1
|
Qiu C, Zheng X, Zhou X, Wang B, Chen T, Xu Y, Yu X, Lu W, Wu Z. The IGF2BP2-circ-DAPK1 axis promotes high-glucose-induced ferroptosis of HUVECs by decreasing NQO1 expression. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167797. [PMID: 40086519 DOI: 10.1016/j.bbadis.2025.167797] [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: 10/08/2024] [Revised: 01/25/2025] [Accepted: 03/07/2025] [Indexed: 03/16/2025]
Abstract
Circular RNAs (circRNAs) are non-coding RNAs with covalently closed loop structures that participate in various biological processes. However, the functions of many circRNAs remain unclear. Endothelial cell dysfunction, which involves abnormal ferroptosis, a unique form of regulated cell death, is a characteristic of various diseases. However, the mechanisms governing ferroptosis in endothelial cells are not fully understood. Here, we investigated the impact of a novel circRNA, circ-DAPK1, on ferroptosis in human umbilical vein endothelial cells (HUVECs) under high-glucose conditions. Our data showed that high-glucose conditions upregulate circ-DAPK1 expression in HUVECs. Overexpression of circ-DAPK1 induced ferroptosis in HUVECs, whereas depletion of circ-DAPK1 mitigated the ferroptosis triggered by high-glucose treatment. Inhibition of ferroptosis reversed the decrease in cell viability induced by high glucose or circ-DAPK1 overexpression. Using RNA immunoprecipitation analyses, we identified several ferroptosis-regulating proteins, including NAD(P)H dehydrogenase [quinone] 1 (NQO1) and insulin-like growth factor 2 mRNA binding protein 2 (IGF2BP2). Mechanistically, circ-DAPK1 interacts with NQO1, enhancing its ubiquitination and accelerating its degradation. NQO1 overexpression partially rescues HUVECs from high-glucose-induced ferroptosis. We also found that IGF2BP2 binds to the m6A site on circ-DAPK1. Depletion of IGF2BP2 in HUVECs reduced circ-DAPK1 expression and inhibited high-glucose-induced ferroptosis. These findings reveal the effects of the IGF2BP2-circ-DAPK1 axis in regulating ferroptosis in HUVECs under high-glucose conditions and extend our understanding of the mechanisms controlling ferroptosis in endothelial cells.
Collapse
Affiliation(s)
- Chenyang Qiu
- Department of Vascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiangtao Zheng
- Department of Vascular Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Xueyuan Western Road, Lucheng District, Wenzhou, China
| | - Xiaoxiang Zhou
- Department of Vascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Bing Wang
- Department of Vascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Tianchi Chen
- Department of Vascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yiting Xu
- Department of Vascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xinyu Yu
- Department of Vascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wei Lu
- Department of Cardiovascular Surgery, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China.
| | - Ziheng Wu
- Department of Vascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
2
|
Pan W, Shao S, Cao X, Dai X, Zheng Y, Cheng J, Feng W, Wu R, Chen Y. In Situ Conversion of Atherosclerotic Plaques' Iron into Nanotheranostics. J Am Chem Soc 2025; 147:3553-3569. [PMID: 39836090 DOI: 10.1021/jacs.4c15068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
The presence of a substantial necrotic core in atherosclerotic plaques markedly heightens the risk of rupture, a consequence of elevated iron levels that exacerbate oxidative stress and lipid peroxidation, thereby sustaining a detrimental cycle of ferroptosis and inflammation. Concurrently targeting both ferroptosis and inflammation is crucial for the effective treatment of vulnerable plaques. In this study, we introduce gallium hexacyanoferrate nanoabsorption catalysts (GaHCF NACs) designed to disrupt this pathological cycle. GaHCF NACs function as highly efficient iron chelators with robust antiferroptosis properties. Through in situ capture of iron within atherosclerotic plaques, these catalysts enhance reactive oxygen species scavenging, initiating an amplified therapeutic response. GaHCF NACs significantly advance plaque regression, stabilization, and vascular functional recovery by inhibiting MAPK13 (p38-δ MAPK) signaling, a key mediator of inflammation and cell death. Importantly, the in situ iron capture process generates a detectable photoacoustic signal, offering a notable diagnostic advantage that allows real-time monitoring of plague status. This multifunctional nanocatalytic platform in situ transforms toxic iron within atherosclerotic plaques into both a therapeutic and diagnostic agent, adapting dynamically to the microenvironment and representing a promising strategy for reducing plaque vulnerability and preventing rupture.
Collapse
Affiliation(s)
- Wenqi Pan
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 200080 Shanghai, P. R. China
| | - Sihui Shao
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 200080 Shanghai, P. R. China
| | - Xinyue Cao
- Materdicine Lab, School of Life Sciences, Shanghai University, 200444 Shanghai, P. R. China
| | - Xinyue Dai
- Materdicine Lab, School of Life Sciences, Shanghai University, 200444 Shanghai, P. R. China
| | - Yi Zheng
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 200080 Shanghai, P. R. China
| | - Jingyun Cheng
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 200080 Shanghai, P. R. China
| | - Wei Feng
- Materdicine Lab, School of Life Sciences, Shanghai University, 200444 Shanghai, P. R. China
| | - Rong Wu
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 200080 Shanghai, P. R. China
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, 200444 Shanghai, P. R. China
| |
Collapse
|
3
|
Niu Q, Zhao L, Wang R, Du L, Shi Q, Jia C, Li G, Jin L, Li F. Predictive value of contrast-enhanced ultrasonography and ultrasound elastography for management of BI-RADS category 4 nonpalpable breast masses. Eur J Radiol 2024; 173:111391. [PMID: 38422608 DOI: 10.1016/j.ejrad.2024.111391] [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: 06/04/2023] [Revised: 02/07/2024] [Accepted: 02/21/2024] [Indexed: 03/02/2024]
Abstract
PURPOSE The objective of this study was to investigate the independent risk factors and associated predictive values of contrast-enhanced ultrasound (CEUS), shear wave elastography (SWE), and strain elastography (SE) for high-risk lesions (HRL) and malignant tumors (MT) among nonpalpable breast masses classified as BI-RADS category 4 on conventional ultrasound. METHODS This prospective study involved consecutively admitted patients with breast tumors from January 2018, aiming to explore the management of BI-RADS category 4 breast tumors using CEUS and elastography. We conducted a retrospective review of patient data, focusing on those with a history of a nonpalpable mass as the primary complaint. Pathologic findings after surgical resection served as the gold standard. The CEUS arterial-phase indices were analyzed using contrast agent arrival-time parametric imaging processing mode, while quantitative and qualitative indices were examined on ES images. Independent risk factors were identified through binary logistic regression multifactorial analysis. The predictive efficacy of different modalities was compared using a receiver operating characteristics curve. Subsequently, a nomogram for predicting the risk of HRL/MT was established based on a multifactorial logistic regression model. RESULTS A total of 146 breast masses from 146 patients were included, comprising 80 benign tumors, 12 HRLs, and 54 MTs based on the final pathology. There was no significant difference in pathologic size between the benign and HRL/MT groups [8.00(6.25,10.00) vs. 9.00(6.00,10.00), P = 0.506]. The diagnostic efficacy of US plus CEUS exceeded that of US plus SWE/SE for BI-RADS 4 nonpalpable masses, with an AUC of 0.954 compared to 0.798/0.741 (P < 0.001). Further stratified analysis revealed a more pronounced improvement for reclassification of BI-RADS 4a masses (AUC: 0.943 vs. 0.762/0.675, P < 0.001) than BI-RADS 4b (AUC:0.950 vs. 0.885/0.796, P>0.05) with the assistance of CEUS than SWE/SE. Employing downgrade CEUS strategies resulted in negative predictive values ranging from 95.2 % to 100.0 % for BI-RADS 4a and 4b masses. Conversely, using upgrade nomogram strategies, which included the independent predictive risk factors of irregular enhanced shape, poor defined enhanced margin, earlier enhanced time, increased surrounding vessels, and presence of contrast agent retention, the diagnostic performance achieved an AUC of 0.947 with good calibration. CONCLUSION After investigating the potential of CEUS and ES in improving risk assessment and diagnostic accuracy for nonpalpable BI-RADS category 4 breast masses, it is evident that CEUS has a more significant impact on enhancing classification compared to ES, particularly for BI-RADS 4a subgroup masses. This finding suggests that CEUS may offer greater benefits in improving risk assessment and diagnostic accuracy for this specific subgroup of breast masses.
Collapse
Affiliation(s)
- Qinghua Niu
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Zhao
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruitao Wang
- Department of Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lianfang Du
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiusheng Shi
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chao Jia
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Gang Li
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lifang Jin
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Fan Li
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
4
|
Zhang L, Li Y, Yang W, Lin L, Li J, Liu D, Li C, Wu J, Li Y. Protocatechuic aldehyde increases pericyte coverage and mitigates pericyte damage to enhance the atherosclerotic plaque stability. Biomed Pharmacother 2023; 168:115742. [PMID: 37871558 DOI: 10.1016/j.biopha.2023.115742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/15/2023] [Accepted: 10/16/2023] [Indexed: 10/25/2023] Open
Abstract
Pericyte dysfunction and loss contribute substantially to the destabilization and rupture of atherosclerotic plaques. Protocatechuic aldehyde (PCAD), a natural polyphenol, exerts anti-atherosclerotic effects. However, the effects and mechanisms of this polyphenol on pericyte recruitment, coverage, and pericyte function remain unknown. We here treated apolipoprotein E-deficient mice having high-fat diet-induced atherosclerosis with PCAD. PCAD achieved therapeutic effects similar to rosuvastatin in lowering lipid levels and thus preventing atherosclerosis progression. With PCAD administration, plaque phenotype exhibited higher stability with markedly reduced lesion vulnerability, which is characterized by reduced lipid content and macrophage accumulation, and a consequent increase in collagen deposition. PCAD therapy increased pericyte coverage in the plaques, reduced VEGF-A production, and inhibited intraplaque neovascularization. PCAD promoted pericyte proliferation, adhesion, and migration to mitigate ox-LDL-induced pericyte dysfunction, which thus maintained the capillary network structure and stability. Furthermore, TGFBR1 silencing partially reversed the protective effect exerted by PCAD on human microvascular pericytes. PCAD increased pericyte coverage and impeded ox-LDL-induced damages through TGF-β1/TGFBR1/Smad2/3 signaling. All these novel findings indicated that PCAD increases pericyte coverage and alleviates pericyte damage to improve the stability of atherosclerotic plaques, which is accomplished by regulating TGF-β1/TGFBR1/Smad2/3 signaling in pericytes.
Collapse
Affiliation(s)
- Lei Zhang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Yuan Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Wenqing Yang
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Lin Lin
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Jie Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Dekun Liu
- Shool of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Chao Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| | - Jibiao Wu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| | - Yunlun Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Department of Cardiovascular, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Shandong Provincial Engineering Laboratory of Traditional Chinese Medicine Precision Therapy for Cardiovascular Diseases, Jinan 250355, China.
| |
Collapse
|
5
|
Burke-Kleinman J, Gotlieb AI. Progression of Arterial Vasa Vasorum from Regulator of Arterial Homeostasis to Promoter of Atherogenesis. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:1468-1484. [PMID: 37356574 DOI: 10.1016/j.ajpath.2023.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/30/2023] [Accepted: 06/08/2023] [Indexed: 06/27/2023]
Abstract
The vasa vasorum (vessels of vessels) are a dynamic microvascular system uniquely distributed to maintain physiological homeostasis of the artery wall by supplying nutrients and oxygen to the outer layers of the artery wall, adventitia, and perivascular adipose tissue, and in large arteries, to the outer portion of the medial layer. Vasa vasorum endothelium and contractile mural cells regulate direct access of bioactive cells and factors present in both the systemic circulation and the arterial perivascular adipose tissue and adventitia to the artery wall. Experimental and human data show that proatherogenic factors and cells gain direct access to the artery wall via the vasa vasorum and may initiate, promote, and destabilize the plaque. Activation and growth of vasa vasorum occur in all blood vessel layers primarily by angiogenesis, producing fragile and permeable new microvessels that may cause plaque hemorrhage and fibrous cap rupture. Ironically, invasive therapies, such as angioplasty and coronary artery bypass grafting, injure the vasa vasorum, leading to treatment failures. The vasa vasorum function both as a master integrator of arterial homeostasis and, once perturbed or injured, as a promotor of atherogenesis. Future studies need to be directed at establishing reliable in vivo and in vitro models to investigate the cellular and molecular regulation of the function and dysfunction of the arterial vasa vasorum.
Collapse
Affiliation(s)
- Jonah Burke-Kleinman
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
| | - Avrum I Gotlieb
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
6
|
Pham TT, Le AH, Dang CP, Chong SY, Do DV, Peng B, Jayasinghe MK, Ong HB, Hoang DV, Louise RA, Loh Y, Hou HW, Wang J, Le MTN. Endocytosis of red blood cell extracellular vesicles by macrophages leads to cytoplasmic heme release and prevents foam cell formation in atherosclerosis. J Extracell Vesicles 2023; 12:e12354. [PMID: 37553837 PMCID: PMC10410060 DOI: 10.1002/jev2.12354] [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: 01/10/2023] [Revised: 07/19/2023] [Accepted: 07/21/2023] [Indexed: 08/10/2023] Open
Abstract
Extracellular vesicles (EVs) can be produced from red blood cells (RBCs) on a large scale and used to deliver therapeutic payloads efficiently. However, not much is known about the native biological properties of RBCEVs. Here, we demonstrate that RBCEVs are primarily taken up by macrophages and monocytes. This uptake is an active process, mediated mainly by endocytosis. Incubation of CD14+ monocytes with RBCEVs induces their differentiation into macrophages with an Mheme-like phenotype, characterized by upregulation of heme oxygenase-1 (HO-1) and the ATP-binding cassette transporter ABCG1. Moreover, macrophages that take up RBCEVs exhibit a reduction in surface CD86 and decreased secretion of TNF-α under inflammatory stimulation. The upregulation of HO-1 is attributed to heme derived from haemoglobin in RBCEVs. Heme is released from internalized RBCEVs in late endosomes and lysosomes via the heme transporter, HRG1. Consequently, RBCEVs exhibit the ability to attenuate foam cell formation from oxidized low-density lipoproteins (oxLDL)-treated macrophages in vitro and reduce atherosclerotic lesions in ApoE knockout mice on a high-fat diet. In summary, our study reveals the uptake mechanism of RBCEVs and their delivery of heme to macrophages, suggesting the potential application of RBCEVs in the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Thach Tuan Pham
- Department of Pharmacology, and Institute for Digital Medicine, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Anh Hong Le
- Department of Pharmacology, and Institute for Digital Medicine, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Cong Phi Dang
- Department of Pharmacology, and Institute for Digital Medicine, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Suet Yen Chong
- Department of Surgery, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Cardiovascular Research Institute, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Dang Vinh Do
- Department of Pharmacology, and Institute for Digital Medicine, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Boya Peng
- Department of Pharmacology, and Institute for Digital Medicine, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Migara Kavishka Jayasinghe
- Department of Pharmacology, and Institute for Digital Medicine, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Hong Boon Ong
- School of Mechanical and Aerospace EngineeringNanyang Technological UniversitySingaporeSingapore
- Lee Kong Chian School of MedicineNanyang Technological UniversitySingaporeSingapore
| | - Dong Van Hoang
- Department of Pharmacology, and Institute for Digital Medicine, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Roma Anne Louise
- Department of Surgery, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Yuin‐Han Loh
- A*STAR Institute of Molecular and Cell BiologySingaporeSingapore
- Department of Biological SciencesNational University of SingaporeSingaporeSingapore
| | - Han Wei Hou
- School of Mechanical and Aerospace EngineeringNanyang Technological UniversitySingaporeSingapore
- Lee Kong Chian School of MedicineNanyang Technological UniversitySingaporeSingapore
| | - Jiong‐Wei Wang
- Department of Surgery, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Cardiovascular Research Institute, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Department of Physiology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Minh TN Le
- Department of Pharmacology, and Institute for Digital Medicine, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Department of Surgery, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- A*STAR Institute of Molecular and Cell BiologySingaporeSingapore
| |
Collapse
|
7
|
Yu Q, Wei P, Xu L, Xia C, Li Y, Liu H, Song X, Tian K, Fu W, Wang R, Wang W, Bai L, Fan J, Liu E, Zhao S. Urotensin II Enhances Advanced Aortic Atherosclerosis Formation and Delays Plaque Regression in Hyperlipidemic Rabbits. Int J Mol Sci 2023; 24:ijms24043819. [PMID: 36835230 PMCID: PMC9963243 DOI: 10.3390/ijms24043819] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 01/30/2023] [Accepted: 02/02/2023] [Indexed: 02/17/2023] Open
Abstract
Accumulated evidence shows that elevated urotensin II (UII) levels are associated with cardiovascular diseases. However, the role of UII in the initiation, progression, and regression of atherosclerosis remains to be verified. Different stages of atherosclerosis were induced in rabbits by a 0.3% high cholesterol diet (HCD) feeding, and either UII (5.4 μg/kg/h) or saline was chronically infused via osmotic mini-pumps. UII promoted atherosclerotic fatty streak formation in ovariectomized female rabbits (34% increase in gross lesion and 93% increase in microscopic lesion), and in male rabbits (39% increase in gross lesion). UII infusion significantly increased the plaque size of the carotid and subclavian arteries (69% increase over the control). In addition, UII infusion significantly enhanced the development of coronary lesions by increasing plaque size and lumen stenosis. Histopathological analysis revealed that aortic lesions in the UII group were characterized by increasing lesional macrophages, lipid deposition, and intra-plaque neovessel formation. UII infusion also significantly delayed the regression of atherosclerosis in rabbits by increasing the intra-plaque macrophage ratio. Furthermore, UII treatment led to a significant increase in NOX2 and HIF-1α/VEGF-A expression accompanied by increased reactive oxygen species levels in cultured macrophages. Tubule formation assays showed that UII exerted a pro-angiogenic effect in cultured endothelial cell lines and this effect was partly inhibited by urantide, a UII receptor antagonist. These findings suggest that UII can accelerate aortic and coronary plaque formation and enhance aortic plaque vulnerability, but delay the regression of atherosclerosis. The role of UII on angiogenesis in the lesion may be involved in complex plaque development.
Collapse
Affiliation(s)
- Qingqing Yu
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
- Laboratory Animal Center, Xi’an Jiaotong University, Xi’an 710061, China
| | - Panpan Wei
- Laboratory Animal Center, Xi’an Jiaotong University, Xi’an 710061, China
| | - Liran Xu
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
| | - Congcong Xia
- Laboratory Animal Center, Xi’an Jiaotong University, Xi’an 710061, China
| | - Yafeng Li
- Laboratory Animal Center, Xi’an Jiaotong University, Xi’an 710061, China
| | - Haole Liu
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
| | - Xiaojie Song
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
| | - Kangli Tian
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
| | - Weilai Fu
- Laboratory Animal Center, Xi’an Jiaotong University, Xi’an 710061, China
| | - Rong Wang
- Laboratory Animal Center, Xi’an Jiaotong University, Xi’an 710061, China
| | - Weirong Wang
- Laboratory Animal Center, Xi’an Jiaotong University, Xi’an 710061, China
| | - Liang Bai
- Laboratory Animal Center, Xi’an Jiaotong University, Xi’an 710061, China
| | - Jianglin Fan
- Department of Molecular Pathology, Faculty of Medicine, Graduate School of Medical Sciences, University of Yamanashi, Tokyo 409-3898, Japan
| | - Enqi Liu
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
- Laboratory Animal Center, Xi’an Jiaotong University, Xi’an 710061, China
| | - Sihai Zhao
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
- Department of Cardiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
- Correspondence: ; Tel.: +86-29-82655361
| |
Collapse
|
8
|
Tissue oxygenation stabilizes neovessels and mitigates hemorrhages in human atherosclerosis-induced angiogenesis. Angiogenesis 2023; 26:63-76. [PMID: 35947328 DOI: 10.1007/s10456-022-09851-8] [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: 04/01/2022] [Accepted: 07/21/2022] [Indexed: 11/01/2022]
Abstract
Progression of atherosclerosis is associated with a maladaptive form of angiogenesis which contributes to intraplaque hemorrhage and plaque disruption. Hypoxia has been implicated in mechanisms of angiogenic neovessel fragility and atherosclerotic plaque destabilization. We used ex vivo and in vivo models to characterize the effect of oxygen (O2) on the formation, stability and tendency to bleed of human plaque-induced neovessels. Plaque explants potently stimulated the ex vivo angiogenic response of rat aortic rings at atmospheric O2 levels. Severe hypoxia (1% O2) inhibited plaque-induced angiogenesis and pericyte recruitment causing neovessel breakdown, whereas increasing O2 levels dose dependently enhanced pericyte numbers and neovessel stability. Plaque fragments implanted subcutaneously with or without aortic rings in SCID mice stimulated the host angiogenic response with plaques causing minimal or no hemorrhages and plaques co-implanted with aortic rings causing marked hemorrhages. Plaque/aortic ring-induced hemorrhages were reduced in mice exposed to moderate hyperoxia (50% O2). Hyperoxia downregulated expression of the hypoxia-sensitive genes Ca9, Ca12 and VegfA and increased influx into implants of mesenchymal cells reactive for the pericyte marker NG2. In both ex vivo and in vivo models, O2 promoted expression of vasostabilizing genes required for pericyte recruitment (Angpt1, Pdgfb), basement membrane assembly (Col4A1), and tight junction formation (Cldn5 and/or Ocln). Our results suggest that formation of neovessels that are stable, pericyte-coated, and resistant to bleeding requires adequate tissue oxygenation. Understanding the mechanisms by which O2 stabilizes neovessels and mitigates neovessel bleeding may lead to new therapies for the prevention of atherosclerosis complications.
Collapse
|
9
|
Wu Q, Ying X, Yu W, Li H, Wei W, Lin X, Zhang X. Identification of ferroptosis-related genes in syncytiotrophoblast-derived extracellular vesicles of preeclampsia. Medicine (Baltimore) 2022; 101:e31583. [PMID: 36343018 PMCID: PMC9646584 DOI: 10.1097/md.0000000000031583] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Preeclampsia (PE), defined as new-onset hypertension and multi-organ systemic complication during pregnancy, is the leading cause of maternal and neonatal mortality and morbidity. With extracellular vesicles research progresses, current data refers to the possibility that ferroptosis may play a role in exosomal effects. Evidence has suggested that ferroptosis may contribute to the pathogenesis of preeclampsia by bioinformatics analyses. The purpose of the current study is to identify the potential ferroptosis-related genes in syncytiotrophoblast-derived extracellular vesicles (STB-EVs) of preeclampsia using bioinformatics analyses. Clinical characteristics and gene expression data of all samples were obtained from the NCBI GEO database. The differentially expressed mRNAs (DE-mRNAs) in STB-EVs of preeclampsia were screened and then were intersected with ferroptosis genes. Functional and pathway enrichment analyses of ferroptosis-related DE-mRNAs in STB-EVs were performed. Ferroptosis-related hub genes in STB-EVs were identified by Cytoscape plugin CytoHubba with a Degree algorithm using a protein-protein interaction network built constructed from the STRING database. The predictive performance of ferroptosis-related hub genes was determined by a univariate analysis of receiver operating characteristic (ROC). The miRNA-hub gene regulatory network was constructed using the miRwalk database. A total of 1976 DE-mRNAs in STB-EVs were identified and the most enriched item identified by gene set enrichment analysis was signaling by G Protein-Coupled Receptors (normalized enrichment score = 1.238). These DE-mRNAs obtained 26 ferroptosis-related DE-mRNAs. Ferroptosis-related DE-mRNAs of gene ontology terms and Encyclopedia of Genes and Genomes pathway enrichment analysis were enriched significantly in response to oxidative stress and ferroptosis. Five hub genes (ALB, NOX4, CDKN2A, TXNRD1, and CAV1) were found in the constructed protein-protein interaction network with ferroptosis-related DE-mRNAs and the areas under the ROC curves for ALB, NOX4, CDKN2A, TXNRD1, and CAV1 were 0.938 (CI: 0.815-1.000), 0.833 (CI: 0.612-1.000), 0.875 (CI: 0.704-1.000), 0.958 (CI: 0.862-1.000), and 0.854 (CI: 0.652-1.000) in univariate analysis of ROC. We constructed a regulatory network of miRNA-hub gene and the findings demonstrate that hsa-miR-26b-5p, hsa-miR-192-5p, hsa-miR-124-3p, hsa-miR-492, hsa-miR-34a-5p and hsa-miR-155-5p could regulate most hub genes. In this study, we identified several central genes closely related to ferroptosis in STB-EVs (ALB, NOX4, CDKN2A, TXNRD1, and CAV1) that are potential biomarkers related to ferroptosis in preeclampsia. Our findings will provide evidence for the involvement of ferroptosis in preeclampsia and improve the understanding of ferroptosis-related molecular pathways in the pathogenesis of preeclampsia.
Collapse
Affiliation(s)
- Quanfeng Wu
- Department of Obstetrics, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Xiang Ying
- Department of Gynecology and Obstetrics, Shanghai Jiaotong University School of Medicine Xinhua Hospital, Shanghai, China
| | - Weiwei Yu
- Department of Obstetrics, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Huanxi Li
- Department of Obstetrics, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Wei Wei
- Department of Obstetrics, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Xueyan Lin
- Department of Obstetrics, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Xueqin Zhang
- Department of Obstetrics, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen, China
- *Correspondence: Xueqin Zhang, Department of Obstetrics, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen 361003, China (e-mail: )
| |
Collapse
|
10
|
Bogdanov L, Shishkova D, Mukhamadiyarov R, Velikanova E, Tsepokina A, Terekhov A, Koshelev V, Kanonykina A, Shabaev A, Frolov A, Zagorodnikov N, Kutikhin A. Excessive Adventitial and Perivascular Vascularisation Correlates with Vascular Inflammation and Intimal Hyperplasia. Int J Mol Sci 2022; 23:ijms232012156. [PMID: 36293013 PMCID: PMC9603343 DOI: 10.3390/ijms232012156] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/09/2022] [Accepted: 10/10/2022] [Indexed: 12/24/2022] Open
Abstract
Albeit multiple studies demonstrated that vasa vasorum (VV) have a crucial importance in vascular pathology, the informative markers and metrics of vascular inflammation defining the development of intimal hyperplasia (IH) have been vaguely studied. Here, we employed two rat models (balloon injury of the abdominal aorta and the same intervention optionally complemented with intravenous injections of calciprotein particles) and a clinical scenario (arterial and venous conduits for coronary artery bypass graft (CABG) surgery) to investigate the pathophysiological interconnections among VV, myeloperoxidase-positive (MPO+) clusters, and IH. We found that the amounts of VV and MPO+ clusters were strongly correlated; further, MPO+ clusters density was significantly associated with balloon-induced IH and increased at calciprotein particle-provoked endothelial dysfunction. Likewise, number and density of VV correlated with IH in bypass grafts for CABG surgery at the pre-intervention stage and were higher in venous conduits which more frequently suffered from IH as compared with arterial grafts. Collectively, our results underline the pathophysiological importance of excessive VV upon the vascular injury or at the exposure to cardiovascular risk factors, highlight MPO+ clusters as an informative marker of adventitial and perivascular inflammation, and propose another mechanistic explanation of a higher long-term patency of arterial grafts upon the CABG surgery.
Collapse
|
11
|
Humar R, Schaer DJ, Vallelian F. Erythrophagocytes in hemolytic anemia, wound healing, and cancer. Trends Mol Med 2022; 28:906-915. [PMID: 36096988 DOI: 10.1016/j.molmed.2022.08.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 08/11/2022] [Accepted: 08/11/2022] [Indexed: 11/18/2022]
Abstract
Hemolysis is a ubiquitous pathology defined as premature red blood cell destruction within the circulation or local tissues. One of the most archetypal functions of macrophages is phagocytosis of damaged or extravasated red blood cells, preventing the extracellular release of toxic hemoglobin and heme. Upon erythrophagocytosis, spiking intracellular heme concentrations drive macrophage transformation into erythrophagocytes, leveraging antioxidative and iron recycling capacities to defend against hemolytic stress. This unique phenotype transformation is coordinated by a regulatory network comprising the transcription factors BACH1, SPI-C, NRF2, and ATF1. Erythrophagocytes negatively regulate inflammation and immunity and may modulate disease-specific outcomes in hemolytic anemia, wound healing, atherosclerosis, and cancer. In this opinion article, we outline the known and presumed functions of erythrophagocytes and their implications for therapeutic innovation and research.
Collapse
Affiliation(s)
- Rok Humar
- Department of Internal Medicine, University Hospital and University of Zurich, Zurich, Switzerland
| | - Dominik J Schaer
- Department of Internal Medicine, University Hospital and University of Zurich, Zurich, Switzerland
| | - Florence Vallelian
- Department of Internal Medicine, University Hospital and University of Zurich, Zurich, Switzerland.
| |
Collapse
|
12
|
Lv X, Wang F, Sun M, Sun C, Fan X, Ma B, Yang Y, Ye Z, Liu P, Wen J. Differential Gene Expression and Immune Cell Infiltration in Carotid Intraplaque Hemorrhage Identified Using Integrated Bioinformatics Analysis. Front Cardiovasc Med 2022; 9:818585. [PMID: 35656397 PMCID: PMC9152291 DOI: 10.3389/fcvm.2022.818585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 03/11/2022] [Indexed: 11/24/2022] Open
Abstract
Background Intraplaque hemorrhage (IPH) is an important feature of unstable plaques and an independent risk factor for cardiovascular events. However, the molecular mechanisms contributing to IPH are incompletely characterized. We aimed to identify novel biomarkers and interventional targets for IPH and to characterize the role of immune cells in IPH pathogenesis. Methods The microarray dataset GSE163154 which contain IPH and non-IPH plaque samples was obtained from the Gene Expression Omnibus (GEO). R software was adopted for identifying differentially expressed genes (DEGs) and conducting functional investigation. The hub genes were carried by protein-protein interaction (PPI) network and were validated by the GSE120521 dataset. CIBERSORT deconvolution was used to determine differential immune cell infiltration and the relationship of immune cells and hub genes. We confirmed expression of proteins encoded by the hub genes by immunohistochemistry and western blotting in 8 human carotid endarterectomy samples with IPH and 8 samples without IPH (non-IPH). Results We detected a total of 438 differentially expressed genes (DEGs), of which 248 were upregulated and 190 were downregulated. DEGs were mainly involved in inflammatory related pathways, including neutrophil activation, neutrophil degranulation, neutrophil-mediated immunity, leukocyte chemotaxis, and lysosomes. The hub genes found through the method of degree in the PPI network showed that ITGB2 and ITGAM might play an important role in IPH. Receiver operating characteristic (ROC) results also showed a good performance of these two genes in the test and validation dataset. We found that the proportions of infiltrating immune cells in IPH and non-IPH samples differed, especially in terms of M0 and M2 macrophages. Immunohistochemistry and western blotting analysis showed that expression levels of ITGB2 and ITGAM increased significantly in carotid atherosclerotic plaques with IPH. Conclusion ITGB2 and ITGAM are key hub genes of IPH and may play an important role in the biological process of IPH. Our findings advance our understanding of the underlying mechanisms of IPH pathogenesis and provide valuable information and directions for future research into novel targets for IPH diagnosis and immunotherapy.
Collapse
Affiliation(s)
- Xiaoshuo Lv
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
- Graduate School of Peking Union Medical College, Beijing, China
| | - Feng Wang
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
- Graduate School of Peking Union Medical College, Beijing, China
| | - Mingsheng Sun
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Congrui Sun
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Xueqiang Fan
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Bo Ma
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Yuguang Yang
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Zhidong Ye
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Peng Liu
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
- Graduate School of Peking Union Medical College, Beijing, China
- Peng Liu
| | - Jianyan Wen
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
- Graduate School of Peking Union Medical College, Beijing, China
- *Correspondence: Jianyan Wen
| |
Collapse
|
13
|
Yuan W, Xia H, Xu Y, Xu C, Chen N, Shao C, Dai Z, Chen R, Tao A. The role of ferroptosis in endothelial cell dysfunction. Cell Cycle 2022; 21:1897-1914. [PMID: 35579940 DOI: 10.1080/15384101.2022.2079054] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Ferroptosis is a form of iron-dependent cell death caused by an excessive accumulation of reactive oxygen species and lipid peroxidation. The importance of ferroptosis in the occurrence and progression of various diseases is gradually being recognized; however, the exact biological effects and potential mechanisms of endothelial cell ferroptosis remain unclear. The endothelium forms the innermost layer of the blood vessels and lymphatic vessels. It acts as an important functional interface, responds to various pathological stimuli and causes endothelial dysfunction. Here, we review recent findings to elucidate the role of ferroptosis in endothelial cells under different pathophysiologic settings.
Collapse
Affiliation(s)
- Wei Yuan
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Hao Xia
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yao Xu
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Chong Xu
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Nan Chen
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Chen Shao
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Zhiyin Dai
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Rui Chen
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Aibin Tao
- Department of Cardiology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| |
Collapse
|
14
|
Functional Phenotypes of Intraplaque Macrophages and Their Distinct Roles in Atherosclerosis Development and Atheroinflammation. Biomedicines 2022; 10:biomedicines10020452. [PMID: 35203661 PMCID: PMC8962399 DOI: 10.3390/biomedicines10020452] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/04/2022] [Accepted: 02/11/2022] [Indexed: 12/01/2022] Open
Abstract
Macrophages are the key inflammatory cell type involved in all stages of atherosclerosis development and progression, as demonstrated by numerous studies. Correspondingly, macrophages are currently regarded as a promising therapeutic target for the development of new treatment approaches. The macrophage population is heterogeneous and dynamic, as these cells can switch between a number of distinct functional states with pro- and anti-atherogenic activity in response to various stimuli. An atherosclerotic plaque microenvironment defined by cytokine levels, cell-to-cell interactions, lipid accumulation, hypoxia, neoangiogenesis, and intraplaque haemorrhage may guide local macrophage polarization processes within the lesion. In this review, we discuss known functional phenotypes of intraplaque macrophages and their distinct contribution to ahteroinflammation.
Collapse
|
15
|
Yang Z, Shi J, Chen L, Fu C, Shi D, Qu H. Role of Pyroptosis and Ferroptosis in the Progression of Atherosclerotic Plaques. Front Cell Dev Biol 2022; 10:811196. [PMID: 35186925 PMCID: PMC8850398 DOI: 10.3389/fcell.2022.811196] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 01/17/2022] [Indexed: 12/12/2022] Open
Abstract
Pyroptosis is a special way of programmed cell death which is dependent on the activation of cysteinyl aspartate specific proteinase 1 (Caspase-1) and Caspase-4/5/11. Ferroptosis is an iron-dependent cell death that characterized by the intra-cellular lipid peroxidation-mediated membrane damage. Pyroptosis or ferroptosis in macrophages, smooth muscle cells, and vascular endothelial cells are believed to be closely related to the progression of atherosclerotic plaques. Therefore, we discuss the role of pyroptosis and ferroptosis in the development of atherosclerotic plaques and may provide new strategies for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Zhen Yang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Cardiovascular Department, Peking University Traditional Chinese Medicine Clinical Medical School (Xiyuan), Beijing, China
- Academy of Integration of Chinese and Western Medicine, Peking University Health Science Center, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
| | - Junhe Shi
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Traditional Chinese Medicine, Beijing, China
| | - Li Chen
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Cardiovascular Department, Peking University Traditional Chinese Medicine Clinical Medical School (Xiyuan), Beijing, China
- Academy of Integration of Chinese and Western Medicine, Peking University Health Science Center, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
| | - Changgeng Fu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Changgeng Fu, ; Dazhuo Shi, ; Hua Qu,
| | - Dazhuo Shi
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Cardiovascular Department, Peking University Traditional Chinese Medicine Clinical Medical School (Xiyuan), Beijing, China
- Academy of Integration of Chinese and Western Medicine, Peking University Health Science Center, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
- *Correspondence: Changgeng Fu, ; Dazhuo Shi, ; Hua Qu,
| | - Hua Qu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Traditional Chinese Medicine, Beijing, China
- *Correspondence: Changgeng Fu, ; Dazhuo Shi, ; Hua Qu,
| |
Collapse
|
16
|
Hyperglycemia Induces Inflammatory Response of Human Macrophages to CD163-Mediated Scavenging of Hemoglobin-Haptoglobin Complexes. Int J Mol Sci 2022; 23:ijms23031385. [PMID: 35163309 PMCID: PMC8836198 DOI: 10.3390/ijms23031385] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/17/2022] [Accepted: 01/19/2022] [Indexed: 01/27/2023] Open
Abstract
Hyperglycemia, a hallmark of diabetes, can induce inflammatory programming of macrophages. The macrophage scavenger receptor CD163 internalizes and degrades hemoglobin-haptoglobin (Hb-Hp) complexes built due to intravascular hemolysis. Clinical studies have demonstrated a correlation between impaired scavenging of Hb-Hp complexes via CD163 and diabetic vascular complications. Our aim was to identify whether hyperglycemia is able to amplify inflammation via Hb-Hp complex interactions with the immune system. M(IFNγ), M(IL-4), and control M0 macrophages were differentiated out of primary human monocytes in normo- (5 mM) and hyperglycemic (25 mM) conditions. CD163 gene expression was decreased 5.53 times in M(IFNγ) with a further decrease of 1.99 times in hyperglycemia. Hyperglycemia suppressed CD163 surface expression in M(IFNγ) (1.43 times). Flow cytometry demonstrated no impairment of Hb-Hp uptake in hyperglycemia. However, hyperglycemia induced an inflammatory response of M(IFNγ) to Hb-Hp1-1 and Hb-Hp2-2 uptake with different dynamics. Hb-Hp1-1 uptake stimulated IL-6 release (3.03 times) after 6 h but suppressed secretion (5.78 times) after 24 h. Contrarily, Hb-Hp2-2 uptake did not affect IL-6 release after 6h but increased secretion after 24 h (3.06 times). Our data show that hyperglycemia induces an inflammatory response of innate immune cells to Hb-Hp1-1 and Hb-Hp2-2 uptake, converting the silent Hb-Hp complex clearance that prevents vascular damage into an inflammatory process, hereby increasing the susceptibility of diabetic patients to vascular complications.
Collapse
|
17
|
Nardi V, Benson J, Bois MC, Saba L, Larson AS, Özcan I, Ahmad A, Morse DW, Meyer FB, Brinjikji W, Lanzino G, Lerman LO, Savastano LE, Lerman A. Carotid Plaques From Symptomatic Patients With Mild Stenosis Is Associated With Intraplaque Hemorrhage. Hypertension 2022; 79:271-282. [PMID: 34878895 DOI: 10.1161/hypertensionaha.121.18128] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Carotid plaque vulnerability features beyond the degree of stenosis may play a key role in the pathogenesis and recurrence of ischemic cerebrovascular events. This study sought to compare intraplaque hemorrhage (IPH) as a marker of plaque vulnerability in symptomatic patients with mild (<50%), moderate (50%-69%), and severe (≥70%) carotid artery stenosis. We included patients who experienced ischemic cerebrovascular events with no other identifiable sources and underwent carotid endarterectomy for mild (n=32), moderate (n=47), and severe (n=58) carotid artery stenosis. The degree of stenosis and imaging hallmarks were assessed by computed tomography angiography or magnetic resonance angiography. Plaque specimens were stained with hematoxylin and eosin and Movat pentachrome staining. Carotid plaques of patients with mild stenosis had a higher extent of IPH (%) on tissue analysis compared with patients with moderate (mild, 15.7% [interquartile range, 7.8%-26.7%]; moderate, 3.9% [0.0%-9.2%]; P<0.001) and severe carotid artery stenosis (mild, 15.7% [interquartile range, 7.8%-26.7%]; severe, 2.5% [interquartile range, 0.0%-11.2%]; P<0.001). When considering the degree of carotid artery stenosis as a continuous variable, a lower lumen narrowing was associated with higher extent of IPH (P<0.001; R, -0.329). Our major finding is the association of IPH with mild carotid artery stenosis based on histological analysis. The current study may suggest that IPH potentially plays a role in the mechanism of stroke in patients with nonobstructive carotid stenosis.
Collapse
Affiliation(s)
- Valentina Nardi
- Department of Cardiovascular Medicine (V.N., I.Ö., A.A., D.W.M., A.L.), Mayo Clinic, Rochester, MN
| | - John Benson
- Department of Radiology (J.C.B., A.S.L., W.B.), Mayo Clinic, Rochester, MN
| | - Melanie C Bois
- Department of Laboratory Medicine and Pathology (M.C.B.), Mayo Clinic, Rochester, MN
| | - Luca Saba
- Department of Radiology, Azienda Ospedaliero Universitaria, Cagliari, Italy (L.S.)
| | - Anthony S Larson
- Department of Radiology (J.C.B., A.S.L., W.B.), Mayo Clinic, Rochester, MN
| | - Ilke Özcan
- Department of Cardiovascular Medicine (V.N., I.Ö., A.A., D.W.M., A.L.), Mayo Clinic, Rochester, MN
| | - Ali Ahmad
- Department of Cardiovascular Medicine (V.N., I.Ö., A.A., D.W.M., A.L.), Mayo Clinic, Rochester, MN
| | - David W Morse
- Department of Cardiovascular Medicine (V.N., I.Ö., A.A., D.W.M., A.L.), Mayo Clinic, Rochester, MN
| | - Fredric B Meyer
- Department of Neurologic Surgery (F.B.M., G.L., L.E.S.), Mayo Clinic, Rochester, MN
| | - Waleed Brinjikji
- Department of Radiology (J.C.B., A.S.L., W.B.), Mayo Clinic, Rochester, MN
| | - Giuseppe Lanzino
- Department of Neurologic Surgery (F.B.M., G.L., L.E.S.), Mayo Clinic, Rochester, MN
| | - Lilach O Lerman
- Department of Nephrology and Hypertension (L.O.L.), Mayo Clinic, Rochester, MN
| | - Luis E Savastano
- Department of Neurologic Surgery (F.B.M., G.L., L.E.S.), Mayo Clinic, Rochester, MN
| | - Amir Lerman
- Department of Cardiovascular Medicine (V.N., I.Ö., A.A., D.W.M., A.L.), Mayo Clinic, Rochester, MN
| |
Collapse
|
18
|
Böhm EW, Pavlaki M, Chalikias G, Mikroulis D, Georgiadis GS, Tziakas DN, Konstantinides S, Schäfer K. Colocalization of Erythrocytes and Vascular Calcification in Human Atherosclerosis: A Systematic Histomorphometric Analysis. TH OPEN 2021; 5:e113-e124. [PMID: 33870075 PMCID: PMC8046517 DOI: 10.1055/s-0041-1725042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 01/18/2021] [Indexed: 11/03/2022] Open
Abstract
Background Intimal calcification typically develops in advanced atherosclerosis, and microcalcification may promote plaque progression and instability. Conversely, intraplaque hemorrhage and erythrocyte extravasation may stimulate osteoblastic differentiation and intralesional calcium phosphate deposition. The presence of erythrocytes and their main cellular components (membranes, hemoglobin, and iron) and colocalization with calcification has never been systematically studied. Methods and Results We examined three types of diseased vascular tissue specimens, namely, degenerative aortic valve stenosis ( n = 46), atherosclerotic carotid artery plaques ( n = 9), and abdominal aortic aneurysms ( n = 14). Biomaterial was obtained from symptomatic patients undergoing elective aortic valve replacement, carotid artery endatherectomy, or aortic aneurysm repair, respectively. Serial sections were stained using Masson-Goldner trichrome, Alizarin red S, and Perl's iron stain to visualize erythrocytes, extracelluar matrix and osteoid, calcium phosphate deposition, or the presence of iron and hemosiderin, respectively. Immunohistochemistry was employed to detect erythrocyte membranes (CD235a), hemoglobin or the hemoglobin scavenger receptor (CD163), endothelial cells (CD31), myofibroblasts (SMA), mesenchymal cells (osteopontin), or osteoblasts (periostin). Our analyses revealed a varying degree of intraplaque hemorrhage and that the majority of extravasated erythrocytes were lysed. Osteoid and calcifications also were frequently present, and erythrocyte membranes were significantly more prevalent in areas with calcification. Areas with extravasated erythrocytes frequently contained CD163-positive cells, although calcification also occurred in areas without CD163 immunosignals. Conclusion Our findings underline the presence of extravasated erythrocytes and their membranes in different types of vascular lesions, and their association with areas of calcification suggests an active role of erythrocytes in vascular disease processes.
Collapse
Affiliation(s)
- Elsa Wilma Böhm
- Department of Cardiology, University Medical Center, Mainz, Germany
| | - Maria Pavlaki
- Department of Cardiology, Democritus University of Thrace, Alexandroupolis, Greece
| | - Georgios Chalikias
- Department of Cardiology, Democritus University of Thrace, Alexandroupolis, Greece
| | - Dimitrios Mikroulis
- Department of Cardiothoracic Surgery, Democritus University of Thrace, Alexandroupolis, Greece
| | - George S Georgiadis
- Department of Vascular Surgery, Democritus University of Thrace, Alexandroupolis, Greece
| | - Dimitrios N Tziakas
- Department of Cardiology, Democritus University of Thrace, Alexandroupolis, Greece
| | | | - Katrin Schäfer
- Department of Cardiology, University Medical Center, Mainz, Germany
| |
Collapse
|
19
|
Baganha F, de Jong RCM, Peters EA, Voorham W, Jukema JW, Delibegovic M, de Vries MR, Quax PHA. Atorvastatin pleiotropically decreases intraplaque angiogenesis and intraplaque haemorrhage by inhibiting ANGPT2 release and VE-Cadherin internalization. Angiogenesis 2021; 24:567-581. [PMID: 33550461 PMCID: PMC8292290 DOI: 10.1007/s10456-021-09767-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 01/04/2021] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Statins pleiotropically provide additional benefits in reducing atherosclerosis, but their effects on intraplaque angiogenesis (IPA) and hemorrhage (IPH) remain unclear. Therefore, we discriminated statin's lipid-lowering dependent and independent effects on IPA and IPH. APPROACH AND RESULTS ApoE3*Leiden mice are statin-responsive due to ApoE and LDLR presence, but also allow to titrate plasma cholesterol levels by diet. Therefore, ApoE3*Leiden mice were fed a high-cholesterol-inducing-diet (HCD) with or without atorvastatin (A) or a moderate-cholesterol-inducing-diet (MCD). Mice underwent vein graft surgery to induce lesions with IPA and IPH. Cholesterol levels were significantly reduced in MCD (56%) and HCD + A (39%) compared to HCD with no significant differences between MCD and HCD + A. Both MCD and HCD + A have a similar reduction in vessel remodeling and inflammation comparing to HCD. IPA was significantly decreased by 30% in HCD + A compared to HCD or MCD. Atorvastatin treatment reduced the presence of immature vessels by 34% vs. HCD and by 25% vs. MCD, resulting in a significant reduction of IPH. Atorvastatin's anti-angiogenic capacity was further illustrated by a dose-dependent reduction of ECs proliferation and migration. Cultured mouse aortic-segments lost sprouting capacity upon atorvastatin treatment and became 30% richer in VE-Cadherin expression and pericyte coverage. Moreover, Atorvastatin inhibited ANGPT2 release and decreased VE-Cadherin(Y685)-phosphorylation in ECs. CONCLUSIONS Atorvastatin has beneficial effects on vessel remodeling due to its lipid-lowering capacity. Atorvastatin has strong pleiotropic effects on IPA by decreasing the number of neovessels and on IPH by increasing vessel maturation. Atorvastatin improves vessel maturation by inhibiting ANGPT2 release and phospho(Y658)-mediated VE-Cadherin internalization.
Collapse
Affiliation(s)
- Fabiana Baganha
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands.,Department of Vascular Surgery/Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, PO Box 9600, 2300 RC, Leiden, The Netherlands.,Aberdeen Cardiovascular and Diabetes Centre, Institute of Medical Sciences, Aberdeen University, Aberdeen, UK
| | - Rob C M de Jong
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands.,Department of Vascular Surgery/Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, PO Box 9600, 2300 RC, Leiden, The Netherlands
| | - Erna A Peters
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands.,Department of Vascular Surgery/Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, PO Box 9600, 2300 RC, Leiden, The Netherlands
| | - Wietske Voorham
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands.,Department of Vascular Surgery/Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, PO Box 9600, 2300 RC, Leiden, The Netherlands
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Mirela Delibegovic
- Aberdeen Cardiovascular and Diabetes Centre, Institute of Medical Sciences, Aberdeen University, Aberdeen, UK
| | - Margreet R de Vries
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands.,Department of Vascular Surgery/Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, PO Box 9600, 2300 RC, Leiden, The Netherlands
| | - Paul H A Quax
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands. .,Department of Vascular Surgery/Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, PO Box 9600, 2300 RC, Leiden, The Netherlands.
| |
Collapse
|
20
|
Liu W, Zong M, Gong HY, Ling LJ, Ye XH, Wang S, Li CY. Comparison of Diagnostic Efficacy Between Contrast-Enhanced Ultrasound and DCE-MRI for Mass- and Non-Mass-Like Enhancement Types in Breast Lesions. Cancer Manag Res 2020; 12:13567-13578. [PMID: 33408526 PMCID: PMC7781362 DOI: 10.2147/cmar.s283656] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 12/16/2020] [Indexed: 01/19/2023] Open
Abstract
Background Contrast-enhanced ultrasound (CEUS) can provide angiogenesis information about breast lesions; however, its diagnostic performance in comparison with that of dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) has not been systematically investigated. This study aimed to evaluate the diagnostic efficacy of CEUS and DCE-MRI in mass-like and non-mass-like enhancement types of breast lesions. Material and Methods A retrospective study was conducted on 252 patients with breast lesions who underwent CEUS and DCE-MRI before surgery between January 2016 and February 2020. Histopathological results were used as reference standards. All patients were classified into mass-like and non-mass-like enhancement lesion groups. The mass-like lesion group was further divided into three categories according to different sizes (group 1: <10 mm, group 2: 10-20 mm, and group 3: >20 mm). Sensitivity, specificity, positive predictive value, negative predictive value, and receiver operating characteristic curve were analyzed to assess the diagnostic performance of these two modalities. Results For mass-like breast lesions, DCE-MRI (Az=0.981) manifested better diagnostic performance than CEUS (Az=0.940) in medium-sized (10-20 mm) tumors (Z=2.018, P=0.043), but both had similar diagnostic performance in smaller (<10 mm) and larger (>20 mm) tumors (P=0.717, P=0.394). For non-mass-like enhancement lesions, CEUS and DCE-MRI showed no significant difference (Z=1.590, P=0.119) and revealed good diagnostic performance (Az=0.859, Az=0.947) in differentiating the two groups. Conclusion For mass-like breast lesions, DCE-MRI showed better diagnostic performance than CEUS in differentiating benign and malignant tumors of medium-sizes (10-20mm) but not of smaller (<10mm) and larger (>20 mm) sizes. For non-mass-like lesions, both modalities showed similar diagnostic performance.
Collapse
Affiliation(s)
- Wei Liu
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People's Republic of China
| | - Min Zong
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People's Republic of China
| | - Hai-Yan Gong
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People's Republic of China
| | - Li-Jun Ling
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People's Republic of China
| | - Xin-Hua Ye
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People's Republic of China
| | - Shui Wang
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People's Republic of China
| | - Cui-Ying Li
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People's Republic of China
| |
Collapse
|
21
|
Cai J, Zhang M, Liu Y, Li H, Shang L, Xu T, Chen Z, Wang F, Qiao T, Li K. Iron accumulation in macrophages promotes the formation of foam cells and development of atherosclerosis. Cell Biosci 2020; 10:137. [PMID: 33292517 PMCID: PMC7691057 DOI: 10.1186/s13578-020-00500-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 11/18/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Macrophages that accumulate in atherosclerotic plaques contribute to progression of the lesions to more advanced and complex plaques. Although iron deposition was found in human atherosclerotic plaques, clinical and pre-clinical studies showed controversial results. Several epidemiological studies did not show the positive correlation between a systemic iron status and an incidence of cardiovascular diseases, suggesting that the iron involvement occurs locally, rather than systemically. RESULTS To determine the direct in vivo effect of iron accumulation in macrophages on the progression of atherosclerosis, we generated Apoe-/- mice with a macrophage-specific ferroportin (Fpn1) deficiency (Apoe-/-Fpn1LysM/LysM). Fpn1 deficiency in macrophages dramatically accelerated the progression of atherosclerosis in mice. Pathophysiological evidence showed elevated levels of reactive oxygen species, aggravated systemic inflammation, and altered plaque-lipid composition. Moreover, Fpn1 deficiency in macrophages significantly inhibited the expression of ABC transporters (ABCA1 and ABCG1) by decreasing the expression of the transcription factor LXRα, which reduced cholesterol efflux and therefore promoted foam cell formation and enhanced plaque formation. Iron chelation relieved the symptoms moderately in vivo, but drastically ex vivo. CONCLUSIONS Macrophage iron content in plaques is a critical factor in progression of atherosclerosis. The interaction of iron and lipid metabolism takes place in macrophage-rich atherosclerotic plaques. And we also suggest that altering intracellular iron levels in macrophages by systemic iron chelation or dietary iron restriction may be a potential supplementary strategy to limit or even regress the progression of atherosclerosis.
Collapse
Affiliation(s)
- Jing Cai
- Department of Vascular Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, People's Republic of China
| | - Meng Zhang
- Department of General Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, People's Republic of China
| | - Yutong Liu
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, People's Republic of China
| | - Huihui Li
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, People's Republic of China
| | - Longcheng Shang
- Department of General Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, People's Republic of China
| | - Tianze Xu
- Department of Vascular Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, People's Republic of China
| | - Zhipeng Chen
- Department of Vascular Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, People's Republic of China
| | - Fudi Wang
- Department of Nutrition, School of Public Health, School of Medicine, Zhejiang University, Hangzhou, China
| | - Tong Qiao
- Department of Vascular Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, People's Republic of China.
| | - Kuanyu Li
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, People's Republic of China.
| |
Collapse
|
22
|
Wunderer F, Traeger L, Sigurslid HH, Meybohm P, Bloch DB, Malhotra R. The role of hepcidin and iron homeostasis in atherosclerosis. Pharmacol Res 2020; 153:104664. [PMID: 31991168 PMCID: PMC7066581 DOI: 10.1016/j.phrs.2020.104664] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 12/20/2019] [Accepted: 01/24/2020] [Indexed: 02/06/2023]
Abstract
Atherosclerotic cardiovascular disease is a major burden on global health and a leading cause of death worldwide. The pathophysiology of this chronic disease is complex, involving inflammation, lipoprotein oxidation and accumulation, plaque formation, and calcification. In 1981, Dr. Jerome Sullivan formulated the 'Iron Hypothesis', suggesting that higher levels of stored iron promote cardiovascular diseases, whereas iron deficiency may have an atheroprotective effect. This hypothesis has stimulated research focused on clarifying the role of iron in the development of atherosclerosis. However, preclinical and clinical studies have produced contradictory results and the observation that patients with hemochromatosis do not appear to have an increased risk of atherosclerosis seemed incongruous with Sullivan's initial hypothesis. The 'paradox' of systemic iron overload not being accompanied by an increased risk for atherosclerosis led to a refinement of the iron hypothesis focusing on intracellular macrophage iron. More recent in vitro and animal studies have elucidated the complex signaling pathways regulating iron, with a particular focus on hepcidin, the master regulator of body iron homeostasis. Bone morphogenetic protein (BMP) signaling is the major pathway that is required for induction of hepcidin expression in response to increasing levels of iron. Strong links between iron homeostasis, BMP signaling, inflammation and atherosclerosis have been established in both mechanistic and human studies. This review summarizes the current understanding of the role of iron homeostasis and hepcidin in the development of atherosclerosis and discusses the BMP-hepcidin-ferroportin axis as a novel therapeutic target for the treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Florian Wunderer
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| | - Lisa Traeger
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Haakon H. Sigurslid
- Cardiovascular Research Center and the Cardiology Division of the Department of medicine, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Patrick Meybohm
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
- Department of Anaesthesiology, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Donald B. Bloch
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston
- Division of Rheumatology, Allergy and Immunology of the Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Rajeev Malhotra
- Cardiovascular Research Center and the Cardiology Division of the Department of medicine, Massachusetts General Hospital and Harvard Medical School, Boston
| |
Collapse
|
23
|
Lenz T, Nicol P, Castellanos MI, Engel LC, Lahmann AL, Alexiou C, Joner M. Small Dimension-Big Impact! Nanoparticle-Enhanced Non-Invasive and Intravascular Molecular Imaging of Atherosclerosis In Vivo. Molecules 2020; 25:E1029. [PMID: 32106607 PMCID: PMC7179220 DOI: 10.3390/molecules25051029] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 02/14/2020] [Accepted: 02/18/2020] [Indexed: 01/16/2023] Open
Abstract
Extensive translational research has provided considerable progress regarding the understanding of atherosclerosis pathophysiology over the last decades. In contrast, implementation of molecular in vivo imaging remains highly limited. In that context, nanoparticles represent a useful tool. Their variable shape and composition assure biocompatibility and stability within the environment of intended use, while the possibility of conjugating different ligands as well as contrast dyes enable targeting of moieties of interest on a molecular level and visualization throughout various imaging modalities. These characteristics have been exploited by a number of preclinical research approaches aimed at advancing understanding of vascular atherosclerotic disease, in order to improve identification of high-risk lesions prior to oftentimes fatal thromboembolic events. Furthermore, the combination of these targeted nanoparticles with therapeutic agents offers the potential of site-targeted drug delivery with minimized systemic secondary effects. This review gives an overview of different groups of targeted nanoparticles, designed for in vivo molecular imaging of atherosclerosis as well as an outlook on potential combined diagnostic and therapeutic applications.
Collapse
Affiliation(s)
- Tobias Lenz
- German Heart Centre Munich, Technical University of Munich, Lazarettstraße 36, 80636 Munich, Germany; (T.L.); (P.N.); (M.I.C.); (L.-C.E.); (A.L.L.)
| | - Philipp Nicol
- German Heart Centre Munich, Technical University of Munich, Lazarettstraße 36, 80636 Munich, Germany; (T.L.); (P.N.); (M.I.C.); (L.-C.E.); (A.L.L.)
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802 Munich, Germany
| | - Maria Isabel Castellanos
- German Heart Centre Munich, Technical University of Munich, Lazarettstraße 36, 80636 Munich, Germany; (T.L.); (P.N.); (M.I.C.); (L.-C.E.); (A.L.L.)
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802 Munich, Germany
| | - Leif-Christopher Engel
- German Heart Centre Munich, Technical University of Munich, Lazarettstraße 36, 80636 Munich, Germany; (T.L.); (P.N.); (M.I.C.); (L.-C.E.); (A.L.L.)
| | - Anna Lena Lahmann
- German Heart Centre Munich, Technical University of Munich, Lazarettstraße 36, 80636 Munich, Germany; (T.L.); (P.N.); (M.I.C.); (L.-C.E.); (A.L.L.)
| | - Christoph Alexiou
- Department of Oto-rhino-laryngology, head and neck surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Professorship, University Hospital Erlangen, 91054 Erlangen, Germany;
| | - Michael Joner
- German Heart Centre Munich, Technical University of Munich, Lazarettstraße 36, 80636 Munich, Germany; (T.L.); (P.N.); (M.I.C.); (L.-C.E.); (A.L.L.)
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802 Munich, Germany
| |
Collapse
|
24
|
Milutinović A, Šuput D, Zorc-Pleskovič R. Pathogenesis of atherosclerosis in the tunica intima, media, and adventitia of coronary arteries: An updated review. Bosn J Basic Med Sci 2020; 20:21-30. [PMID: 31465719 PMCID: PMC7029210 DOI: 10.17305/bjbms.2019.4320] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 07/31/2019] [Indexed: 02/07/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory disease of arteries and it affects the structure and function of all three layers of the coronary artery wall. Current theories suggest that the dysfunction of endothelial cells is one of the initial steps in the development of atherosclerosis. The view that the tunica intima normally consists of a single layer of endothelial cells attached to the subendothelial layer and internal elastic membrane has been questioned in recent years. The structure of intima changes with age and it becomes multilayered due to migration of smooth muscle cells from the media to intima. At this stage, the migration and proliferation of smooth muscle cells do not cause pathological changes in the intima. The multilayering of intima is classically considered to be an important stage in the development of atherosclerosis, but in fact atherosclerotic plaques develop only focally due to the interplay of various processes that involve the resident and invading inflammatory cells. The tunica media consists of multiple layers of smooth muscle cells that produce the extracellular matrix, and this layer normally does not contain microvessels. During the development of atherosclerosis, the microvessels from the tunica adventitia or from the lumen may penetrate thickened media to provide nutrition and oxygenation. According to some theories, the endothelial dysfunction of these nutritive vessels may significantly contribute to the atherosclerosis of coronary arteries. The adventitia contains fibroblasts, progenitor cells, immune cells, microvessels, and adrenergic nerves. The degree of inflammatory cell infiltration into the adventitia, which can lead to the formation of tertiary lymphoid organs, correlates with the severity of atherosclerotic plaques. Coronary arteries are surrounded by perivascular adipose tissue that also participates in the atherosclerotic process.
Collapse
Affiliation(s)
- Aleksandra Milutinović
- Institute of Histology and Embryology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia.
| | - Dušan Šuput
- Institute of Pathophysiology, Medical Faculty, University of Ljubljana, Ljubljana, Slovenia.
| | - Ruda Zorc-Pleskovič
- Institute of Histology and Embryology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia; International Center for Cardiovascular Diseases MC Medicor d.d., Izola, Slovenia.
| |
Collapse
|
25
|
Wu C, Daugherty A, Lu HS. Updates on Approaches for Studying Atherosclerosis. Arterioscler Thromb Vasc Biol 2020; 39:e108-e117. [PMID: 30917052 DOI: 10.1161/atvbaha.119.312001] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Congqing Wu
- From the Saha Cardiovascular Research Center (C.W., A.D., H.S.L.), University of Kentucky, Lexington
| | - Alan Daugherty
- From the Saha Cardiovascular Research Center (C.W., A.D., H.S.L.), University of Kentucky, Lexington.,Department of Physiology (A.D., H.S.L.), University of Kentucky, Lexington
| | - Hong S Lu
- From the Saha Cardiovascular Research Center (C.W., A.D., H.S.L.), University of Kentucky, Lexington.,Department of Physiology (A.D., H.S.L.), University of Kentucky, Lexington
| |
Collapse
|
26
|
Malhotra R, Wunderer F, Barnes HJ, Bagchi A, Buswell MD, O'Rourke CD, Slocum CL, Ledsky CD, Peneyra KM, Sigurslid H, Corman B, Johansson KB, Rhee DK, Bloch KD, Bloch DB. Hepcidin Deficiency Protects Against Atherosclerosis. Arterioscler Thromb Vasc Biol 2019; 39:178-187. [PMID: 30587002 DOI: 10.1161/atvbaha.118.312215] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Objective- Inflammatory stimuli enhance the progression of atherosclerotic disease. Inflammation also increases the expression of hepcidin, a hormonal regulator of iron homeostasis, which decreases intestinal iron absorption, reduces serum iron levels and traps iron within macrophages. The role of macrophage iron in the development of atherosclerosis remains incompletely understood. The objective of this study was to investigate the effects of hepcidin deficiency and decreased macrophage iron on the development of atherosclerosis. Approach and Results- Hepcidin- and LDL (low-density lipoprotein) receptor-deficient ( Hamp-/-/ Ldlr-/-) mice and Hamp+/+/ Ldlr-/- control mice were fed a high-fat diet for 21 weeks. Compared with control mice, Hamp-/-/ Ldlr-/- mice had decreased aortic macrophage activity and atherosclerosis. Because hepcidin deficiency is associated with both increased serum iron and decreased macrophage iron, the possibility that increased serum iron was responsible for decreased atherosclerosis in Hamp-/-/ Ldlr-/- mice was considered. Hamp+/+/ Ldlr-/- mice were treated with iron dextran so as to produce a 2-fold increase in serum iron. Increased serum iron did not decrease atherosclerosis in Hamp+/+/ Ldlr-/- mice. Aortic macrophages from Hamp-/-/ Ldlr-/- mice had less labile free iron and exhibited a reduced proinflammatory (M1) phenotype compared with macrophages from Hamp+/+/ Ldlr-/- mice. THP1 human macrophages treated with an iron chelator were used to model hepcidin deficiency in vitro. Treatment with an iron chelator reduced LPS (lipopolysaccharide)-induced M1 phenotypic expression and decreased uptake of oxidized LDL. Conclusions- In summary, in a hyperlipidemic mouse model, hepcidin deficiency was associated with decreased macrophage iron, a reduced aortic macrophage inflammatory phenotype and protection from atherosclerosis. The results indicate that decreasing hepcidin activity, with the resulting decrease in macrophage iron, may prove to be a novel strategy for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Rajeev Malhotra
- From the Cardiovascular Research Center and Cardiology Division of the Department of Medicine (R.M., H.J.B., M.D.B., C.L.S., H.S., D.K.R., K.D.B.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Florian Wunderer
- the Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care, and Pain Medicine (F.W., A.B., C.D.O., C.D.L., K.M.P., B.C., K.B.J., K.D.B., D.B.B.), Massachusetts General Hospital and Harvard Medical School, Boston.,Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Frankfurt am Main, Germany (F.W.)
| | - Hanna J Barnes
- From the Cardiovascular Research Center and Cardiology Division of the Department of Medicine (R.M., H.J.B., M.D.B., C.L.S., H.S., D.K.R., K.D.B.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Aranya Bagchi
- the Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care, and Pain Medicine (F.W., A.B., C.D.O., C.D.L., K.M.P., B.C., K.B.J., K.D.B., D.B.B.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Mary D Buswell
- From the Cardiovascular Research Center and Cardiology Division of the Department of Medicine (R.M., H.J.B., M.D.B., C.L.S., H.S., D.K.R., K.D.B.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Caitlin D O'Rourke
- the Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care, and Pain Medicine (F.W., A.B., C.D.O., C.D.L., K.M.P., B.C., K.B.J., K.D.B., D.B.B.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Charles L Slocum
- From the Cardiovascular Research Center and Cardiology Division of the Department of Medicine (R.M., H.J.B., M.D.B., C.L.S., H.S., D.K.R., K.D.B.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Clara D Ledsky
- the Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care, and Pain Medicine (F.W., A.B., C.D.O., C.D.L., K.M.P., B.C., K.B.J., K.D.B., D.B.B.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Kathryn M Peneyra
- the Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care, and Pain Medicine (F.W., A.B., C.D.O., C.D.L., K.M.P., B.C., K.B.J., K.D.B., D.B.B.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Haakon Sigurslid
- From the Cardiovascular Research Center and Cardiology Division of the Department of Medicine (R.M., H.J.B., M.D.B., C.L.S., H.S., D.K.R., K.D.B.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Benjamin Corman
- the Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care, and Pain Medicine (F.W., A.B., C.D.O., C.D.L., K.M.P., B.C., K.B.J., K.D.B., D.B.B.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Kimberly B Johansson
- the Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care, and Pain Medicine (F.W., A.B., C.D.O., C.D.L., K.M.P., B.C., K.B.J., K.D.B., D.B.B.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - David K Rhee
- From the Cardiovascular Research Center and Cardiology Division of the Department of Medicine (R.M., H.J.B., M.D.B., C.L.S., H.S., D.K.R., K.D.B.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Kenneth D Bloch
- From the Cardiovascular Research Center and Cardiology Division of the Department of Medicine (R.M., H.J.B., M.D.B., C.L.S., H.S., D.K.R., K.D.B.), Massachusetts General Hospital and Harvard Medical School, Boston.,the Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care, and Pain Medicine (F.W., A.B., C.D.O., C.D.L., K.M.P., B.C., K.B.J., K.D.B., D.B.B.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Donald B Bloch
- the Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care, and Pain Medicine (F.W., A.B., C.D.O., C.D.L., K.M.P., B.C., K.B.J., K.D.B., D.B.B.), Massachusetts General Hospital and Harvard Medical School, Boston.,Division of Rheumatology, Allergy and Immunology of the Department of Medicine (D.B.B.), Massachusetts General Hospital and Harvard Medical School, Boston
| |
Collapse
|
27
|
Wu Y, Zhang Y, Dai L, Wang Q, Xue L, Su Z, Zhang C. An apoptotic body-biomimic liposome in situ upregulates anti-inflammatory macrophages for stabilization of atherosclerotic plaques. J Control Release 2019; 316:236-249. [DOI: 10.1016/j.jconrel.2019.10.043] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 10/19/2019] [Accepted: 10/22/2019] [Indexed: 01/31/2023]
|
28
|
Cornelissen A, Guo L, Sakamoto A, Virmani R, Finn AV. New insights into the role of iron in inflammation and atherosclerosis. EBioMedicine 2019; 47:598-606. [PMID: 31416722 PMCID: PMC6796517 DOI: 10.1016/j.ebiom.2019.08.014] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 08/05/2019] [Accepted: 08/06/2019] [Indexed: 12/24/2022] Open
Abstract
Iron is fundamental for life-essential processes. However, it can also cause oxidative damage, which is thought to trigger numerous pathologies, including cardiovascular diseases. The role of iron in the pathogenesis of atherosclerosis is still not completely understood. Macrophages are both key players in the handling of iron throughout the body and in the onset, progression and destabilization of atherosclerotic plaques. Iron itself might impact atherosclerosis through its effects on macrophages. However, while targeting iron metabolism within macrophages may have some beneficial effects on preventing atherosclerotic plaque progression there may also be negative consequences. Thus, the prevailing view of iron being capable of accelerating the progression of coronary disease through lipid peroxidation may not fully take into account the multi-faceted role of iron in pathogenesis of atherosclerosis. In this review, we will summarize the current understanding of iron metabolism in the context of the complex interplay between iron, inflammation, and atherosclerosis.
Collapse
Affiliation(s)
- Anne Cornelissen
- CVPath Institute, Gaithersburg, MD, USA; University Hospital RWTH Aachen, Department of Cardiology, Aachen, Germany.
| | - Liang Guo
- CVPath Institute, Gaithersburg, MD, USA.
| | | | | | - Aloke V Finn
- CVPath Institute, Gaithersburg, MD, USA; University of Maryland, School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
29
|
Duran EK, Cook NR, Bobadilla M, Kim E, Manson JE, Buring JE, Ridker PM, Pradhan AD. Plasma Placental Growth Factor Concentrations Are Elevated Well in Advance of Type 2 Diabetes Mellitus Onset: Prospective Data From the WHS. J Am Heart Assoc 2019; 8:e012790. [PMID: 31322059 PMCID: PMC6761678 DOI: 10.1161/jaha.119.012790] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Background Pathologic angiogenesis is a hallmark of type 2 diabetes mellitus (T2DM) microvascular complications and may modulate adipogenesis and precede the onset of clinical diabetes mellitus; however, longitudinal data are unavailable. Placental growth factor is a potent proangiogenic factor that stimulates the formation of mature and durable vessels but is understudied in human diseases. Methods and Results We conducted a prospective case‐cohort study of baseline placental growth factor and incident T2DM within the WHS (Women's Health Study). A random sample of incident T2DM cases (n=491) occurring over a 15‐year follow‐up period was selected and compared with a reference subcohort (n=561). Case subjects were matched to the reference risk set on 5‐year age groups and race. All subjects in this analysis were required to have a hemoglobin A1c <6.5% at WHS enrollment. Median baseline levels of placental growth factor were higher in case subjects compare to the reference subcohort (18.0 pg/mL versus 17.2 pg/mL) but were only weakly correlated with glycemic measures and not associated with obesity. The risk of diabetes mellitus increased across placental growth factor quartile in the base model (hazard ratios, 1.00, 1.14, 1.46, and 2.14; P‐trend<0.001) and in multivariable‐adjusted models accounting for clinical T2DM risk factors (hazard ratios, 1.00, 1.17, 1.45, and 2.61; P‐trend<0.001). These findings were not substantially altered by further adjustment for high‐sensitivity C‐reactive protein, hemoglobin A1c, or fasting insulin and remained robust in sensitivity analyses excluding those diagnosed within 2 years of enrollment and those with baseline hemoglobin A1c ≥6.0%. Conclusions Elevated placental growth factor levels are associated with future T2DM independent of traditional risk factors, measures of glycemia, insulin resistance, and high‐sensitivity C‐reactive protein. These prospective data suggest that pathologic angiogenesis may occur well before the clinical onset of T2DM and thus may have relevance to vascular complications of this disease. Clinical Trial Registration URL: http://www.clinicaltrials.gov. Unique identifier: NCT00000479.
Collapse
Affiliation(s)
- Edward K Duran
- Division of Preventive Medicine Brigham and Women's Hospital Harvard Medical School Boston MA
| | - Nancy R Cook
- Division of Preventive Medicine Brigham and Women's Hospital Harvard Medical School Boston MA
| | | | - Eunjung Kim
- Division of Preventive Medicine Brigham and Women's Hospital Harvard Medical School Boston MA
| | - JoAnn E Manson
- Division of Preventive Medicine Brigham and Women's Hospital Harvard Medical School Boston MA
| | - Julie E Buring
- Division of Preventive Medicine Brigham and Women's Hospital Harvard Medical School Boston MA
| | - Paul M Ridker
- Division of Preventive Medicine Brigham and Women's Hospital Harvard Medical School Boston MA.,Division of Cardiovascular Medicine Brigham and Women's Hospital Harvard Medical School Boston MA
| | - Aruna D Pradhan
- Division of Preventive Medicine Brigham and Women's Hospital Harvard Medical School Boston MA.,Division of Cardiovascular Medicine VA Boston Medical Center Boston MA
| |
Collapse
|
30
|
Aplin AC, Nicosia RF. The plaque-aortic ring assay: a new method to study human atherosclerosis-induced angiogenesis. Angiogenesis 2019; 22:421-431. [DOI: 10.1007/s10456-019-09667-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 03/26/2019] [Indexed: 12/18/2022]
|
31
|
Flores AM, Ye J, Jarr KU, Hosseini-Nassab N, Smith BR, Leeper NJ. Nanoparticle Therapy for Vascular Diseases. Arterioscler Thromb Vasc Biol 2019; 39:635-646. [PMID: 30786744 PMCID: PMC6436996 DOI: 10.1161/atvbaha.118.311569] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Nanoparticles promise to advance strategies to treat vascular disease. Since being harnessed by the cancer field to deliver safer and more effective chemotherapeutics, nanoparticles have been translated into applications for cardiovascular disease. Systemic exposure and drug-drug interactions remain a concern for nearly all cardiovascular therapies, including statins, antithrombotic, and thrombolytic agents. Moreover, off-target effects and poor bioavailability have limited the development of completely new approaches to treat vascular disease. Through the rational design of nanoparticles, nano-based delivery systems enable more efficient delivery of a drug to its therapeutic target or even directly to the diseased site, overcoming biological barriers and enhancing a drug's therapeutic index. In addition, advances in molecular imaging have led to the development of theranostic nanoparticles that may simultaneously act as carriers of both therapeutic and imaging payloads. The following is a summary of nanoparticle therapy for atherosclerosis, thrombosis, and restenosis and an overview of recent major advances in the targeted treatment of vascular disease.
Collapse
Affiliation(s)
- Alyssa M. Flores
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine., Hanover, NH
- Geisel School of Medicine at Dartmouth, Hanover, NH
| | - Jianqin Ye
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine., Hanover, NH
| | - Kai-Uwe Jarr
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine., Hanover, NH
| | - Niloufar Hosseini-Nassab
- Department of Radiology, Stanford University School of Medicine, Michigan State University, East Lansing, MI, USA
| | - Bryan R. Smith
- Department of Biomedical Engineering and Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
| | - Nicholas J. Leeper
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine., Hanover, NH
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA
- Stanford Cardiovascular Institute, Stanford, CA 94305, USA
| |
Collapse
|
32
|
Jin F, Zheng X, Yang Y, Yao G, Ye L, Doeppner TR, Hermann DM, Wang H, Dai Y. Impairment of hypoxia-induced angiogenesis by LDL involves a HIF-centered signaling network linking inflammatory TNFα and angiogenic VEGF. Aging (Albany NY) 2019; 11:328-349. [PMID: 30659163 PMCID: PMC6366960 DOI: 10.18632/aging.101726] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 12/12/2018] [Indexed: 02/06/2023]
Abstract
Hypoxia inducible factors (HIFs) mediate angiogenesis via up-regulation of various pro-angiogenic factors (particularly VEGF) in response to hypoxia. Here, we report that hypoxia unexpectedly induced robust production of the pro-inflammatory factor TNFα by endothelial cells (ECs), suggesting an autocrine loop that in turn activated HIFs via an NF-κB-dependent process, resulting in production of VEGF and thereby promotion of angiogenesis. In contrast, low-density lipoprotein (LDL) prevented expression of HIFs in ECs exposed to either hypoxia or TNFα, while knockdown of either HIF-1α or HIF-2α strikingly attenuated hypoxia-induced production of VEGF by ECs as well as EC colony formation and tube formation. Significantly, LDL attenuated hypoxia-induced angiogenesis by disrupting the TNFα/NF-κB/HIF/VEGF signaling cascade via down-regulation of the TNF receptor TNF-R1, rather than TNFα itself, and multiple key components of both canonical and non-canonical NF-κB pathways. By doing so, LDL was able to either inhibit or down-regulate a wide spectrum of HIF-dependent pro-angiogenic downstream targets and signals. Together, these findings argue existence of a self-regulatory TNFα/NF-κB/HIF/VEGF signaling network in ECs, which mediates and fine-tones angiogenesis, at least in response to hypoxia. They also suggest that LDL impairs angiogenesis by disrupting this network, which might represent a novel mechanism underlying anti-angiogenic property of LDL.
Collapse
Affiliation(s)
- Fengyan Jin
- Department of Hematology, Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, China
| | - Xiangyu Zheng
- Department of Neurology, the First Hospital of Jilin University, Changchun, Jilin, China
| | - Yanping Yang
- Department of Hematology, Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, China
| | - Gang Yao
- Department of Neurology, the Second Affiliated Hospital of Jilin University, Changchun, Jilin, China
| | - Long Ye
- Laboratory of Cancer Precision Medicine, the First Hospital of Jilin University, Changchun, Jilin, China
| | - Thorsten R. Doeppner
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Dirk M. Hermann
- Department of Neurology, University Duisburg-Essen Medical School, Essen, Germany
| | - Haifeng Wang
- Department of Neurosurgery, the First Hospital of Jilin University, Changchun, Jilin, China
| | - Yun Dai
- Laboratory of Cancer Precision Medicine, the First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
33
|
VEGF-A/VEGFR-2 and FGF-2/FGFR-1 but not PDGF-BB/PDGFR-β play important roles in promoting immature and inflammatory intraplaque angiogenesis. PLoS One 2018; 13:e0201395. [PMID: 30125282 PMCID: PMC6101364 DOI: 10.1371/journal.pone.0201395] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Accepted: 07/13/2018] [Indexed: 12/18/2022] Open
Abstract
Various angiogenic factors have been shown to play important roles in intraplaque angiogenesis, while little is known about the dynamic expression change and interplay between various angiogenic factors and intraplaque angiogenesis under high cholesterol conditions. New Zealand rabbits underwent balloon injury of the abdominal artery and then were assigned to a control group (n = 15, normal chow) or high cholesterol group (n = 25, 1% high cholesterol diet). At weeks 4, 6, 8, 10, and 12 after acclimation, rabbits (high cholesterol group, n = 5; control group, n = 3) were euthanized. No lesions were observed in the control group. From week 4 to week 12, the expression of vascular endothelial growth factor A (VEGF-A), VEGF receptor 2 (VEGFR-2), fibroblast growth factor 2 (FGF-2), FGF receptor 1 (FGFR-1), platelet-derived growth factor-BB (PDGF-BB), and tumor necrosis factor alpha (TNF-α), the vulnerability index (VI) and the microvessel density (MVD) were significantly elevated in the high cholesterol group; however, PDGF receptor β (PDGFR-β) expression showed little change. Analysis by double-label immunofluorescence (CD31 and Ng2) and FITC-dextran indicated that the neovessels within the plaque were leaky due to a lack of pericytes. As indicated by Pearson’s correlation analysis, there was a highly positive correlation between the VI, MVD, macrophage content, and TNF-α level, and the levels of VEGF-A/VEGFR-2 and FGF-2/FGFR-1. However, no correlations were observed between PDGFR-β levels and the VI or MVD. High expression of VEGF-A/VEGFR-2 and FGF-2/FGFR-1 but not of PDGF-BB/PDGFR-β may contribute to immature and inflammatory intraplaque angiogenesis and plaque instability in a rabbit model of atherosclerosis.
Collapse
|
34
|
Melo NDOR, Juanes CDC, Alves MFDA, Silva ETM, Jamacaru FVF, Lemos TLGD, Dornelas CA. Effect of red propolis on hamster cheek pouch angiogenesis in a new sponge implant model. Acta Cir Bras 2018; 33:420-430. [DOI: 10.1590/s0102-865020180050000004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 04/13/2018] [Indexed: 12/22/2022] Open
|
35
|
Baetta R, Pontremoli M, Fernandez AM, Spickett CM, Banfi C. Reprint of: Proteomics in cardiovascular diseases: Unveiling sex and gender differences in the era of precision medicine. J Proteomics 2018; 178:57-72. [PMID: 29622522 DOI: 10.1016/j.jprot.2018.03.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 11/06/2017] [Accepted: 11/17/2017] [Indexed: 01/19/2023]
Abstract
Cardiovascular diseases (CVDs) represent the most important cause of mortality in women and in men. Contrary to the long-standing notion that the effects of the major risk factors on CVD outcomes are the same in both sexes, recent evidence recognizes new, potentially independent, sex/gender-related risk factors for CVDs, and sex/gender-differences in the clinical presentation of CVDs have been demonstrated. Furthermore, some therapeutic options may not be equally effective and safe in men and women. In this context, proteomics offers an extremely useful and versatile analytical platform for biomedical researches that expand from the screening of early diagnostic and prognostic biomarkers to the investigation of the molecular mechanisms underlying CDVs. In this review, we summarized the current applications of proteomics in the cardiovascular field, with emphasis on sex and gender-related differences in CVDs. SIGNIFICANCE Increasing evidence supports the profound effect of sex and gender on cardiovascular physio-pathology and the response to drugs. A clear understanding of the mechanisms underlying sexual dimorphisms in CVDs would not only improve our knowledge of the etiology of these diseases, but could also inform health policy makers and guideline committees in tailoring specific interventions for the prevention, treatment and management of CVDs in both men and women.
Collapse
|
36
|
Guo L, Akahori H, Harari E, Smith SL, Polavarapu R, Karmali V, Otsuka F, Gannon RL, Braumann RE, Dickinson MH, Gupta A, Jenkins AL, Lipinski MJ, Kim J, Chhour P, de Vries PS, Jinnouchi H, Kutys R, Mori H, Kutyna MD, Torii S, Sakamoto A, Choi CU, Cheng Q, Grove ML, Sawan MA, Zhang Y, Cao Y, Kolodgie FD, Cormode DP, Arking DE, Boerwinkle E, Morrison AC, Erdmann J, Sotoodehnia N, Virmani R, Finn AV. CD163+ macrophages promote angiogenesis and vascular permeability accompanied by inflammation in atherosclerosis. J Clin Invest 2018; 128:1106-1124. [PMID: 29457790 PMCID: PMC5824873 DOI: 10.1172/jci93025] [Citation(s) in RCA: 234] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 01/02/2018] [Indexed: 12/26/2022] Open
Abstract
Intake of hemoglobin by the hemoglobin-haptoglobin receptor CD163 leads to a distinct alternative non-foam cell antiinflammatory macrophage phenotype that was previously considered atheroprotective. Here, we reveal an unexpected but important pathogenic role for these macrophages in atherosclerosis. Using human atherosclerotic samples, cultured cells, and a mouse model of advanced atherosclerosis, we investigated the role of intraplaque hemorrhage on macrophage function with respect to angiogenesis, vascular permeability, inflammation, and plaque progression. In human atherosclerotic lesions, CD163+ macrophages were associated with plaque progression, microvascularity, and a high level of HIF1α and VEGF-A expression. We observed irregular vascular endothelial cadherin in intraplaque microvessels surrounded by CD163+ macrophages. Within these cells, activation of HIF1α via inhibition of prolyl hydroxylases promoted VEGF-mediated increases in intraplaque angiogenesis, vascular permeability, and inflammatory cell recruitment. CD163+ macrophages increased intraplaque endothelial VCAM expression and plaque inflammation. Subjects with homozygous minor alleles of the SNP rs7136716 had elevated microvessel density, increased expression of CD163 in ruptured coronary plaques, and a higher risk of myocardial infarction and coronary heart disease in population cohorts. Thus, our findings highlight a nonlipid-driven mechanism by which alternative macrophages promote plaque angiogenesis, leakiness, inflammation, and progression via the CD163/HIF1α/VEGF-A pathway.
Collapse
MESH Headings
- Adult
- Animals
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Antigens, Differentiation, Myelomonocytic/genetics
- Antigens, Differentiation, Myelomonocytic/metabolism
- Atherosclerosis/metabolism
- Coronary Disease/metabolism
- Coronary Vessels/metabolism
- Disease Progression
- Female
- Hemoglobins/metabolism
- Humans
- Inflammation/metabolism
- Macrophages/cytology
- Macrophages/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Middle Aged
- Myocardial Infarction/metabolism
- Neovascularization, Pathologic
- Oxidative Stress
- Permeability
- Phenotype
- Polymorphism, Single Nucleotide
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- Liang Guo
- CVPath Institute, Gaithersburg, Maryland, USA
| | - Hirokuni Akahori
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | | | | | - Rohini Polavarapu
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Vinit Karmali
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | | | | | | | | | - Anuj Gupta
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Audrey L. Jenkins
- MedStar Heart and Vascular Institute and MedStar Health Research Institute, MedStar Washington Hospital Center, Washington, DC, USA
| | - Michael J. Lipinski
- MedStar Heart and Vascular Institute and MedStar Health Research Institute, MedStar Washington Hospital Center, Washington, DC, USA
| | - Johoon Kim
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Peter Chhour
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Paul S. de Vries
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | | | | | | | | | - Sho Torii
- CVPath Institute, Gaithersburg, Maryland, USA
| | | | - Cheol Ung Choi
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Qi Cheng
- CVPath Institute, Gaithersburg, Maryland, USA
| | - Megan L. Grove
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Mariem A. Sawan
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Yin Zhang
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | - Yihai Cao
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | | | - David P. Cormode
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Dan E. Arking
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Eric Boerwinkle
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, Texas, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, USA
| | - Alanna C. Morrison
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Jeanette Erdmann
- Institute for Cardiogenetics, University of Lübeck, and German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Germany
| | - Nona Sotoodehnia
- Division of Cardiology, Department of Medicine and Epidemiology, Cardiovascular Health Research Unit, University of Washington, Seattle, Washington, USA
| | | | - Aloke V. Finn
- CVPath Institute, Gaithersburg, Maryland, USA
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
37
|
Pourcet B, Staels B. Alternative macrophages in atherosclerosis: not always protective! J Clin Invest 2018; 128:910-912. [PMID: 29457787 DOI: 10.1172/jci120123] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disease of the vasculature that is initiated by cholesterol deposition into the arterial wall, which triggers the infiltration of immune and inflammatory cells, including monocytes and macrophages. As atherosclerotic plaques progress, localized hypoxia promotes compensatory angiogenesis from the vasa vasorum. Immature neovessels are prone to leakage, thus destabilizing the plaque and leading to intraplaque hemorrhage. Macrophages with different phenotypes, ranging from classical inflammatory subtypes to alternatively activated antiinflammatory macrophages, have been identified in atherosclerotic lesions. Antiinflammatory hemoglobin-scavenging CD163+ macrophages are present in neovessel- and hemorrhage-rich areas; however, the role of these macrophages in atherogenesis has been unclear. In this issue of the JCI, Guo, Akahori, and colleagues show that CD163+ macrophages promote angiogenesis, vessel permeability, and leucocyte infiltration in human and mouse atherosclerotic lesions through a mechanism involving hemoglobin:haptoglobin/CD163/HIF1α-mediated VEGF induction. This study thus identifies proatherogenic properties of CD163+ macrophages, which previously were thought to be beneficial.
Collapse
|
38
|
Yan A, Yue T, Li L, Li W, Li Q, Li J. Bromodomain-containing protein 7 deficiency augments atherosclerotic lesions in ApoE -/- mice. Biochem Biophys Res Commun 2018; 495:2202-2208. [PMID: 29273506 DOI: 10.1016/j.bbrc.2017.12.103] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Accepted: 12/18/2017] [Indexed: 11/16/2022]
Abstract
Atherosclerotic plaque formation is characterized by the persistence of lipid-laden macrophages on the inner walls of arteries. Chronic inflammation and imbalanced macrophage function are likely to play a critical role. Herein, we investigated whether bromodomain-containing protein 7 (Brd7), a member of the bromodomain-containing protein family, regulates atherosclerosis, and if so, which mechanisms are responsible for the process. We found that Brd7 is expressed in mouse atherosclerotic plaques, and mostly in macrophages. Inhibition of Brd7 accelerates atherosclerotic lesion formation in ApoE-/- mice by promoting NF-κB-mediated inflammation. Furthermore, Brd7 inhibition alters the phenotype of macrophages and promotes plaque instability, at least partly via STAT6 signaling. Our data define a previously undescribed role of Brd7 in the development of atherosclerosis.
Collapse
Affiliation(s)
- Aiguo Yan
- Department of Geriatrics, Zibo Center Hospital, Zibo, Shandong 255036, China
| | - Tao Yue
- Department of Geriatrics, Zibo Center Hospital, Zibo, Shandong 255036, China.
| | - Li Li
- Department of Geriatrics, Zibo Center Hospital, Zibo, Shandong 255036, China.
| | - Wei Li
- Department of Geriatrics, Zibo Center Hospital, Zibo, Shandong 255036, China
| | - Qinghua Li
- Department of Emergency, Zibo Center Hospital, Zibo, Shandong 255036, China
| | - Jun Li
- Department of Neurosurgery, Zibo Center Hospital, Zibo, Shandong 255036, China
| |
Collapse
|
39
|
Chen YL, Liu FQ, Guo Y, Cheng J, Yang L, Lu M, Li P, Xu J, Yu T, Wang ZG, Cao Y, Ran HT. PA/US dual-modality imaging to guide VEGFR-2 targeted photothermal therapy using ZnPc-/PFH-loaded polymeric nanoparticles. Biomater Sci 2018; 6:2130-2143. [PMID: 29916500 DOI: 10.1039/c8bm00213d] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Angiogenesis is a common pathological characteristic of many solid tumors and vulnerable atherosclerotic plaques.
Collapse
|
40
|
Baetta R, Pontremoli M, Martinez Fernandez A, Spickett CM, Banfi C. Proteomics in cardiovascular diseases: Unveiling sex and gender differences in the era of precision medicine. J Proteomics 2017; 173:62-76. [PMID: 29180046 DOI: 10.1016/j.jprot.2017.11.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 11/06/2017] [Accepted: 11/17/2017] [Indexed: 02/07/2023]
Abstract
Cardiovascular diseases (CVDs) represent the most important cause of mortality in women and in men. Contrary to the long-standing notion that the effects of the major risk factors on CVD outcomes are the same in both sexes, recent evidence recognizes new, potentially independent, sex/gender-related risk factors for CVDs, and sex/gender-differences in the clinical presentation of CVDs have been demonstrated. Furthermore, some therapeutic options may not be equally effective and safe in men and women. In this context, proteomics offers an extremely useful and versatile analytical platform for biomedical researches that expand from the screening of early diagnostic and prognostic biomarkers to the investigation of the molecular mechanisms underlying CDVs. In this review, we summarized the current applications of proteomics in the cardiovascular field, with emphasis on sex and gender-related differences in CVDs. SIGNIFICANCE Increasing evidence supports the profound effect of sex and gender on cardiovascular physio-pathology and the response to drugs. A clear understanding of the mechanisms underlying sexual dimorphisms in CVDs would not only improve our knowledge of the etiology of these diseases, but could also inform health policy makers and guideline committees in tailoring specific interventions for the prevention, treatment and management of CVDs in both men and women.
Collapse
|