1
|
McGonagle D, Giryes S. An immunology model for accelerated coronary atherosclerosis and unexplained sudden death in the COVID-19 era. Autoimmun Rev 2024; 23:103642. [PMID: 39313122 DOI: 10.1016/j.autrev.2024.103642] [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: 07/28/2024] [Revised: 09/09/2024] [Accepted: 09/17/2024] [Indexed: 09/25/2024]
Abstract
The immunological basis for cardiac deaths remote from potential triggering viral infection, including SARS-CoV-2 infection, remains enigmatic. Cardiac surface inflammation, including the pericardium, epicardium and superficial myocardium with associated coronary artery vasculitis in infant Kawasaki Disease (KD) and multisystem inflammatory syndrome in children (MIS-C) is well recognised. In this perspective, we review the evidence pointing towards prominent post-viral infection related epicardial inflammation in older subjects, resulting in atherosclerotic plaque destabilisation with seemingly unrelated myocardial infarction that may be temporally distant from the actual infectious triggers. Cardiac surface inflammation in the relatively immune cell rich tissues in the territory though where the coronary arteries traverse is common in the adult post-COVD pneumonic phase and is also well described after vaccination including pre-COVID era vaccinations. Immunologically, the pericardium/epicardium tissue was known to be critical for coronary artery territory atherosclerotic disease prior to the COVID-19 era and may be linked to the involvement of the coronary artery vasa vasorum that physiologically oxygenates the coronary artery walls. We highlight how viral infection or vaccination-associated diffuse epicardial tissue inflammation adjacent to the coronary artery vasa vasorum territory represents a critical unifying concept for seemingly unrelated fatal coronary artery atherosclerotic disease, that could occur soon after or remote from infection or vaccination in adults. Mechanistically, such epicardial inflammation impacting coronary artery vasa vasorum immunity acts as gateways towards the slow destabilisation of pre-existing atherosclerotic plaques, with resultant myocardial infarction and other cardiac pathology. This model offers immunologists and academic cardiologists an immunopathological roadmap between innocuous viral infections or vaccinations and seemingly temporally remote "unrelated" atherosclerotic disease with excess cardiac deaths.
Collapse
Affiliation(s)
- Dennis McGonagle
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), University of Leeds, Leeds, United Kingdom; National Institute for Health Research (NIHR) Leeds Biomedical Research Centre (BRC), Leeds Teaching Hospitals, Leeds, United Kingdom.
| | - Sami Giryes
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), University of Leeds, Leeds, United Kingdom; B. Shine Rheumatology Institute, Rambam Healthcare Campus, Haifa, Israel
| |
Collapse
|
2
|
Gerasimovskaya E, Patil RS, Davies A, Maloney ME, Simon L, Mohamed B, Cherian-Shaw M, Verin AD. Extracellular purines in lung endothelial permeability and pulmonary diseases. Front Physiol 2024; 15:1450673. [PMID: 39234309 PMCID: PMC11372795 DOI: 10.3389/fphys.2024.1450673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 08/05/2024] [Indexed: 09/06/2024] Open
Abstract
The purinergic signaling system is an evolutionarily conserved and critical regulatory circuit that maintains homeostatic balance across various organ systems and cell types by providing compensatory responses to diverse pathologies. Despite cardiovascular diseases taking a leading position in human morbidity and mortality worldwide, pulmonary diseases represent significant health concerns as well. The endothelium of both pulmonary and systemic circulation (bronchial vessels) plays a pivotal role in maintaining lung tissue homeostasis by providing an active barrier and modulating adhesion and infiltration of inflammatory cells. However, investigations into purinergic regulation of lung endothelium have remained limited, despite widespread recognition of the role of extracellular nucleotides and adenosine in hypoxic, inflammatory, and immune responses within the pulmonary microenvironment. In this review, we provide an overview of the basic aspects of purinergic signaling in vascular endothelium and highlight recent studies focusing on pulmonary microvascular endothelial cells and endothelial cells from the pulmonary artery vasa vasorum. Through this compilation of research findings, we aim to shed light on the emerging insights into the purinergic modulation of pulmonary endothelial function and its implications for lung health and disease.
Collapse
Affiliation(s)
| | - Rahul S Patil
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Adrian Davies
- Department of Internal Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - McKenzie E Maloney
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
- Office of Academic Affairs, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Liselle Simon
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Basmah Mohamed
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Mary Cherian-Shaw
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Alexander D Verin
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States
| |
Collapse
|
3
|
Danilov VV, Laptev VV, Klyshnikov KY, Stepanov AD, Bogdanov LA, Antonova LV, Krivkina EO, Kutikhin AG, Ovcharenko EA. ML-driven segmentation of microvascular features during histological examination of tissue-engineered vascular grafts. Front Bioeng Biotechnol 2024; 12:1411680. [PMID: 38988863 PMCID: PMC11233802 DOI: 10.3389/fbioe.2024.1411680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 05/21/2024] [Indexed: 07/12/2024] Open
Abstract
Introduction The development of next-generation tissue-engineered medical devices such as tissue-engineered vascular grafts (TEVGs) is a leading trend in translational medicine. Microscopic examination is an indispensable part of animal experimentation, and histopathological analysis of regenerated tissue is crucial for assessing the outcomes of implanted medical devices. However, the objective quantification of regenerated tissues can be challenging due to their unusual and complex architecture. To address these challenges, research and development of advanced ML-driven tools for performing adequate histological analysis appears to be an extremely promising direction. Methods We compiled a dataset of 104 representative whole slide images (WSIs) of TEVGs which were collected after a 6-month implantation into the sheep carotid artery. The histological examination aimed to analyze the patterns of vascular tissue regeneration in TEVGs in situ. Having performed an automated slicing of these WSIs by the Entropy Masker algorithm, we filtered and then manually annotated 1,401 patches to identify 9 histological features: arteriole lumen, arteriole media, arteriole adventitia, venule lumen, venule wall, capillary lumen, capillary wall, immune cells, and nerve trunks. To segment and quantify these features, we rigorously tuned and evaluated the performance of six deep learning models (U-Net, LinkNet, FPN, PSPNet, DeepLabV3, and MA-Net). Results After rigorous hyperparameter optimization, all six deep learning models achieved mean Dice Similarity Coefficients (DSC) exceeding 0.823. Notably, FPN and PSPNet exhibited the fastest convergence rates. MA-Net stood out with the highest mean DSC of 0.875, demonstrating superior performance in arteriole segmentation. DeepLabV3 performed well in segmenting venous and capillary structures, while FPN exhibited proficiency in identifying immune cells and nerve trunks. An ensemble of these three models attained an average DSC of 0.889, surpassing their individual performances. Conclusion This study showcases the potential of ML-driven segmentation in the analysis of histological images of tissue-engineered vascular grafts. Through the creation of a unique dataset and the optimization of deep neural network hyperparameters, we developed and validated an ensemble model, establishing an effective tool for detecting key histological features essential for understanding vascular tissue regeneration. These advances herald a significant improvement in ML-assisted workflows for tissue engineering research and development.
Collapse
Affiliation(s)
| | - Vladislav V Laptev
- Siberian State Medical University, Tomsk, Russia
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - Kirill Yu Klyshnikov
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - Alexander D Stepanov
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - Leo A Bogdanov
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - Larisa V Antonova
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - Evgenia O Krivkina
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - Anton G Kutikhin
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - Evgeny A Ovcharenko
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| |
Collapse
|
4
|
Yamamoto K, Mizutani K, Akiyama T, Nogawa H, Toda M. Vasa vasorum: The role in intracranial physiology and pathophysiology. Surg Neurol Int 2024; 15:188. [PMID: 38974550 PMCID: PMC11225505 DOI: 10.25259/sni_214_2024] [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: 03/23/2024] [Accepted: 05/04/2024] [Indexed: 07/09/2024] Open
Abstract
Background Vasa vasorum (VVs) is a Latin word representing vessels of vessels. VVs are usually found on the adventitia of the parent vessel and infrequently reach the media and intima, depending on the size and type of the parent vessels and physiological and pathological conditions. The VVs include arteries, capillaries, veins, and lymphatic vessels, involving the oxygenation and nourishment of the vessel's wall to sustain its healthy state. Accumulated studies have revealed that VVs are involved in various intracranial lesions, including atherosclerotic diseases, aneurysms, and shunt diseases. The current review aims to review and integrate past and recent findings and knowledge on VVs and to facilitate our understanding of VVs and intracranial pathology involving VVs. Methods A literature review was carried out with a focus on the role of VVs by searching the Pubmed database. Results We identified 71 articles that discuss the role of VVs. We discussed the anatomical structure, physiological significance, and pathological significance of the VV. Conclusion VV is not only involved in the nutrition and metabolism of the vascular wall but is also deeply involved in the pathogenesis of inflammation, ischemia, and thrombosis of the vascular wall. In addition, in the central nervous system, intracranial vascular wall nutrient particularities and VVs are closely related to the pathogenesis of cerebral aneurysms, subarachnoid hemorrhage, arteriovenous shunt disease, atherosclerotic lesions, and other conditions.
Collapse
Affiliation(s)
| | - Katsuhiro Mizutani
- Department of Neurosurgery, Keio University, School of Medicine, Shinjuku, Japan
| | | | | | | |
Collapse
|
5
|
Matsui C, Tsukuura R, Sakai H, Escandón JM, Mohammad A, Yamamoto T. Evaluation of the Superficial Collecting Lymph Vessels' Vasa Vasorum in Lymphoedematous Limbs Using Video Capillaroscopy. Eur J Vasc Endovasc Surg 2024; 67:1008-1014. [PMID: 38000693 DOI: 10.1016/j.ejvs.2023.11.035] [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: 06/26/2023] [Revised: 10/06/2023] [Accepted: 11/17/2023] [Indexed: 11/26/2023]
Abstract
OBJECTIVE The pre-collecting and collecting lymph vessels have smooth muscle cells, and sufficient perfusion is vital to maintain their function. Although the vasa vasorum of the collecting lymph vessels (VVCL) have been histologically investigated, little is known about their physiology. This study aimed to investigate the relationship between morphology and blood flow of the VVCL in lymphoedematous limbs. METHODS Medical records of lower extremity lymphoedema patients who underwent video capillaroscopy observation during supermicrosurgical lymphaticovenous anastomosis (LVA) surgery were reviewed. The collecting lymph vessels, dissected for LVA, were examined under video capillaroscopy (GOKO Bscan-ZD, GOKO Imaging Devices Co., Japan) with a magnification of 175x and 620x. Blood flow velocity of the VVCL was calculated by measuring the red blood cell movement using software (GOKO-VIP ver. 1.0.0.4, GOKO Imaging Devices Co., Japan). Based on the video capillaroscopy findings, the VVCL were grouped according to their morphology; the VVCL morphology types and blood flow velocity were then compared according to the lymphosclerosis severity grade. RESULTS Sixty-seven lymph vessels in 20 lower extremity lymphoedema patients were evaluated, including s0 in 19 (28.4%), s1 in 34 (50.7%), s2 in 10 (14.9%), and s3 in four (6.0%) lymph vessels. The VVCLs were grouped into four types: type 1 (n = 4), type 2 (n = 37), type 3 (n = 19), and type 4 (n = 7). Blood flow velocity of the VVCL ranged 0 - 189.3 μm/sec (average 26.40 μm/sec). There were statistically significant differences in VVCL morphology (p < .001) and blood flow velocity (p < .001) according to lymphosclerotic severity. CONCLUSION Vasa vasorum of the collecting lymph vessels could be grouped into four types with different characteristics. Morphological and physiological changes of the VVCL were related to sclerotic changes of the collecting lymph vessels.
Collapse
Affiliation(s)
- Chihiro Matsui
- Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, Tokyo, Japan; Department of Plastic and Reconstructive Surgery, National Centre for Global Health and Medicine, Tokyo, Japan
| | - Reiko Tsukuura
- Department of Plastic and Reconstructive Surgery, National Centre for Global Health and Medicine, Tokyo, Japan
| | - Hayahito Sakai
- Department of Plastic and Reconstructive Surgery, National Centre for Global Health and Medicine, Tokyo, Japan
| | - Joseph M Escandón
- Division of Plastic and Reconstructive Surgery, Strong Memorial Hospital, University of Rochester Medical Centre, New York, NY, USA
| | - Arbab Mohammad
- Aarupadai Veedu Medical College and Hospital, Puducherry, India
| | - Takumi Yamamoto
- Department of Plastic and Reconstructive Surgery, National Centre for Global Health and Medicine, Tokyo, Japan.
| |
Collapse
|
6
|
Xiao M, Malmi MA, Schocken DD, Zgibor JC, Alman AC. Longitudinal blood glucose level and increased silent myocardial infarction: a pooled analysis of four cohort studies. Cardiovasc Diabetol 2024; 23:130. [PMID: 38637769 PMCID: PMC11027351 DOI: 10.1186/s12933-024-02212-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 03/25/2024] [Indexed: 04/20/2024] Open
Abstract
BACKGROUND Fasting glucose (FG) demonstrates dynamic fluctuations over time and is associated with cardiovascular outcomes, yet current research is limited by small sample sizes and relies solely on baseline glycemic levels. Our research aims to investigate the longitudinal association between FG and silent myocardial infarction (SMI) and also delves into the nuanced aspect of dose response in a large pooled dataset of four cohort studies. METHODS We analyzed data from 24,732 individuals from four prospective cohort studies who were free of myocardial infarction history at baseline. We calculated average FG and intra-individual FG variability (coefficient of variation), while SMI cases were identified using 12-lead ECG exams with the Minnesota codes and medical history. FG was measured for each subject during the study's follow-up period. We applied a Cox regression model with time-dependent variables to assess the association between FG and SMI with adjustment for age, gender, race, Study, smoking, longitudinal BMI, low-density lipoprotein level, blood pressure, and serum creatinine. RESULTS The average mean age of the study population was 60.5 (sd: 10.3) years with median fasting glucose of 97.3 mg/dL at baseline. During an average of 9 years of follow-up, 357 SMI events were observed (incidence rate, 1.3 per 1000 person-years). The association between FG and SMI was linear and each 25 mg/dL increment in FG was associated with a 15% increase in the risk of SMI. This association remained significant after adjusting for the use of lipid-lowering medication, antihypertensive medication, antidiabetic medication, and insulin treatment (HR 1.08, 95% CI 1.01-1.16). Higher average FG (HR per 25 mg/dL increase: 1.17, 95% CI 1.08-1.26) and variability of FG (HR per 1 sd increase: 1.23, 95% CI 1.12-1.34) over visits were also correlated with increased SMI risk. CONCLUSIONS Higher longitudinal FG and larger intra-individual variability in FG over time were associated in a dose-response manner with a higher SMI risk. These findings support the significance of routine cardiac screening for subjects with elevated FG, with and without diabetes.
Collapse
Affiliation(s)
- Mianli Xiao
- College of Public Health, University of South Florida, Tampa, FL, USA
| | - Markku A Malmi
- College of Public Health, University of South Florida, Tampa, FL, USA
| | | | - Janice C Zgibor
- College of Public Health, University of South Florida, Tampa, FL, USA
| | - Amy C Alman
- College of Public Health, University of South Florida, Tampa, FL, USA.
| |
Collapse
|
7
|
Miceli G, Basso MG, Pintus C, Pennacchio AR, Cocciola E, Cuffaro M, Profita M, Rizzo G, Tuttolomondo A. Molecular Pathways of Vulnerable Carotid Plaques at Risk of Ischemic Stroke: A Narrative Review. Int J Mol Sci 2024; 25:4351. [PMID: 38673936 PMCID: PMC11050267 DOI: 10.3390/ijms25084351] [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: 02/26/2024] [Revised: 04/05/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
The concept of vulnerable carotid plaques is pivotal in understanding the pathophysiology of ischemic stroke secondary to large-artery atherosclerosis. In macroscopic evaluation, vulnerable plaques are characterized by one or more of the following features: microcalcification; neovascularization; lipid-rich necrotic cores (LRNCs); intraplaque hemorrhage (IPH); thin fibrous caps; plaque surface ulceration; huge dimensions, suggesting stenosis; and plaque rupture. Recognizing these macroscopic characteristics is crucial for estimating the risk of cerebrovascular events, also in the case of non-significant (less than 50%) stenosis. Inflammatory biomarkers, such as cytokines and adhesion molecules, lipid-related markers like oxidized low-density lipoprotein (LDL), and proteolytic enzymes capable of degrading extracellular matrix components are among the key molecules that are scrutinized for their associative roles in plaque instability. Through their quantification and evaluation, these biomarkers reveal intricate molecular cross-talk governing plaque inflammation, rupture potential, and thrombogenicity. The current evidence demonstrates that plaque vulnerability phenotypes are multiple and heterogeneous and are associated with many highly complex molecular pathways that determine the activation of an immune-mediated cascade that culminates in thromboinflammation. This narrative review provides a comprehensive analysis of the current knowledge on molecular biomarkers expressed by symptomatic carotid plaques. It explores the association of these biomarkers with the structural and compositional attributes that characterize vulnerable plaques.
Collapse
Affiliation(s)
- Giuseppe Miceli
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (G.M.); (M.G.B.); (C.P.); (A.R.P.); (E.C.); (M.C.); (M.P.); (G.R.)
- Internal Medicine and Stroke Care Ward, University Hospital, Policlinico “P. Giaccone”, 90127 Palermo, Italy
| | - Maria Grazia Basso
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (G.M.); (M.G.B.); (C.P.); (A.R.P.); (E.C.); (M.C.); (M.P.); (G.R.)
- Internal Medicine and Stroke Care Ward, University Hospital, Policlinico “P. Giaccone”, 90127 Palermo, Italy
| | - Chiara Pintus
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (G.M.); (M.G.B.); (C.P.); (A.R.P.); (E.C.); (M.C.); (M.P.); (G.R.)
- Internal Medicine and Stroke Care Ward, University Hospital, Policlinico “P. Giaccone”, 90127 Palermo, Italy
| | - Andrea Roberta Pennacchio
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (G.M.); (M.G.B.); (C.P.); (A.R.P.); (E.C.); (M.C.); (M.P.); (G.R.)
- Internal Medicine and Stroke Care Ward, University Hospital, Policlinico “P. Giaccone”, 90127 Palermo, Italy
| | - Elena Cocciola
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (G.M.); (M.G.B.); (C.P.); (A.R.P.); (E.C.); (M.C.); (M.P.); (G.R.)
- Internal Medicine and Stroke Care Ward, University Hospital, Policlinico “P. Giaccone”, 90127 Palermo, Italy
| | - Mariagiovanna Cuffaro
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (G.M.); (M.G.B.); (C.P.); (A.R.P.); (E.C.); (M.C.); (M.P.); (G.R.)
- Internal Medicine and Stroke Care Ward, University Hospital, Policlinico “P. Giaccone”, 90127 Palermo, Italy
| | - Martina Profita
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (G.M.); (M.G.B.); (C.P.); (A.R.P.); (E.C.); (M.C.); (M.P.); (G.R.)
- Internal Medicine and Stroke Care Ward, University Hospital, Policlinico “P. Giaccone”, 90127 Palermo, Italy
| | - Giuliana Rizzo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (G.M.); (M.G.B.); (C.P.); (A.R.P.); (E.C.); (M.C.); (M.P.); (G.R.)
- Internal Medicine and Stroke Care Ward, University Hospital, Policlinico “P. Giaccone”, 90127 Palermo, Italy
| | - Antonino Tuttolomondo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (G.M.); (M.G.B.); (C.P.); (A.R.P.); (E.C.); (M.C.); (M.P.); (G.R.)
- Internal Medicine and Stroke Care Ward, University Hospital, Policlinico “P. Giaccone”, 90127 Palermo, Italy
| |
Collapse
|
8
|
Qi Z, Yan Z, Zhu K, Wang Y, Fan Y, Li T, Zhang J. Novel treatment from a botanical formulation Si-Miao-Yong-an decoction inhibits vasa vasorum angiogenesis and stabilizes atherosclerosis plaques via the Wnt1/β-catenin signalling pathway. PHARMACEUTICAL BIOLOGY 2023; 61:1364-1373. [PMID: 37651108 PMCID: PMC10472848 DOI: 10.1080/13880209.2023.2249061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 07/03/2023] [Accepted: 08/12/2023] [Indexed: 09/01/2023]
Abstract
CONTEXT Si-Miao-Yong-An (SMYA) has been widely used for the clinical treatment of atherosclerosis (AS). Yet, its complete mechanism of action is not fully understood. OBJECTIVE To investigate the mechanism by which SMYA stabilizes AS plaques from the perspective of inhibiting vasa vasorum (VV) angiogenesis. MATERIALS AND METHODS We used male ApoE-/- mice to establish an AS model. The mice were divided into model, SMYA (11.7 mg/kg/d), and simvastatin (SVTT) (2.6 mg/kg/d) groups. Mice were given SMYA or SVTT by daily gavage for 8 weeks. HE staining, immunofluorescence double-labelling staining, and immunohistochemical staining were used to observe the pathological changes in the plaques. Finally, the protein and mRNA expression levels of the Wnt1/β-catenin signalling pathway were detected by Western blot and qRT-PCR, respectively. RESULTS SMYA significantly attenuated cholesterol crystallization, and lipid accumulation in AS plaques, resulting in smaller plaque size (0.25 mm2 vs. 0.46 mm2), and lowering ratio of plaque to lumen area (20.04% vs. 38.33%) and VV density (50.64/mm2 vs. 98.02/mm2). Meanwhile, SMYA suppressed both the positive area percentage of Wnt1 (2.53 vs. 3.56), β-catenin (3.33 vs. 5.65) and Cyclin D1 (2.10 vs. 3.27) proteins in the aortic root plaques, and mRNA expression of Wnt1 (1.38 vs. 2.09), β-catenin (2.05 vs. 3.25) and Cyclin D1 (1.39 vs. 2.57). DISCUSSION AND CONCLUSIONS SMYA has a protective effect against AS, which may be related to its anti-VV angiogenesis in plaques, suggesting that SMYA has the potential as a novel botanical formulation in the treatment of AS.
Collapse
Affiliation(s)
- Zhongwen Qi
- Postdoctoral Research Station of China Academy of Chinese Medical Sciences, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, P.R. China
- Institute of Gerontology, China Academy of Chinese Medical Sciences, Beijing, P.R. China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Zhipeng Yan
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Ke Zhu
- Zhengzhou Hospital of Traditional Chinese Medicine, Zhengzhou, P.R. China
| | - Yueyao Wang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Yajie Fan
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Tingting Li
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Junping Zhang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| |
Collapse
|
9
|
Yu S, Huo R, Qiao H, Ning Z, Xu H, Yang D, Shen R, Xu N, Han H, Chen S, Liu Y, Zhao X. Carotid artery perivascular adipose tissue on magnetic resonance imaging: a potential indicator for carotid vulnerable atherosclerotic plaque. Quant Imaging Med Surg 2023; 13:7695-7705. [PMID: 38106263 PMCID: PMC10722049 DOI: 10.21037/qims-23-280] [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: 03/07/2023] [Accepted: 08/24/2023] [Indexed: 12/19/2023]
Abstract
Background Magnetic resonance imaging (MRI) has the potential in assessing the inflammation of perivascular adipose tissue (PVAT) due to its excellent soft tissue contrast. However, evidence is lacking for the association between carotid PVAT measured by MRI and carotid vulnerable atherosclerotic plaques. This study aimed to investigate the association between signal intensity of PVAT and vulnerable plaques in carotid arteries using multi-contrast magnetic resonance (MR) vessel wall imaging. Methods In this cross-sectional study, a total of 104 patients (mean age, 64.9±7.0 years; 86 men) with unilateral moderate-to-severe atherosclerotic stenosis referred to carotid endarterectomy (CEA) were recruited from April 2018 to December 2020 at Department of Neurosurgery of Peking University Third Hospital. All patients underwent multi-contrast MR vessel wall imaging including time-of-flight (ToF) MR angiography, black-blood T1-weighted (T1w) and T2-weighted (T2w) and simultaneous non-contrast angiography and intraplaque hemorrhage (IPH) imaging sequences. Patients with contraindications to endarterectomy or MRI examinations were excluded. The signal-to-noise ratio (SNR) and contrast-to-noise ratio (CNR) of PVAT were measured on ToF images and vulnerable plaque characteristics including IPH, large lipid-rich necrotic core (LRNC), and fibrous cap rupture (FCR) were identified. The SNR and CNR of PVAT were compared between slices with and without vulnerable plaque features using Mann-Whitney U test and their associations were analyzed using the generalized linear mixed model (GLMM). Results Carotid artery slices with IPH (30.93±14.56 vs. 27.34±10.02; P<0.001), FCR (30.35±13.82 vs. 27.53±10.37; P=0.006), and vulnerable plaque (29.15±12.52 vs. 27.32±10.05; P=0.016) had significantly higher value of SNR of PVAT compared to those without. After adjusting for clinical confounders, the SNR of PVAT was significantly associated with presence of IPH [odds ratio (OR) =0.627, 95% confidence interval (CI): 0.465-0.847, Puncorr=0.002, PFDR=0.016] and vulnerable plaque (OR =0.762, 95% CI: 0.629-0.924, Puncorr=0.006, PFDR=0.020). However, no significant association was found between the CNR of PVAT and presence of vulnerable plaque features (all P>0.05). Conclusions The SNR of carotid artery PVAT measured by ToF MR angiography is independently associated with vulnerable atherosclerotic plaque features, suggesting that the signal intensity of PVAT might be an effective indicator for vulnerable plaque.
Collapse
Affiliation(s)
- Shuwan Yu
- Center for Biomedical Imaging Research, Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, China
| | - Ran Huo
- Department of Radiology, Peking University Third Hospital, Beijing, China
| | - Huiyu Qiao
- Center for Biomedical Imaging Research, Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, China
- Tsinghua University-Peking University Joint Center for Life Sciences, Beijing, China
| | - Zihan Ning
- Center for Biomedical Imaging Research, Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, China
| | - Huimin Xu
- Department of Radiology, Peking University Third Hospital, Beijing, China
| | - Dandan Yang
- Department of Radiology, Beijing Geriatric Hospital, Beijing, China
| | - Rui Shen
- Center for Biomedical Imaging Research, Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, China
| | - Ning Xu
- Center for Biomedical Imaging Research, Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, China
| | - Hualu Han
- Center for Biomedical Imaging Research, Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, China
| | - Shuo Chen
- Center for Biomedical Imaging Research, Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, China
- Tsinghua University-Peking University Joint Center for Life Sciences, Beijing, China
| | - Ying Liu
- Department of Radiology, Peking University Third Hospital, Beijing, China
| | - Xihai Zhao
- Center for Biomedical Imaging Research, Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, China
| |
Collapse
|
10
|
Jamgotchian L, Devel L, Thai R, Poupel L, Huby T, Gautier E, Le Goff W, Lesnik P, Gravel E, Doris E. Targeted delivery of LXR-agonists to atherosclerotic lesions mediated by polydiacetylene micelles. NANOSCALE 2023; 15:18864-18870. [PMID: 37966726 DOI: 10.1039/d3nr04778d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2023]
Abstract
We report the development of compact and stabilized micelles incorporating a synthetic LXR agonist prodrug for the passive targeting of atherosclerotic lesions and therapeutic intervention. In vivo studies showed that the nanohybrid micelles exhibited favorable pharmacokinetics/biodistribution and were able to upregulate, to some extent, LXR target genes with no alteration of lipid metabolism.
Collapse
Affiliation(s)
- Lucie Jamgotchian
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SCBM, 91191 Gif-sur-Yvette, France.
| | - Laurent Devel
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SIMOS, 91191 Gif-sur-Yvette, France.
| | - Robert Thai
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SIMOS, 91191 Gif-sur-Yvette, France.
| | - Lucie Poupel
- Inovarion, 251 rue saint Jacques, 75005 Paris, France
| | - Thierry Huby
- Sorbonne Université, INSERM UMRS-1166, Institute of Cardiometabolism and Nutrition (ICAN), 75013 Paris, France.
| | - Emmanuel Gautier
- Sorbonne Université, INSERM UMRS-1166, Institute of Cardiometabolism and Nutrition (ICAN), 75013 Paris, France.
| | - Wilfried Le Goff
- Sorbonne Université, INSERM UMRS-1166, Institute of Cardiometabolism and Nutrition (ICAN), 75013 Paris, France.
| | - Philippe Lesnik
- Sorbonne Université, INSERM UMRS-1166, Institute of Cardiometabolism and Nutrition (ICAN), 75013 Paris, France.
| | - Edmond Gravel
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SCBM, 91191 Gif-sur-Yvette, France.
| | - Eric Doris
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SCBM, 91191 Gif-sur-Yvette, France.
| |
Collapse
|
11
|
Yan A, Gotlieb AI. The microenvironment of the atheroma expresses phenotypes of plaque instability. Cardiovasc Pathol 2023; 67:107572. [PMID: 37595697 DOI: 10.1016/j.carpath.2023.107572] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/06/2023] [Accepted: 08/14/2023] [Indexed: 08/20/2023] Open
Abstract
Data from histopathology studies of human atherosclerotic tissue specimens and from vascular imaging studies support the concept that the local arterial microenvironment of a stable atheroma promotes destabilizing conditions that result in the transition to an unstable atheroma. Destabilization is characterized by several different plaque phenotypes that cause major clinical events such as acute coronary syndrome and cerebrovascular strokes. There are several rupture-associated phenotypes causing thrombotic vascular occlusion including simple fibrous cap rupture of an atheroma, fibrous cap rupture at site of previous rupture-and-repair of an atheroma, and nodular calcification with rupture. Endothelial erosion without rupture has more recently been shown to be a common phenotype to promote thrombosis as well. Microenvironment features that are linked to these phenotypes of plaque instability are neovascularization arising from the vasa vasorum network leading to necrotic core expansion, intraplaque hemorrhage, and cap rupture; activation of adventitial and perivascular adipose tissue cells leading to secretion of cytokines, growth factors, adipokines in the outer artery wall that destabilize plaque structure; and vascular smooth muscle cell phenotypic switching through transdifferentiation and stem/progenitor cell activation resulting in the promotion of inflammation, calcification, and secretion of extracellular matrix, altering fibrous cap structure, and necrotic core growth. As the technology evolves, studies using noninvasive vascular imaging will be able to investigate the transition of stable to unstable atheromas in real time. A limitation in the field, however, is that reliable and predictable experimental models of spontaneous plaque rupture and/or erosion are not currently available to study the cell and molecular mechanisms that regulate the conversion of the stable atheroma to an unstable plaque.
Collapse
Affiliation(s)
- Angela Yan
- 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
|
12
|
Abela GS, Katkoori VR, Pathak DR, Bumpers HL, Leja M, Abideen ZU, Boumegouas M, Perry D, Al-Janadi A, Richard JE, Barnaba C, Meza IGM. Cholesterol crystals induce mechanical trauma, inflammation, and neo-vascularization in solid cancers as in atherosclerosis. AMERICAN HEART JOURNAL PLUS : CARDIOLOGY RESEARCH AND PRACTICE 2023; 35:100317. [PMID: 37981958 PMCID: PMC10655498 DOI: 10.1016/j.ahjo.2023.100317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/21/2023]
Abstract
Background and aims Cancer and atherosclerosis share common risk factors and inflammatory pathways that promote their proliferation via vascular endothelial growth factor (VEGF). Because CCs cause mechanical injury and inflammation in atherosclerosis, we investigated their presence in solid cancers and their activation of IL-1β, VEGF, CD44, and Ubiquityl-Histone H2B (Ub-H2B), that promote cancer growth. Methods Tumor specimens from eleven different types of human cancers and atherosclerotic plaques were assessed for CCs, free cholesterol content and IL1-β by microscopy, immunohistochemistry, and biochemical analysis. Breast and colon cancer cell lines were cultured with and without CCs to select for expression of VEGF, CD44, and Ub-H2B. Western blot and immunofluorescence were performed on cells to assess the effect of CCs on signaling pathways. Results Cancers displayed higher CC content (+2.29 ± 0.74 vs +1.46 ± 0.84, p < 0.0001), distribution (5.06 ± 3.13 vs 2.86 ± 2.18, p < 0.001) and free cholesterol (3.63 ± 4.02 vs 1.52 ± 0.56 μg/mg, p < 0.01) than cancer free marginal tissues and similarly for atherosclerotic plaques and margins (+2.31 ± 0.51 vs +1.44 ± 0.79, p < 0.02; 14.0 ± 5.74 vs 8.14 ± 5.52, p < 0.03; 0.19 ± 0.14 vs 0.09 ± 0.04 μg/mg, p < 0.02) respectively. Cancers displayed significantly increased expression of IL1-β compared to marginal tissues. CCs treated cancer cells had increased expression of VEGF, CD44, and Ub-H2B compared to control. By microscopy, CCs were found perforating cancer tumors similar to plaque rupture. Conclusions These findings suggest that CCs can induce trauma and activate cytokines that enhance cancer growth as in atherosclerosis.
Collapse
Affiliation(s)
- George S. Abela
- Department of Medicine, Division of Cardiology, Michigan State University, East Lansing, MI, USA
- Department of Physiology, Division of Pathology, Michigan State University, East Lansing, MI, USA
| | - Venkat R. Katkoori
- Department of Physiology, Division of Pathology, Michigan State University, East Lansing, MI, USA
| | - Dorothy R. Pathak
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA
| | - Harvey L. Bumpers
- Department of Surgery, Michigan State University, East Lansing, MI, USA
| | - Monika Leja
- Department of Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Zain ul Abideen
- Department of Medicine, Division of Cardiology, Michigan State University, East Lansing, MI, USA
| | - Manel Boumegouas
- Department of Medicine, Division of Cardiology, Michigan State University, East Lansing, MI, USA
| | - Daniel Perry
- Department of Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Anas Al-Janadi
- Department of Cancer Care Services, Corewell Health, Grand Rapids, MI, USA
| | | | - Carlo Barnaba
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
| | - Ilce G. Medina Meza
- Biosystems and Agricultural Engineering, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
13
|
Kirkgöz K. C-Reactive Protein in Atherosclerosis-More than a Biomarker, but not Just a Culprit. Rev Cardiovasc Med 2023; 24:297. [PMID: 39077585 PMCID: PMC11262456 DOI: 10.31083/j.rcm2410297] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 08/11/2023] [Accepted: 08/21/2023] [Indexed: 07/31/2024] Open
Abstract
C-reactive protein (CRP) is a pentraxin that is mainly synthesized in the liver in response to inflammatory cytokines. It exists in two functionally and structurally distinct isoforms. The first is a highly pro-inflammatory and mostly tissue-bound monomeric isoform (mCRP). The second is circulating pentameric CRP (pCRP), which also serves as a substrate for the formation of mCRP. CRP is elevated during inflammatory conditions and is associated with a higher risk of cardiovascular disease. The aim of this review is to examine the current state of knowledge regarding the role of these two distinct CRP isoforms on atherogenesis. This should allow further evaluation of CRP as a potential therapeutic target for atherosclerosis. While it seems clear that CRP should be used as a therapeutic target for atherosclerosis and cardiovascular disease, questions remain about how this can be achieved. Current data suggests that CRP is more than just a biomarker of atherosclerosis and cardiovascular disease. Indeed, recent evidence shows that mCRP in particular is strongly atherogenic, whereas pCRP may be partially protective against atherogenesis. Thus, further investigation is needed to determine how the two CRP isoforms contribute to atherogenesis and the development of cardiovascular disease.
Collapse
Affiliation(s)
- Kürsat Kirkgöz
- University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| |
Collapse
|
14
|
Badimon L, Arderiu G. Atherosclerotic Plaque VASA Vasorum in Diabetic Macroangiopathy: WHEN IS Important, but also HOW IS Needed. Thromb Haemost 2023; 123:999-1002. [PMID: 37353212 DOI: 10.1055/a-2116-7261] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/25/2023]
Affiliation(s)
- Lina Badimon
- Cardiovascular Program-ICCC, IR-Hospital de la Santa Creu i Sant Pau, IIBSantPau, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV CB16/11/00226), Madrid, Spain
| | - Gemma Arderiu
- Cardiovascular Program-ICCC, IR-Hospital de la Santa Creu i Sant Pau, IIBSantPau, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV CB16/11/00226), Madrid, Spain
| |
Collapse
|
15
|
Belli M, Bellia A, Sergi D, Barone L, Lauro D, Barillà F. Glucose variability: a new risk factor for cardiovascular disease. Acta Diabetol 2023; 60:1291-1299. [PMID: 37341768 PMCID: PMC10442283 DOI: 10.1007/s00592-023-02097-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 04/11/2023] [Indexed: 06/22/2023]
Abstract
AIMS AND DATA SYNTHESIS Glucose variability (GV) is increasingly considered an additional index of glycemic control. Growing evidence indicates that GV is associated with diabetic vascular complications, thus being a relevant point to address in diabetes management. GV can be measured using various parameters, but to date, a gold standard has not been identified. This underscores the need for further studies in this field also to identify the optimal treatment. CONCLUSIONS We reviewed the definition of GV, the pathogenetic mechanisms of atherosclerosis, and its relationship with diabetic complications.
Collapse
Affiliation(s)
- Martina Belli
- Division of Cardiology, Department of Systems Medicine, Tor Vergata University, 00133, Rome, Italy
- Cardiovascular Imaging Unit, San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Alfonso Bellia
- Department of Systems Medicine, Tor Vergata University, 00133, Rome, Italy
| | - Domenico Sergi
- Division of Cardiology, Department of Systems Medicine, Tor Vergata University, 00133, Rome, Italy
| | - Lucy Barone
- Division of Cardiology, Department of Systems Medicine, Tor Vergata University, 00133, Rome, Italy
| | - Davide Lauro
- Department of Systems Medicine, Tor Vergata University, 00133, Rome, Italy
| | - Francesco Barillà
- Division of Cardiology, Department of Systems Medicine, Tor Vergata University, 00133, Rome, Italy.
| |
Collapse
|
16
|
Markova V, Bogdanov L, Velikanova E, Kanonykina A, Frolov A, Shishkova D, Lazebnaya A, Kutikhin A. Endothelial Cell Markers Are Inferior to Vascular Smooth Muscle Cells Markers in Staining Vasa Vasorum and Are Non-Specific for Distinct Endothelial Cell Lineages in Clinical Samples. Int J Mol Sci 2023; 24:ijms24031959. [PMID: 36768296 PMCID: PMC9916324 DOI: 10.3390/ijms24031959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/15/2023] [Accepted: 01/18/2023] [Indexed: 01/20/2023] Open
Abstract
Current techniques for the detection of vasa vasorum (VV) in vascular pathology include staining for endothelial cell (EC) markers such as CD31 or VE-cadherin. However, this approach does not permit an objective assessment of vascular geometry upon vasospasm and the clinical relevance of endothelial specification markers found in developmental biology studies remains unclear. Here, we performed a combined immunostaining of rat abdominal aorta (rAA) and human saphenous vein (hSV) for various EC or vascular smooth muscle cell (VSMC) markers and found that the latter (e.g., alpha smooth muscle actin (α-SMA) or smooth muscle myosin heavy chain (SM-MHC)) ensure a several-fold higher signal-to-noise ratio irrespective of the primary antibody origin, fluorophore, or VV type (arterioles, venules, or capillaries). Further, α-SMA or SM-MHC staining allowed unbiased evaluation of the VV area under vasospasm. Screening of the molecular markers of endothelial heterogeneity (mechanosensitive transcription factors KLF2 and KLF4, arterial transcription factors HES1, HEY1, and ERG, venous transcription factor NR2F2, and venous/lymphatic markers PROX1, LYVE1, VEGFR3, and NRP2) have not revealed specific markers of any lineage in hSV (although KLF2 and PROX1 were restricted to venous endothelium in rAA), suggesting the need in high-throughput searches for the clinically relevant signatures of arterial, venous, lymphatic, or capillary differentiation.
Collapse
|
17
|
Bax M, Romanov V, Junday K, Giannoulatou E, Martinac B, Kovacic JC, Liu R, Iismaa SE, Graham RM. Arterial dissections: Common features and new perspectives. Front Cardiovasc Med 2022; 9:1055862. [PMID: 36561772 PMCID: PMC9763901 DOI: 10.3389/fcvm.2022.1055862] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 11/16/2022] [Indexed: 12/12/2022] Open
Abstract
Arterial dissections, which involve an abrupt tear in the wall of a major artery resulting in the intramural accumulation of blood, are a family of catastrophic disorders causing major, potentially fatal sequelae. Involving diverse vascular beds, including the aorta or coronary, cervical, pulmonary, and visceral arteries, each type of dissection is devastating in its own way. Traditionally they have been studied in isolation, rather than collectively, owing largely to the distinct clinical consequences of dissections in different anatomical locations - such as stroke, myocardial infarction, and renal failure. Here, we review the shared and unique features of these arteriopathies to provide a better understanding of this family of disorders. Arterial dissections occur commonly in the young to middle-aged, and often in conjunction with hypertension and/or migraine; the latter suggesting they are part of a generalized vasculopathy. Genetic studies as well as cellular and molecular investigations of arterial dissections reveal striking similarities between dissection types, particularly their pathophysiology, which includes the presence or absence of an intimal tear and vasa vasorum dysfunction as a cause of intramural hemorrhage. Pathway perturbations common to all types of dissections include disruption of TGF-β signaling, the extracellular matrix, the cytoskeleton or metabolism, as evidenced by the finding of mutations in critical genes regulating these processes, including LRP1, collagen genes, fibrillin and TGF-β receptors, or their coupled pathways. Perturbances in these connected signaling pathways contribute to phenotype switching in endothelial and vascular smooth muscle cells of the affected artery, in which their physiological quiescent state is lost and replaced by a proliferative activated phenotype. Of interest, dissections in various anatomical locations are associated with distinct sex and age predilections, suggesting involvement of gene and environment interactions in disease pathogenesis. Importantly, these cellular mechanisms are potentially therapeutically targetable. Consideration of arterial dissections as a collective pathology allows insight from the better characterized dissection types, such as that involving the thoracic aorta, to be leveraged to inform the less common forms of dissections, including the potential to apply known therapeutic interventions already clinically available for the former.
Collapse
Affiliation(s)
- Monique Bax
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- UNSW Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
| | - Valentin Romanov
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- UNSW Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
| | - Keerat Junday
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- UNSW Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
| | - Eleni Giannoulatou
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- UNSW Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
| | - Boris Martinac
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- UNSW Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
| | - Jason C. Kovacic
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- UNSW Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
- St. Vincent’s Hospital, Darlinghurst, NSW, Australia
- Icahn School of Medicine at Mount Sinai, Cardiovascular Research Institute, New York, NY, United States
| | - Renjing Liu
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- UNSW Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
| | - Siiri E. Iismaa
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- UNSW Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
| | - Robert M. Graham
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- UNSW Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
- St. Vincent’s Hospital, Darlinghurst, NSW, Australia
| |
Collapse
|
18
|
Sun L, Jiang Q, Xie Y, Wang S, Zhang Z. Optical coherence tomography of the pulmonary arteries in children with congenital heart diseases: A systematic review. Pediatr Investig 2022; 6:264-270. [PMID: 36582270 PMCID: PMC9789933 DOI: 10.1002/ped4.12353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 10/14/2022] [Indexed: 12/05/2022] Open
Abstract
Importance Optical coherence tomography (OCT) is a high-resolution intravascular imaging tool and has shown promise for providing real-time quantitative and qualitative descriptions of pulmonary vascular structures in vivo in adult pulmonary hypertension (PH), while not popular in pediatric patients with congenital heart diseases (CHD). Objective The aim of this review is to summarize all the available evidence on the use of OCT for imaging pulmonary vascular remodeling in pediatric patients. Methods We conducted the systematic literature resources (Cochran Library database, Medline via PubMed, EMBASE, and Web of Knowledge) from January 2010 to December 2021 and the search terms were "PH", "child", "children", "pediatric", "OCT", "CHD", "pulmonary vessels", "pulmonary artery wall". Studies in which OCT was used to image the pulmonary vessels in pediatric patients with CHD were considered for inclusion. Results Five studies met the inclusion criteria. These five papers discussed the study of OCT in the pulmonary vasculature of different types of CHD, including common simple CHD, complex cyanotic CHD, and Williams-Beuren syndrome. In biventricular anatomy, pulmonary vascular remodeling was primarily reflected by pulmonary intima thickening from two-dimensional OCT. In single-ventricle anatomy, due to the state of hypoxia, the morphology of pulmonary vessels was indirectly reflected by the number and shape of nourishing vessels from three-dimensional OCT. Interpretation OCT may be an adequate imaging procedure for the demonstration of pulmonary vascular structures and provide additional information in pediatric patients.
Collapse
Affiliation(s)
- Ling Sun
- Department of Pediatric CardiologyGuangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangdong Cardiovascular InstituteGuangdong Provincial Key Laboratory of South China Structural Heart DiseaseGuangzhouGuangdongChina
| | - Qiuping Jiang
- Department of Pediatric CardiologyGuangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangdong Cardiovascular InstituteGuangdong Provincial Key Laboratory of South China Structural Heart DiseaseGuangzhouGuangdongChina
| | - Yumei Xie
- Department of Pediatric CardiologyGuangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangdong Cardiovascular InstituteGuangdong Provincial Key Laboratory of South China Structural Heart DiseaseGuangzhouGuangdongChina
| | - Shushui Wang
- Department of Pediatric CardiologyGuangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangdong Cardiovascular InstituteGuangdong Provincial Key Laboratory of South China Structural Heart DiseaseGuangzhouGuangdongChina
| | - Zhiwei Zhang
- Department of Pediatric CardiologyGuangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangdong Cardiovascular InstituteGuangdong Provincial Key Laboratory of South China Structural Heart DiseaseGuangzhouGuangdongChina
| |
Collapse
|
19
|
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: 10] [Impact Index Per Article: 5.0] [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
|
20
|
Kurozumi A, Hara H, Nagai R, Hiroi Y. Acute myocardial infarction immediately after second vaccination for coronavirus disease 2019. Clin Case Rep 2022; 10:e6431. [PMID: 36245459 PMCID: PMC9552979 DOI: 10.1002/ccr3.6431] [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: 02/18/2022] [Revised: 04/27/2022] [Accepted: 09/26/2022] [Indexed: 11/11/2022] Open
Abstract
We present a serious and rare case of acute myocardial infarction soon after the administration of second vaccination for coronavirus disease 2019. Patient's culprit lesion in the right coronary artery was identified and appropriately treated using intravascular imaging. Postvaccination monitoring of patients who are at high risk of cardiovascular diseases is critical. Rare but severe cases of acute myocardial infarction following vaccination for coronavirus disease 2019 have been reported. Physicians should consider this rare side effect as a possible differential diagnosis and appropriately manage such patients.
Collapse
Affiliation(s)
- Atsumasa Kurozumi
- Department of CardiologyNational Center for Global Health and MedicineShinjuku‐city, TokyoJapan
| | - Hisao Hara
- Department of CardiologyNational Center for Global Health and MedicineShinjuku‐city, TokyoJapan
| | - Ran Nagai
- Department of CardiologyNational Center for Global Health and MedicineShinjuku‐city, TokyoJapan
| | - Yukio Hiroi
- Department of CardiologyNational Center for Global Health and MedicineShinjuku‐city, TokyoJapan
| |
Collapse
|
21
|
Chen Y, Qin W, Li L, Wu P, Wei D. Mitophagy: Critical Role in Atherosclerosis Progression. DNA Cell Biol 2022; 41:851-860. [PMID: 36036955 DOI: 10.1089/dna.2022.0249] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Autophagy maintains intracellular homeostasis in the cardiovascular system, including in cardiomyocytes, endothelial cells (ECs), and arterial smooth muscle cells. Mitophagy, a selective autophagy that specifically removes damaged and dysfunctional mitochondria, is particularly important for cardiovascular homeostasis. Dysfunctional mitophagy contributes to cardiovascular disease, particularly atherosclerosis (AS). This review focuses on the advances of regulator mechanisms of mitophagy and its potential roles in AS. The findings are beneficial to understanding the pathological processes of atherosclerotic lesions and provide new ideas for the prevention and clinical treatment of AS.
Collapse
Affiliation(s)
- Yanmei Chen
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, China
| | - Wenhua Qin
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, China
| | - Lu Li
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, China
| | - Peng Wu
- Yueyang Maternal-Child Medicine Health Hospital Hunan, Province Innovative Training Base for Medical Postgraduates, University of China South China and Yueyang Women & Children's Medical Center, Yueyang, China.,Hengyang Maternal and Child Health Hospital, Hengyang, China
| | - Dangheng Wei
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, China.,Yueyang Maternal-Child Medicine Health Hospital Hunan, Province Innovative Training Base for Medical Postgraduates, University of China South China and Yueyang Women & Children's Medical Center, Yueyang, China
| |
Collapse
|
22
|
Psychological stress-induced increase in the cardio-ankle vascular index (CAVI) may be a predictor of cardiovascular events. Hypertens Res 2022; 45:1672-1674. [PMID: 35974174 DOI: 10.1038/s41440-022-00996-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 07/09/2022] [Accepted: 07/15/2022] [Indexed: 11/09/2022]
|
23
|
Ahmadloo S, Ling KH, Fazli A, Larijani G, Ghodsian N, Mohammadi S, Amini N, Hosseinpour Sarmadi V, Ismail P. Signature pattern of gene expression and signaling pathway in premature diabetic patients uncover their correlation to early age coronary heart disease. Diabetol Metab Syndr 2022; 14:107. [PMID: 35906673 PMCID: PMC9336005 DOI: 10.1186/s13098-022-00878-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 07/21/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Coronary Heart Disease (CHD) is the leading cause of death in industrialized countries. There is currently no direct relation between CHD and type 2 diabetes mellitus (T2D), one of the major modifiable risk factors for CHD. This study was carried out for genes expression profiling of T2D associated genes to identify related biological processes/es and modulated signaling pathway/s of male subjects with CHD. METHOD the subjects were divided into four groups based on their disease, including control, type 2 diabetes mellitus (T2D), CHD, and CHD + T2D groups. The RNA was extracted from their blood, and RT2 Profiler™ PCR Array was utilized to determine gene profiling between groups. Finally, the PCR Array results were validated by using Q-RT-PCR in a more extensive and independent population. RESULT PCR Array results revealed that the T2D and T2D + CHD groups shared 11 genes significantly up-regulated in both groups. Further analysis showed that the mRNA levels of AKT2, IL12B, IL6, IRS1, IRS2, MAPK14, and NFKB1 increased. Consequently, the mRNA levels of AQP2, FOXP3, G6PD, and PIK3R1 declined in the T2D + CHD group compared to the T2D group. Furthermore, in silico analysis indicated 36 Gene Ontology terms and 59 signaling pathways were significantly enriched in both groups, which may be a culprit in susceptibility of diabetic patients to CHD development. CONCLUSION Finally, the results revealed six genes as a hub gene in altering various biological processes and signaling pathways. The expression trend of these identified genes might be used as potential markers and diagnostic tools for the early identification of the vulnerability of T2D patients to develop premature CHD.
Collapse
Affiliation(s)
- Salma Ahmadloo
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
- Vaccination Department, Pasteur Institute of Iran, Tehran, Iran
| | - King-Hwa Ling
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
- Genetics and Regenerative Medicine Research Center, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Ahmad Fazli
- Department of Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Ghazaleh Larijani
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Nooshin Ghodsian
- Department of Biomedical Engineering, University of British Columbia, Vancouver, Canada
| | - Sanaz Mohammadi
- Faculty of Biological Science and Technology, Shahid Beheshti University, Tehran, Iran
| | - Naser Amini
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Institutes of Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Vahid Hosseinpour Sarmadi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
- Institutes of Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Patimah Ismail
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia.
| |
Collapse
|
24
|
Hillock-Watling C, Gotlieb AI. The pathobiology of perivascular adipose tissue (PVAT), the fourth layer of the blood vessel wall. Cardiovasc Pathol 2022; 61:107459. [PMID: 35907442 DOI: 10.1016/j.carpath.2022.107459] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 06/24/2022] [Accepted: 07/21/2022] [Indexed: 12/21/2022] Open
Abstract
The perivascular adipose tissue (PVAT) is an adipose tissue depot which surrounds most human blood vessels. It is metabolically active and has both a protective and a pathogenic role in vascular biology and pathobiology. It regulates vascular homeostasis and promotes vascular dysfunction. The purpose of this review is to consider the origin, structure, function, and dysfunction of this unique adipose depot consisting of white (WAT), brown (BAT) and beige adipose tissue, to support the concept that PVAT may be considered the fourth layer of the normal arterial wall (tunica adiposa), in which dysfunction creates a microenvironment that regulates, in part, the initiation and growth of the fibro-inflammatory lipid atherosclerotic plaque. Experimental in-vivo and in-vitro studies and human investigations show that the adipocytes, extracellular matrix, nerve fibers and vasa vasorum found in PVAT form a functional adipose tissue unit adjacent to, but not anatomically separated from, the adventitia. PVAT maintains and regulates the structure and function of the normal arterial wall through autocrine and paracrine mechanisms, that include modulation of medial smooth muscle cell contractility and secretion of anti-inflammatory molecules. PVAT shows regional phenotypic heterogeneity which may be important in its effect on the wall of specific sections of the aorta and its muscular branches during perturbations and various injuries including obesity and diabetes. In atherosclerosis, a pan-vascular microenvironment is created that functionally links the intima-medial atherosclerotic plaque to the adventitia and PVAT beneath the plaque, highlighting the local impact of PVAT on atherogenesis. PVAT adipocytes have inflammatory effects which in response to injury show activation and phenotypic changes, some of which are considered to have direct and indirect effects on the intima and media during the initiation, growth, and development of complicated atherosclerotic plaques. Thus, it is important to maintain the integrity of the full vascular microenvironment so that design of experimental and human studies include investigation of PVAT. The era of discarding PVAT tissue in both experimental and human research and clinical vascular studies should end.
Collapse
Affiliation(s)
- Cassie Hillock-Watling
- 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
|
25
|
The Cerebral Arterial Wall in the Development and Growth of Intracranial Aneurysms. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12125964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
A considerable number of people harbor intracranial aneurysms (IA), which is a focal or segmental disease of the arterial wall. The pathophysiologic mechanisms of IAs formation, growth, and rupture are complex. The mechanism also differs with respect to the type of aneurysm. In broad aspects, aneurysms may be considered a disease of the vessel wall. In addition to the classic risk factors and the genetic/environmental conditions, altered structural and pathologic events along with the interaction of the surrounding environment and luminal flow dynamics contribute to the aneurysm’s development and growth. In this review, we have tried to simplify the complex interaction of a multitude of events in relation to vessel wall in the formation and growth of IAs.
Collapse
|
26
|
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
|
27
|
Cui L, Xing Y, Wang L, Liu K, Chen H, Li C, Chen Y. Carotid Intraplaque Neovascularization and Future Vascular Events in Patients With Asymptomatic Carotid Stenosis. Front Pharmacol 2022; 13:804810. [PMID: 35273496 PMCID: PMC8902365 DOI: 10.3389/fphar.2022.804810] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 01/24/2022] [Indexed: 11/27/2022] Open
Abstract
Objective: Intraplaque neovascularization is a marker of plaque vulnerability and is used to predict the risk of future vascular events in patients with symptomatic carotid stenosis; however, its association with asymptomatic carotid stenosis has not been prospectively evaluated. Therefore, this study aimed to explore the association between intraplaque neovascularization assessed using contrast-enhanced ultrasound and the occurrence of future ischemic events in asymptomatic patients diagnosed with carotid stenosis. Methods: We recruited patients with asymptomatic carotid stenosis from our center. Contrast-enhanced ultrasound was performed at baseline. The outcomes were ischemic stroke and cardiovascular events. We plotted Kaplan-Meier survival curves and performed a log-rank test to compare endpoint event probability in patients with and without grade 2 intraplaque neovascularization. Univariate and multivariate Cox proportional hazards models were used to assess predictors of future vascular events. Results: The data of 50 participants were included in the analysis (median follow-up, 43.7 months). Endpoint events occurred in 12 participants (24%). The Kaplan-Meier survival curves showed that patients with grade 2 intraplaque neovascularization had a higher probability of future vascular events than those with grades 0 and 1 (p < .05). Grade 2 intraplaque neovascularization (hazard ratio: 4.530, 95% confidence interval, 1.337–15.343, p < .05) was an independent predictor of future vascular events in patients with asymptomatic carotid stenosis. Conclusion: Grade 2 intraplaque neovascularization assessed using contrast-enhanced ultrasound independently predicted future ischemic events in patients with asymptomatic carotid stenosis, and contrast-enhanced ultrasound may be an effective screening method to identify high-risk subgroups of patients with asymptomatic carotid stenosis.
Collapse
Affiliation(s)
- Liuping Cui
- Department of Neurology, the First Hospital of Jilin University, Changchun, China
| | - Yingqi Xing
- Department of Vascular Ultrasonography, Xuanwu Hospital, Capital Medical University, Beijing, China.,Beijing Diagnostic Center of Vascular Ultrasound, Beijing, China.,Center of Vascular Ultrasonography, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Lijuan Wang
- Department of Neurology, the First Hospital of Jilin University, Changchun, China
| | - Kangding Liu
- Department of Neurology, the First Hospital of Jilin University, Changchun, China
| | - Hongxiu Chen
- Department of Vascular Ultrasonography, Xuanwu Hospital, Capital Medical University, Beijing, China.,Beijing Diagnostic Center of Vascular Ultrasound, Beijing, China.,Center of Vascular Ultrasonography, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Cong Li
- Department of Neurology, the First Hospital of Jilin University, Changchun, China
| | - Ying Chen
- Department of Neurology, the First Hospital of Jilin University, Changchun, China
| |
Collapse
|
28
|
Ma Y, Ma Q, Wang X, Yu T, Dang Y, Shang J, Li G, Hou Y. Incremental Prognostic Value of Pericoronary Adipose Tissue Thickness Measured Using Cardiac Magnetic Resonance Imaging After Revascularization in Patients With ST-Elevation Myocardial Infarction. Front Cardiovasc Med 2022; 9:781402. [PMID: 35317286 PMCID: PMC8934413 DOI: 10.3389/fcvm.2022.781402] [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: 09/22/2021] [Accepted: 02/07/2022] [Indexed: 11/13/2022] Open
Abstract
Background and AimPericoronary adipose tissue (PCAT) reflects pericoronary inflammation and is associated with coronary artery disease. We aimed to identify the association between local PCTA thickness using cardiac magnetic resonance (CMR) and prognosis of patients with ST-elevation myocardial infarction (STEMI), and to investigate the incremental prognostic value of PCAT thickness in STEMI after reperfusion.MethodsA total of 245 patients with STEMI (mean age, 55.61 ± 10.52 years) who underwent CMR imaging within 1 week of percutaneous coronary intervention therapy and 35 matched controls (mean age, 53.89 ± 9.45 years) were enrolled. PCAT thickness indexed to body surface area at five locations, ventricular volume and function, infarct-related parameters, and global strain indices were evaluated using CMR. Associations between PCAT thickness index and 1-year major adverse cardiovascular events (MACE) after STEMI were calculated. The prognostic value of the standard model based on features of clinical and CMR and updated model including PACT thickness index were further assessed.ResultsPatients with MACE had a more significant increase in PCAT thickness index at superior interventricular groove (SIVGi) than patients without MACE. The SIVGi was significantly associated with left ventricular ejection fraction (LVEF), infarct size, and global deformation. SIVGi > 4.98 mm/m2 was an independent predictor of MACE (hazard ratio, 3.2; 95% CI: 1.6–6.38; p < 0.001). The updated model significantly improved the power of prediction and had better discrimination ability than that of the standard model for predicting 1-year MACE (areas under the ROC curve [AUC] = 0.8 [95% CI: 0.74–0.87] vs. AUC = 0.76 [95% CI: 0.68–0.83], p < 0.05; category-free net reclassification index [cfNRI] = 0.38 [95% CI: 0.1–0.53, p = 0.01]; integrated discrimination improvement [IDI] = 0.09 [95% CI: 0.01–0.18, p = 0.02]).ConclusionsThis study demonstrated SIVGi as an independent predictor conferred incremental value over standard model based on clinical and CMR factors in 1-year MACE predictions for STEMI.
Collapse
Affiliation(s)
- Yue Ma
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Quanmei Ma
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaonan Wang
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Tongtong Yu
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yuxue Dang
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jin Shang
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Guangxiao Li
- Department of Medical Record Management Center, The First Hospital of China Medical University, Shenyang, China
| | - Yang Hou
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
- *Correspondence: Yang Hou
| |
Collapse
|
29
|
Sánchez E, Santos MD, Nuñez-Garcia M, Bueno M, Sajoux I, Yeramian A, Lecube A. Randomized Clinical Trial to Evaluate the Morphological Changes in the Adventitial Vasa Vasorum Density and Biological Markers of Endothelial Dysfunction in Subjects with Moderate Obesity Undergoing a Very Low-Calorie Ketogenic Diet. Nutrients 2021; 14:33. [PMID: 35010908 PMCID: PMC8746664 DOI: 10.3390/nu14010033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 12/18/2021] [Accepted: 12/20/2021] [Indexed: 12/13/2022] Open
Abstract
Weight loss after bariatric surgery decreases the earlier expansion of the adventitial vasa vasorum (VV), a biomarker of early atheromatous disease. However, no data are available regarding weight loss achieved by very low calorie ketogenic diets (VLCKD) on VV and lipid-based atherogenic indices. A randomized clinical trial was performed to examine changes in adventitial VV density in 20 patients with moderate obesity who underwent a 6-month very low calorie ketogenic diet (VLCKD, 600-800 kcal/day), and 10 participants with hypocaloric diet based on the Mediterranean Diet (MedDiet, estimated reduction of 500 kcal on the usual intake). Contrast-enhanced carotid ultrasound was used to assess the VV. Body composition analysis was also used. The atherogenic index of plasma (log (triglycerides to high-density lipoprotein cholesterol ratio)) and the triglyceride-glucose index were calculated. Serum concentrations of soluble intercellular adhesion molecule 1 (sICAM-1), and soluble vascular cell adhesion molecule 1 (sVCAM-1) were measured. The impact of weight on quality of life-lite (IWQOL-Lite) questionnaire was administered. Participants of intervention groups displayed a similar VV values. Significant improvements of BMI (-5.3 [-6.9 to -3.6] kg/m2, p < 0.001), total body fat (-7.0 [-10.7 to -3.3] %, p = 0.003), and IWQOL-Lite score (-41.4 [-75.2 to -7.6], p = 0.027) were observed in VLCKD group in comparison with MedDiet group. Although after a 6-months follow-up period VV density (mean, right and left sides) did not change significantly in any group, participants in the VLCKD exhibited a significantly decrease both in their atherogenic index of plasma and serum concentration of sICAM-1. A 6-month intervention with VLCKD do not impact in the density of the adventitial VV in subjects with moderate obesity, but induces significant changes in markers of endothelial dysfunction and CV risk.
Collapse
Affiliation(s)
- Enric Sánchez
- Obesity, Diabetes and Metabolism (ODIM) Research Group, IRBLleida, University of Lleida, 25198 Lleida, Spain; (E.S.); (M.-D.S.); (M.B.)
| | - Maria-Dolores Santos
- Obesity, Diabetes and Metabolism (ODIM) Research Group, IRBLleida, University of Lleida, 25198 Lleida, Spain; (E.S.); (M.-D.S.); (M.B.)
- Endocrinology and Nutrition Department, Arnau de Vilanova University Hospital, 25198 Lleida, Spain;
| | | | - Marta Bueno
- Obesity, Diabetes and Metabolism (ODIM) Research Group, IRBLleida, University of Lleida, 25198 Lleida, Spain; (E.S.); (M.-D.S.); (M.B.)
- Endocrinology and Nutrition Department, Arnau de Vilanova University Hospital, 25198 Lleida, Spain;
| | - Ignacio Sajoux
- Pronokal Group, 08009 Barcelona, Spain; (M.N.-G.); (I.S.)
| | - Andree Yeramian
- Endocrinology and Nutrition Department, Arnau de Vilanova University Hospital, 25198 Lleida, Spain;
| | - Albert Lecube
- Obesity, Diabetes and Metabolism (ODIM) Research Group, IRBLleida, University of Lleida, 25198 Lleida, Spain; (E.S.); (M.-D.S.); (M.B.)
- Endocrinology and Nutrition Department, Arnau de Vilanova University Hospital, 25198 Lleida, Spain;
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| |
Collapse
|
30
|
Botts SR, Fish JE, Howe KL. Dysfunctional Vascular Endothelium as a Driver of Atherosclerosis: Emerging Insights Into Pathogenesis and Treatment. Front Pharmacol 2021; 12:787541. [PMID: 35002720 PMCID: PMC8727904 DOI: 10.3389/fphar.2021.787541] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 12/06/2021] [Indexed: 12/28/2022] Open
Abstract
Atherosclerosis, the chronic accumulation of cholesterol-rich plaque within arteries, is associated with a broad spectrum of cardiovascular diseases including myocardial infarction, aortic aneurysm, peripheral vascular disease, and stroke. Atherosclerotic cardiovascular disease remains a leading cause of mortality in high-income countries and recent years have witnessed a notable increase in prevalence within low- and middle-income regions of the world. Considering this prominent and evolving global burden, there is a need to identify the cellular mechanisms that underlie the pathogenesis of atherosclerosis to discover novel therapeutic targets for preventing or mitigating its clinical sequelae. Despite decades of research, we still do not fully understand the complex cell-cell interactions that drive atherosclerosis, but new investigative approaches are rapidly shedding light on these essential mechanisms. The vascular endothelium resides at the interface of systemic circulation and the underlying vessel wall and plays an essential role in governing pathophysiological processes during atherogenesis. In this review, we present emerging evidence that implicates the activated endothelium as a driver of atherosclerosis by directing site-specificity of plaque formation and by promoting plaque development through intracellular processes, which regulate endothelial cell proliferation and turnover, metabolism, permeability, and plasticity. Moreover, we highlight novel mechanisms of intercellular communication by which endothelial cells modulate the activity of key vascular cell populations involved in atherogenesis, and discuss how endothelial cells contribute to resolution biology - a process that is dysregulated in advanced plaques. Finally, we describe important future directions for preclinical atherosclerosis research, including epigenetic and targeted therapies, to limit the progression of atherosclerosis in at-risk or affected patients.
Collapse
Affiliation(s)
- Steven R. Botts
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Jason E. Fish
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
| | - Kathryn L. Howe
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
- Division of Vascular Surgery, Department of Surgery, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
31
|
Bazban-Shotorbani S, Gavins F, Kant K, Dufva M, Kamaly N. A Biomicrofluidic Screening Platform for Dysfunctional Endothelium‐Targeted Nanoparticles and Therapeutics. ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202100092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Affiliation(s)
- Salime Bazban-Shotorbani
- Department of Health Technology DTU Health Tech Technical University of Denmark Lyngby 2800 Kgs. Denmark
- Department of Chemistry Molecular Sciences Research Hub (MSRH) Imperial College London London W12 0BZ UK
| | - Felicity Gavins
- Department of Life Sciences Centre for Inflammation Research and Translational Medicine (CIRTM) Brunel University London London UB8 3PH UK
| | - Krishna Kant
- Department of Physical Chemistry Biomedical Research Center of Galicia (CINBIO) University of Vigo Vigo 36310 Spain
| | - Martin Dufva
- Department of Health Technology DTU Health Tech Technical University of Denmark Lyngby 2800 Kgs. Denmark
| | - Nazila Kamaly
- Department of Chemistry Molecular Sciences Research Hub (MSRH) Imperial College London London W12 0BZ UK
| |
Collapse
|
32
|
Xia N, Hasselwander S, Reifenberg G, Habermeier A, Closs EI, Mimmler M, Jung R, Karbach S, Lagrange J, Wenzel P, Daiber A, Münzel T, Hövelmeyer N, Waisman A, Li H. B Lymphocyte-Deficiency in Mice Causes Vascular Dysfunction by Inducing Neutrophilia. Biomedicines 2021; 9:biomedicines9111686. [PMID: 34829915 PMCID: PMC8615852 DOI: 10.3390/biomedicines9111686] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 10/17/2021] [Accepted: 11/10/2021] [Indexed: 11/16/2022] Open
Abstract
B lymphocytes have been implicated in the development of insulin resistance, atherosclerosis and certain types of hypertension. In contrast to these studies, which were performed under pathological conditions, the present study provides evidence for the protective effect of B lymphocytes in maintaining vascular homeostasis under physiological conditions. In young mice not exposed to any known risk factors, the lack of B cells led to massive endothelial dysfunction. The vascular dysfunction in B cell-deficient mice was associated with an increased number of neutrophils in the circulating blood. Neutrophil depletion in B cell-deficient mice resulted in the complete normalization of vascular function, indicating a causal role of neutrophilia. Moreover, vascular function in B cell-deficient mice could be restored by adoptive transfer of naive B-1 cells isolated from wild-type mice. Interestingly, B-1 cell transfer also reduced the number of neutrophils in the recipient mice, further supporting the involvement of neutrophils in the vascular pathology caused by B cell-deficiency. In conclusion, we report in the present study the hitherto undescribed role of B lymphocytes in regulating vascular function. B cell dysregulation may represent a crucial mechanism in vascular pathology.
Collapse
Affiliation(s)
- Ning Xia
- Department of Pharmacology, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany; (N.X.); (S.H.); (G.R.); (A.H.); (E.I.C.); (M.M.)
| | - Solveig Hasselwander
- Department of Pharmacology, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany; (N.X.); (S.H.); (G.R.); (A.H.); (E.I.C.); (M.M.)
| | - Gisela Reifenberg
- Department of Pharmacology, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany; (N.X.); (S.H.); (G.R.); (A.H.); (E.I.C.); (M.M.)
| | - Alice Habermeier
- Department of Pharmacology, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany; (N.X.); (S.H.); (G.R.); (A.H.); (E.I.C.); (M.M.)
| | - Ellen I. Closs
- Department of Pharmacology, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany; (N.X.); (S.H.); (G.R.); (A.H.); (E.I.C.); (M.M.)
| | - Maximilian Mimmler
- Department of Pharmacology, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany; (N.X.); (S.H.); (G.R.); (A.H.); (E.I.C.); (M.M.)
| | - Rebecca Jung
- Institute for Molecular Medicine, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany; (R.J.); (N.H.); (A.W.)
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany; (S.K.); (J.L.); (P.W.)
| | - Susanne Karbach
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany; (S.K.); (J.L.); (P.W.)
- Department of Cardiology, Cardiology 1, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany; (A.D.); (T.M.)
| | - Jérémy Lagrange
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany; (S.K.); (J.L.); (P.W.)
- Department of Cardiology, Cardiology 1, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany; (A.D.); (T.M.)
| | - Philip Wenzel
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany; (S.K.); (J.L.); (P.W.)
- Department of Cardiology, Cardiology 1, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany; (A.D.); (T.M.)
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, 55131 Mainz, Germany
| | - Andreas Daiber
- Department of Cardiology, Cardiology 1, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany; (A.D.); (T.M.)
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, 55131 Mainz, Germany
| | - Thomas Münzel
- Department of Cardiology, Cardiology 1, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany; (A.D.); (T.M.)
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, 55131 Mainz, Germany
| | - Nadine Hövelmeyer
- Institute for Molecular Medicine, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany; (R.J.); (N.H.); (A.W.)
- Research Center for Immunotherapy (FZI), Johannes Gutenberg University Medical Center, 55131 Mainz, Germany
| | - Ari Waisman
- Institute for Molecular Medicine, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany; (R.J.); (N.H.); (A.W.)
- Research Center for Immunotherapy (FZI), Johannes Gutenberg University Medical Center, 55131 Mainz, Germany
| | - Huige Li
- Department of Pharmacology, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany; (N.X.); (S.H.); (G.R.); (A.H.); (E.I.C.); (M.M.)
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, 55131 Mainz, Germany
- Correspondence: ; Tel.: +49-(6131)-17-9348; Fax: +49-(6131)-17-9329
| |
Collapse
|
33
|
Unno N, Tanaka H, Yata T, Kayama T, Yamanaka Y, Tsuyuki H, Sano M, Inuzuka K, Naruse E, Takeuchi H. K-134, a phosphodiesterase 3 inhibitor, reduces vascular inflammation and hypoxia, and prevents rupture of experimental abdominal aortic aneurysms. JVS Vasc Sci 2021; 1:219-232. [PMID: 34617050 PMCID: PMC8489215 DOI: 10.1016/j.jvssci.2020.09.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 09/21/2020] [Indexed: 11/10/2022] Open
Abstract
Objective Abdominal aortic aneurysm (AAA) is a chronic inflammatory disease, which frequently results in fatal rupture; however, no pharmacologic treatment exists to inhibit AAA growth and prevent rupture. In this study, we investigated whether K-134, a novel phosphodiesterase 3 inhibitor, could limit the progression and rupture of AAA using multiple experimental models. Methods A hypoperfusion-induced AAA rat model was developed by inserting of a small catheter and via tight ligation of the infrarenal aorta. Rats were fed with a 0.15% K-134-containing diet (K-134(+) group) or a normal diet (K-134(-) group) from 7 days before the experiment to 28 days after model creation (pretreatment protocol). After the administration period, elastin fragmentation, macrophage infiltration, reactive oxygen species expression, matrix metalloproteinase levels, aneurysmal tissue hypoxia, and adventitial vasa vasorum (VV) stenosis were assessed. In the delayed treatment protocol, rats with AAA >3 mm were randomly divided to K-134(+) or K-134(-) group 7 days after model creation, and the effect of K-134 on suppressing preexisting AAA was examined. Further, elastase-induced rat model and angiotensin II-infused ApoE-/- mouse model were also used to examine the ability of K-134 to prevent rupture. Results K-134 prevented AAA rupture and significantly improved survival in the pretreatment protocol (P < .01). In the K-134(+) group, elastin degeneration was prevented; macrophage infiltration and reactive oxygen species production were significantly decreased. At 14 days, the enzymatic activity of matrix metalloproteinase-9 was significantly decreased. Further, K-134 inhibited intimal hyperplasia and VV stenosis. Expressions of hypoxic markers, hypoxia-inducible factor-1α, and pimonidazole, in the aneurysmal wall were also attenuated. In the delayed treatment protocol, K-134 also improved survival of rats with preexisting AAA. Similarly, in the elastase-induced rat model and angiotensin II-infused ApoE-/- mouse model, K-134 inhibited rupture and significantly improved survival (P < .01). Conclusions K-134 prevented the rupture of AAA and improved survival through suppressing inflammatory reaction. The inhibition of intimal hyperplasia in the adventitial VV may be associated with reduced hypoxia in the aneurysmal tissue. (JVS–Vascular Science 2020;1:219-32.) Clinical Relevance This study shows that K-134, a novel phosphodiesterase 3 inhibitor, suppressed abdominal aortic aneurysm (AAA) rupture. Considering that K-134 had already undergone a phase Ⅱ study in the United States for claudication in peripheral artery occlusive disease patients with good tolerance, K-134 may become a promising new therapeutic option for AAAs and could undergo clinical trials for patients with small AAA.
Collapse
Affiliation(s)
- Naoki Unno
- Division of Vascular Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan.,Second Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan.,Division of Vascular Surgery, Hamamatsu Medical Center, Hamamatsu, Japan
| | - Hiroki Tanaka
- Division of Vascular Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Tatsuro Yata
- Division of Vascular Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan.,Second Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Takafumi Kayama
- Division of Vascular Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan.,Second Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yuta Yamanaka
- Division of Vascular Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan.,Second Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hajime Tsuyuki
- Division of Vascular Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan.,Second Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Masaki Sano
- Division of Vascular Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan.,Second Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kazunori Inuzuka
- Division of Vascular Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan.,Second Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Ena Naruse
- Division of Vascular Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan.,Second Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hiroya Takeuchi
- Second Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| |
Collapse
|
34
|
Characteristics of atherosclerosis in femoropopliteal artery and its clinical relevance. Atherosclerosis 2021; 335:31-40. [PMID: 34547588 DOI: 10.1016/j.atherosclerosis.2021.09.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 08/28/2021] [Accepted: 09/09/2021] [Indexed: 12/12/2022]
Abstract
Atherosclerosis is a systemic disease with different faces. Despite identical or similar pathogenetic mechanisms, atherosclerotic lesions and their clinical manifestations vary in different parts of the vascular system. Peripheral arterial disease (PAD) represents one of the most frequent clinical manifestations of atherosclerosis with predominant location in the superficial femoral artery (SFA). Morphological characteristics of atherosclerotic plaques in peripheral arteries differ from lesions in the coronary and carotid arteries. Plaques in SFA have more fibrotic components, less lipids and inflammatory cells, which makes them more stable and less prone to rupture. Factors that determine the different structure of plaques in SFA compared to coronary arteries include hemodynamic forces, vasa vasorum and calcification. Low shear stress in SFA in the adductor canal is one of the factors which determines frequent atherosclerotic lesions in this region. Lower lipid content and fewer inflammatory cells explain higher stability of SFA plaques. The specific structure of SFA plaques may require preventive and therapeutic measures, which to some extent differ from prevention of coronary atherosclerosis and may include inhibition of fibrotic proliferation in SFA plaques and calcification. Revascularization of PAD differs from procedures used in coronary arteries and requires specific technical expertise and devices.
Collapse
|
35
|
Hayabuchi Y, Homma Y, Kagami S. Three-dimensional imaging of pulmonary arterial vasa vasorum using optical coherence tomography in patients after bidirectional Glenn and Fontan procedures. Eur Heart J Cardiovasc Imaging 2021; 22:941-949. [PMID: 32413104 DOI: 10.1093/ehjci/jeaa098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 01/31/2020] [Accepted: 04/16/2020] [Indexed: 11/13/2022] Open
Abstract
AIMS We evaluated pulmonary arterial (PA) vasa vasorum (VV) in Fontan candidate patients with a novel three-dimensional (3D) imaging technique using optical coherence tomography (OCT). METHODS AND RESULTS This prospective study assessed the development of adventitial VV in the distal PA of 10 patients with bidirectional Glenn circulation (BDG group, 1.6 ± 0.3 years) and Fontan circulation (Fontan group, 3.3 ± 0.3 years), and in 20 children with normal PA haemodynamics and morphology (Control group, 1.5 ± 0.3 years). We assessed the PA VV with two-dimensional (2D) cross-sectional, multi-planar reconstruction (MPR), and volume rendering (VR) imaging. VV development was evaluated by the VV area/volume ratio, defined as the VV area/volume divided by the adventitial area/volume. Compared to the control group, the observed VV number and diameter on 3D images of MPR and VR were significantly higher, and curved and torturous-shaped VV were more frequently observed in the BDG and Fontan groups (P < 0.001, all). The median VV volume ratio was significantly greater in the BDG than in the control group (3.38% vs. 0.61%; P < 0.001). Although the VV volume ratio decreased significantly after the Fontan procedure (2.64%, P = 0.005 vs. BDG), the ratio remained higher than in the control group (P < 0.001 vs. control). CONCLUSION 3D OCT imaging is a novel method that can be used to evaluate adventitial PA VV and may provide pathophysiological insight into the role of the PA VV in these patients.
Collapse
Affiliation(s)
- Yasunobu Hayabuchi
- Department of Pediatrics, Tokushima University, Kuramoto-cho-3, Tokushima 770-8503, Japan
| | - Yukako Homma
- Department of Pediatrics, Tokushima University, Kuramoto-cho-3, Tokushima 770-8503, Japan
| | - Shoji Kagami
- Department of Pediatrics, Tokushima University, Kuramoto-cho-3, Tokushima 770-8503, Japan
| |
Collapse
|
36
|
Guggenberger KV, Torre GD, Ludwig U, Vogel P, Weng AM, Vogt ML, Fröhlich M, Schmalzing M, Raithel E, Forman C, Urbach H, Meckel S, Bley TA. Vasa vasorum of proximal cerebral arteries after dural crossing - potential imaging confounder in diagnosing intracranial vasculitis in elderly subjects on black-blood MRI. Eur Radiol 2021; 32:1276-1284. [PMID: 34347156 PMCID: PMC8795054 DOI: 10.1007/s00330-021-08181-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 05/18/2021] [Accepted: 05/23/2021] [Indexed: 11/24/2022]
Abstract
Objectives Vessel wall enhancement (VWE) may be commonly seen on MRI images of asymptomatic subjects. This study aimed to characterize the VWE of the proximal internal carotid (ICA) and vertebral arteries (VA) in a non-vasculitic elderly patient cohort. Methods Cranial MRI scans at 3 Tesla were performed in 43 patients (aged ≥ 50 years) with known malignancy for exclusion of cerebral metastases. For vessel wall imaging (VWI), a high-resolution compressed-sensing black-blood 3D T1-weighted fast (turbo) spin echo sequence (T1 CS-SPACE prototype) was applied post gadolinium with an isotropic resolution of 0.55 mm. Bilateral proximal intradural ICA and VA segments were evaluated for presence, morphology, and longitudinal extension of VWE. Results Concentric VWE of the proximal intradural ICA was found in 13 (30%) patients, and of the proximal intradural VA in 39 (91%) patients. Mean longitudinal extension of VWE after dural entry was 13 mm in the VA and 2 mm in the ICA. In 14 of 39 patients (36%) with proximal intradural VWE, morphology of VWE was suggestive of the mere presence of vasa vasorum. In 25 patients (64 %), morphology indicated atherosclerotic lesions in addition to vasa vasorum. Conclusions Vasa vasorum may account for concentric VWE within the proximal 2 mm of the ICA and 13 mm of the VA after dural entry in elderly subjects. Concentric VWE in these locations should not be confused with large artery vasculitis. Distal to these segments, VWE may be more likely related to pathologic conditions such as vasculitis. Key Points • Vasa vasorum may account for concentric VWE within the proximal 2 mm of the ICA and 13 mm of the VA after dural entry in non-vasculitic elderly people. • Concentric enhancement within the proximal 2 mm of the intradural ICA and within the proximal 13 mm of the intradural VA portions should not be misinterpreted as vasculitis. • Distal of this, VWE is likely related to pathologic conditions, in case of concentric VWE suggestive of vasculitis.
Collapse
Affiliation(s)
- Konstanze Viktoria Guggenberger
- Department of Diagnostic and Interventional Radiology, University Hospital Wuerzburg, Faculty of Medicine, University of Wuerzburg, Oberduerrbacher Straße 6, 97080, Wuerzburg, Germany.
| | - Giulia Dalla Torre
- Department of Diagnostic and Interventional Radiology, University Hospital Wuerzburg, Faculty of Medicine, University of Wuerzburg, Oberduerrbacher Straße 6, 97080, Wuerzburg, Germany
| | - Ute Ludwig
- Department of Radiology, Medical Physics, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Patrick Vogel
- Department of Diagnostic and Interventional Radiology, University Hospital Wuerzburg, Faculty of Medicine, University of Wuerzburg, Oberduerrbacher Straße 6, 97080, Wuerzburg, Germany.,Department of Experimental Physics 5 (Biophysics), University of Wuerzburg, 97074, Wuerzburg, Germany
| | - Andreas Max Weng
- Department of Diagnostic and Interventional Radiology, University Hospital Wuerzburg, Faculty of Medicine, University of Wuerzburg, Oberduerrbacher Straße 6, 97080, Wuerzburg, Germany
| | - Marius Lothar Vogt
- Department of Diagnostic and Interventional Neuroradiology, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Matthias Fröhlich
- Department of Internal Medicine II, Rheumatology and Clinical Immunology, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Marc Schmalzing
- Department of Internal Medicine II, Rheumatology and Clinical Immunology, University Hospital Wuerzburg, Wuerzburg, Germany
| | | | | | - Horst Urbach
- Department of Neuroradiology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Breisacher Straße 64, 79106, Freiburg, Germany
| | - Stephan Meckel
- Department of Neuroradiology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Breisacher Straße 64, 79106, Freiburg, Germany
| | - Thorsten Alexander Bley
- Department of Diagnostic and Interventional Radiology, University Hospital Wuerzburg, Faculty of Medicine, University of Wuerzburg, Oberduerrbacher Straße 6, 97080, Wuerzburg, Germany
| |
Collapse
|
37
|
Chen L, Fang Z, Wang X, Sun X, Ge X, Cheng C, Hu H. G protein-coupled receptor 39 activation alleviates oxidized low-density lipoprotein-induced macrophage inflammatory response, lipid accumulation and apoptosis by inducing A20 expression. Bioengineered 2021; 12:4070-4080. [PMID: 34288802 PMCID: PMC8806696 DOI: 10.1080/21655979.2021.1952917] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
G protein-coupled receptor 39 (GPR39) agonist weakens oxidized low-density lipoprotein (ox-LDL)-induced attachment of monocytes to vascular endothelial cells and thus alleviates atherosclerosis. This study looks at whether GPR39 protects macrophages against ox-LDL-induced inflammation and apoptosis and ameliorates lipid accumulation in atherosclerosis and investigates its mechanism. Following inducement of ox-LDL, the expression of GPR39 and tumor necrosis factor alpha-induced protein 3 (TNFAIP3, also known as A20) in Raw 264.7 cells was detected by RT-qPCR and western blotting. The viability of macrophages treated with GPR39 agonist was detected by a cell counting kit 8 kit. GPR39 and A20 expression in ox-LDL-challenged macrophages was assayed by RT-qPCR and western blot with or without GPR30 agonist. After transfection of small interfering RNA (siRNA)-A20, the expression of pro-inflammatory cytokine tumor necrosis factor-α (TNF-α), interleukin (IL)-1β and IL-6 and anti-inflammatory cytokine IL-10 as well as NF-κB p65 and COX2 was detected. Lipid accumulation was observed through Oil Red O Staining. Total cholesterol (TC) and free cholesterol (FC) in macrophages were detected by commercial kits. Lastly, macrophage apoptosis was observed through TUNEL, and apoptosis-related proteins were detected by western blotting . Results indicated that decreased expression of GPR39 and A20 was observed in ox-LDL-induced macrophages. GPR39 agonist significantly increased A20 expression in ox-LDL-treated macrophages. Furthermore, A20 interference reversed the inhibitory effect of GPR39 agonist on ox-LDL-induced inflammation, lipid accumulation, TC and FC overexpression as well as cell apoptosis. In conclusion, activating GPR39 alleviates ox-LDL-induced macrophage inflammation, lipid accumulation and apoptosis in an A20-dependent manner.
Collapse
Affiliation(s)
- Lu Chen
- Department of Vascular Surgery, Provincial Hospital Affiliated to Anhui Medical University, Hefei City, Anhui Province, China
| | - Zhengdong Fang
- Department of Vascular Surgery, The First Affiliated Hospital of USTC, Hefei City, Anhui Province, China
| | - Xiaotian Wang
- Department of Vascular Surgery, The First Affiliated Hospital of USTC, Hefei City, Anhui Province, China
| | - Xiaojie Sun
- Department of Vascular Surgery, The First Affiliated Hospital of USTC, Hefei City, Anhui Province, China
| | - Xinbao Ge
- Department of Vascular Surgery, The First Affiliated Hospital of USTC, Hefei City, Anhui Province, China
| | - Can Cheng
- Department of Vascular Surgery, The First Affiliated Hospital of USTC, Hefei City, Anhui Province, China
| | - Hejie Hu
- Department of Vascular Surgery, The First Affiliated Hospital of USTC, Hefei City, Anhui Province, China
| |
Collapse
|
38
|
Xue Q, Chen L, Yu J, Sun K, Ye L, Zheng J. Downregulation of Interleukin-13 Receptor α2 Inhibits Angiogenic Formation Mediated by Chitinase 3-Like 1 in Late Atherosclerotic Lesions of apoE -/- Mice. Front Physiol 2021; 12:690109. [PMID: 34349665 PMCID: PMC8327173 DOI: 10.3389/fphys.2021.690109] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 06/03/2021] [Indexed: 11/13/2022] Open
Abstract
Aim: Chitinase 3-like 1 (CHI3L1) has the potential to prompt proliferation and angiogenic formation. Interleukin-13 receptor α2 (IL-13Rα2) was regarded as a receptor of CHI3L1; however, it is unknown whether CHI3L1 adjusts the neovascularization in late atherosclerotic lesions of apoE -/- mice via IL-13Rα2. Methods: Silicone collars were placed around one of the common carotid arteries of apoE -/- mice fed with a high-fat diet. The mice were further injected with Ad.CHI3L1 alone or Ad.CHI3L1 + Ad.IL-13Rα2 shRNA through the caudal vein. The plaque areas in the whole aorta and aortic root were evaluated by Oil Red O staining and H&E staining. The contents of CD31, CD42b, and collagen in carotid plaques were investigated by immunohistochemistry and Masson trichrome staining. The role of CHI3L1 in migration and tube formation of human umbilical vein endothelial cells (HUVECs) was determined by transwell and Matrigel tests. The effect of CHI3L1 on the expression of AKT and extracellular signal-regulated kinase (ERK) was evaluated with the Western blot. Results: The plaque loads in the aorta were significantly more extensive in apoE -/- mice injected with Ad.CHI3L1 than those with Ad.CHI3L1 + Ad.IL-13Rα2 shRNA. CHI3L1 significantly increased the contents of CD31 and CD42b and decreased the element of collagen in late-stage atherosclerotic lesions of the carotid arteries. The effects of CHI3L1 on migration, tube formation, and upregulation of phospho-AKT and phospho-ERK of HUVECs were prohibited by inhibitors of phosphatidylinositol 3-kinase (PI3K) and mitogen-activated protein kinase kinase (MEK) as well as IL-13Rα2 shRNA. Conclusion: To some extent, CHI3L1 promotes migration and tube formation of HUVECs and neovascularization in atherosclerotic plaques possibly mediated by IL-13Rα2 through AKT and ERK signal pathways.
Collapse
Affiliation(s)
- Qi Xue
- Department of Cardiology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Lei Chen
- Department of Pathology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Jianwu Yu
- Department of Cardiology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Kewang Sun
- Department of Cardiology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Lifang Ye
- Department of Cardiology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Jianlei Zheng
- Department of Cardiology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China.,Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
39
|
Abstract
Abdominal aortic aneurysm (AAA) is a common disease associated with significant cardiovascular morbidity and mortality. Up to now, there is still controversy on the choice of treatment method of AAA. Even so, the mechanisms of AAA progression are poorly defined, making targeting new therapies problematic. Current evidence favors an interaction of the hemodynamic microenvironment with local and systemic immune responses. In this review, we aim to provide an update of mechanisms in AAA progression, involving hemodynamics, perivascular adipose tissue, adventitial fibroblasts, vasa vasorum remodeling, intraluminal thrombus, and distribution of macrophage subtypes.
Collapse
Affiliation(s)
- Jiang-Ping Gao
- Department of Vascular Surgery, Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, China
| | - Wei Guo
- Department of Vascular Surgery, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
40
|
Influence of Disorders of Fatty Acid Metabolism, Arterial Wall Hypoxia, and Intraplaque Hemorrhages on Lipid Accumulation in Atherosclerotic Vessels. ACTA BIOMEDICA SCIENTIFICA 2021. [DOI: 10.29413/abs.2021-6.2.8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The review describes a number of competing views on the main causes of cholesterol accumulation in atherosclerotic vessels. On the one hand, unregulated cholesterol influx into arterial intima is primarily related to the increasing proportion of atherogenic lipoproteins in the lipoprotein spectrum of blood. On the other hand, the leading role in this process is assigned to the increased permeability of endothelium for atherogenic lipoproteins. The increased ability of arterial intima connective tissue to bind atherogenic blood lipoproteins is also considered to be the leading cause of cholesterol accumulation in the vascular wall. The key role in cholesterol accumulation is also assigned to unregulated (by a negative feedback mechanism) absorption of atherogenic lipoproteins by foam cells. It is suggested that the main cause of abundant cholesterol accumulation in atherosclerotic vessels is significant inflow of this lipid into the vascular wall during vasa vasorum hemorrhages.The article also provides arguments, according to which disorder of fatty acid metabolism in arterial wall cells can initiate accumulation of neutral lipids in them, contribute to the inflammation and negatively affect the mechanical conditions around the vasa vasorum in the arterial walls. As a result, the impact of pulse waves on the luminal surface of the arteries will lead to frequent hemorrhages of these microvessels. At the same time, adaptive-muscular intima hyperplasia, which develops in arterial channel areas subjected to high hemodynamic loads, causes local hypoxia in a vascular wall. As a result, arterial wall cells undergo even more severe lipid transformation. Hypoxia also stimulates vascularization of the arterial wall, which contributes to hemorrhages in it. With hemorrhages, free erythrocyte cholesterol penetrates into the forming atherosclerotic plaque, a part of this cholesterol forms cholesterol esters inside the arterial cells. The saturation of erythrocyte membranes with this lipid in conditions of hypercholesterolemia and atherogenic dyslipoproteinemia contributes to the process of cholesterol accumulation in arteries.
Collapse
|
41
|
Rios-Navarro C, Daghbouche-Rubio N, Gavara J, de Dios E, Perez N, Vila JM, Chorro FJ, Ruiz-Sauri A, Bodi V. Ischemia-reperfusion injury to coronary arteries: Comprehensive microscopic study after reperfused myocardial infarction. Ann Anat 2021; 238:151785. [PMID: 34144157 DOI: 10.1016/j.aanat.2021.151785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 05/25/2021] [Accepted: 05/26/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Coronary arteries supply oxygen and nutrients to the heart. We evaluated the dynamics of microscopic damage throughout the ischemia-reperfusion process in the wall of coronary arteries following myocardial infarction (MI). METHODS In a swine model of reperfused MI, animals were divided into one control and four MI groups: 90-min ischemia without reperfusion, or followed by one minute, one week or one month reperfusion. Left anterior descending (LAD; infarct-related artery) and control right coronary arteries (RCA) were isolated. Taking the balloon inflation region as a reference, we isolated the proximal and distal LAD areas, performing histological staining and immunohistochemistry. RESULTS Although mild changes in tunica intima were observed during ischemia, an almost complete absence of endothelium, and abnormal breaks in the internal elastic layer were found post-revascularization. In tunica media, increased thickness was observed soon after reperfusion, whereas larger thickness, disorganized muscle cell distribution and edema were found one week after reperfusion. This damage was more pronounced in distal rather than proximal LAD, whereas no changes were detected in RCA. In the tunica adventitia, vasa vasorum density decayed during ischemia in both LAD regions, but was restored after one month. Leukocyte adhesion to the artery was observed post-revascularization, developing into a massive presence in the three layers one week post-reperfusion. CONCLUSIONS Ischemia-reperfusion can itself induce damage in the wall of the epicardial coronary artery, becoming more pronounced in the region distal to balloon inflation. Exploring these abnormalities will provide insight into the pathophysiology of coronary circulation and MI.
Collapse
Affiliation(s)
| | | | - Jose Gavara
- INCLIVA Health Research Institute, Valencia, Spain
| | - Elena de Dios
- Department of Medicine, School of Medicine, University of Valencia, Valencia, Spain; Centro de Investigación Biomédica en Red - Cardiovascular, Spain
| | - Nerea Perez
- INCLIVA Health Research Institute, Valencia, Spain
| | - Jose M Vila
- INCLIVA Health Research Institute, Valencia, Spain; Department of Physiology, School of Medicine, University of Valencia, Valencia, Spain
| | - Francisco J Chorro
- INCLIVA Health Research Institute, Valencia, Spain; Department of Medicine, School of Medicine, University of Valencia, Valencia, Spain; Centro de Investigación Biomédica en Red - Cardiovascular, Spain; Cardiology Department, Hospital Clinico Universitario, Valencia, Spain
| | - Amparo Ruiz-Sauri
- INCLIVA Health Research Institute, Valencia, Spain; Department of Pathology, School of Medicine, University of Valencia, Valencia, Spain.
| | - Vicente Bodi
- INCLIVA Health Research Institute, Valencia, Spain; Department of Medicine, School of Medicine, University of Valencia, Valencia, Spain; Centro de Investigación Biomédica en Red - Cardiovascular, Spain; Cardiology Department, Hospital Clinico Universitario, Valencia, Spain.
| |
Collapse
|
42
|
Li L, Dmytriw AA, Jiao L. Response to: effect of vasa vasorum in cerebrovascular compensation: 2 case reports. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1029. [PMID: 34277829 PMCID: PMC8267289 DOI: 10.21037/atm-21-855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 04/25/2021] [Indexed: 11/16/2022]
Affiliation(s)
- Long Li
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Adam A Dmytriw
- Neuroradiology & Neurointervention Service, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Liqun Jiao
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.,Department of Interventional Neuroradiology, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
43
|
Turpin C, Catan A, Meilhac O, Bourdon E, Canonne-Hergaux F, Rondeau P. Erythrocytes: Central Actors in Multiple Scenes of Atherosclerosis. Int J Mol Sci 2021; 22:ijms22115843. [PMID: 34072544 PMCID: PMC8198892 DOI: 10.3390/ijms22115843] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 05/26/2021] [Accepted: 05/26/2021] [Indexed: 12/16/2022] Open
Abstract
The development and progression of atherosclerosis (ATH) involves lipid accumulation, oxidative stress and both vascular and blood cell dysfunction. Erythrocytes, the main circulating cells in the body, exert determinant roles in the gas transport between tissues. Erythrocytes have long been considered as simple bystanders in cardiovascular diseases, including ATH. This review highlights recent knowledge concerning the role of erythrocytes being more than just passive gas carriers, as potent contributors to atherosclerotic plaque progression. Erythrocyte physiology and ATH pathology is first described. Then, a specific chapter delineates the numerous links between erythrocytes and atherogenesis. In particular, we discuss the impact of extravasated erythrocytes in plaque iron homeostasis with potential pathological consequences. Hyperglycaemia is recognised as a significant aggravating contributor to the development of ATH. Then, a special focus is made on glycoxidative modifications of erythrocytes and their role in ATH. This chapter includes recent data proposing glycoxidised erythrocytes as putative contributors to enhanced atherothrombosis in diabetic patients.
Collapse
Affiliation(s)
- Chloé Turpin
- Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), INSERM, UMR 1188, Université de La Réunion, 97400 Saint Denis, France; (C.T.); (A.C.); (O.M.); (E.B.)
| | - Aurélie Catan
- Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), INSERM, UMR 1188, Université de La Réunion, 97400 Saint Denis, France; (C.T.); (A.C.); (O.M.); (E.B.)
| | - Olivier Meilhac
- Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), INSERM, UMR 1188, Université de La Réunion, 97400 Saint Denis, France; (C.T.); (A.C.); (O.M.); (E.B.)
- Centre Hospitalier Universitaire de La Réunion, 97400 Saint Denis, France
| | - Emmanuel Bourdon
- Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), INSERM, UMR 1188, Université de La Réunion, 97400 Saint Denis, France; (C.T.); (A.C.); (O.M.); (E.B.)
| | | | - Philippe Rondeau
- Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), INSERM, UMR 1188, Université de La Réunion, 97400 Saint Denis, France; (C.T.); (A.C.); (O.M.); (E.B.)
- Correspondence: ; Tel.: +262(0)-2-62-93-88-43; Fax: +262-(0)-2-62-93-88-01
| |
Collapse
|
44
|
Yang X, Wang L, Zhang Z, Hu J, Liu X, Wen H, Liu M, Zhang X, Dai H, Ni M, Li R, Guo R, Zhang L, Luan X, Lin H, Dong M, Lu H. Ginsenoside Rb 1 Enhances Plaque Stability and Inhibits Adventitial Vasa Vasorum via the Modulation of miR-33 and PEDF. Front Cardiovasc Med 2021; 8:654670. [PMID: 34124194 PMCID: PMC8192703 DOI: 10.3389/fcvm.2021.654670] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 04/20/2021] [Indexed: 01/25/2023] Open
Abstract
Background: Atherosclerosis is closely associated with proliferation of the adventitial vasa vasorum, leading to the atherosclerotic plaque progression and vulnerability. In this report, we investigated the role of Ginsenoside Rb1 (Rb1) on atherosclerotic plaque stabilization and adventitial vasa vasorum (VV) along with the mechanisms involved. Methods and Results: Apolipoprotein E-deficient (ApoE-/-) mice were fed with a high-fat diet for 20 weeks, and then Ginsenoside Rb1 (50 mg/kg/d, intraperitoneal) was given for 4 weeks. Rb1 treatment significantly inhibited adventitial VV proliferation, alleviated inflammation, decreased plaque burden, and stabilized atherosclerotic plaques in apoE-/- mice. However, the beneficial effects of Rb1 on atherosclerotic lesion was attenuated by overexpression of miR-33. The analysis from atherosclerotic plaque revealed that Rb1 treatment could result in an induction of Pigment epithelium-derived factor (PEDF) expression and reduction of the miR-33 generation. Overexpression of miR-33 significantly reverted the Rb1-mediated elevation of PEDF and anti-angiogenic effect. Conclusions: Ginsenoside Rb1 attenuates plaque growth and enhances plaque stability partially through inhibiting adventitial vasa vasorum proliferation and inflammation in apoE-/- mice. The anti-angiogenic and anti-inflammation effects of Rb1 are exerted via the modulation of miR-33 and its target gene PEDF.
Collapse
Affiliation(s)
- Xiaoyan Yang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Cardiology, Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital Affiliated to Capital Medical University, Beijing, China
| | - Lei Wang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zihao Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jiayi Hu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaoling Liu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Hao Wen
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,The Second School of Clinical Medicine, Binzhou Medical University, Yantai, China
| | - Minghao Liu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xue Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Cardiology, Qingdao Municipal Hospital, Qingdao, China
| | - Hongyan Dai
- Department of Cardiology, Qingdao Municipal Hospital, Qingdao, China
| | - Mei Ni
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Rui Li
- Department of Cardiology, China-Japan Friendship Hospital, Ministry of Health, Beijing, China
| | - Rong Guo
- Department of Cardiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Lei Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaorong Luan
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Huili Lin
- Department of Cardiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Mei Dong
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Huixia Lu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
45
|
Iannuzzi A, Rubba P, Gentile M, Mallardo V, Calcaterra I, Bresciani A, Covetti G, Cuomo G, Merone P, Di Lorenzo A, Alfieri R, Aliberti E, Giallauria F, Di Minno MND, Iannuzzo G. Carotid Atherosclerosis, Ultrasound and Lipoproteins. Biomedicines 2021; 9:biomedicines9050521. [PMID: 34066616 PMCID: PMC8148516 DOI: 10.3390/biomedicines9050521] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 04/30/2021] [Accepted: 05/04/2021] [Indexed: 12/13/2022] Open
Abstract
Carotid artery plaques are considered a measure of atherosclerosis and are associated with an increased risk of atherosclerotic cardiovascular disease, particularly ischemic strokes. Monitoring of patients with an elevated risk of stroke is critical in developing better prevention strategies. Non-invasive imaging allows us to directly see atherosclerosis in vessels and many features that are related to plaque vulnerability. A large body of evidence has demonstrated a strong correlation between some lipid parameters and carotid atherosclerosis. In this article, we review the relationship between lipids and atherosclerosis with a focus on carotid ultrasound, the most common method to estimate atherosclerotic load.
Collapse
Affiliation(s)
- Arcangelo Iannuzzi
- Department of Medicine and Medical Specialties, A. Cardarelli Hospital, 80131 Naples, Italy; (A.B.); (G.C.)
- Correspondence:
| | - Paolo Rubba
- Department of Clinical Medicine, Surgery Federico II University, 80131 Naples, Italy; (P.R.); (M.G.); (V.M.); (I.C.); (M.N.D.D.M.); (G.I.)
| | - Marco Gentile
- Department of Clinical Medicine, Surgery Federico II University, 80131 Naples, Italy; (P.R.); (M.G.); (V.M.); (I.C.); (M.N.D.D.M.); (G.I.)
| | - Vania Mallardo
- Department of Clinical Medicine, Surgery Federico II University, 80131 Naples, Italy; (P.R.); (M.G.); (V.M.); (I.C.); (M.N.D.D.M.); (G.I.)
| | - Ilenia Calcaterra
- Department of Clinical Medicine, Surgery Federico II University, 80131 Naples, Italy; (P.R.); (M.G.); (V.M.); (I.C.); (M.N.D.D.M.); (G.I.)
| | - Alessandro Bresciani
- Department of Medicine and Medical Specialties, A. Cardarelli Hospital, 80131 Naples, Italy; (A.B.); (G.C.)
| | - Giuseppe Covetti
- Department of Medicine and Medical Specialties, A. Cardarelli Hospital, 80131 Naples, Italy; (A.B.); (G.C.)
| | - Gianluigi Cuomo
- Department of Translational Medical Sciences, Federico II University of Naples, 80131 Naples, Italy; (G.C.); (P.M.); (A.D.L.); (R.A.); (F.G.)
| | - Pasquale Merone
- Department of Translational Medical Sciences, Federico II University of Naples, 80131 Naples, Italy; (G.C.); (P.M.); (A.D.L.); (R.A.); (F.G.)
| | - Anna Di Lorenzo
- Department of Translational Medical Sciences, Federico II University of Naples, 80131 Naples, Italy; (G.C.); (P.M.); (A.D.L.); (R.A.); (F.G.)
| | - Roberta Alfieri
- Department of Translational Medical Sciences, Federico II University of Naples, 80131 Naples, Italy; (G.C.); (P.M.); (A.D.L.); (R.A.); (F.G.)
| | - Emilio Aliberti
- North Tees University Hospital, Stockton-on Tees TS19 8PE, UK;
| | - Francesco Giallauria
- Department of Translational Medical Sciences, Federico II University of Naples, 80131 Naples, Italy; (G.C.); (P.M.); (A.D.L.); (R.A.); (F.G.)
| | - Matteo Nicola Dario Di Minno
- Department of Clinical Medicine, Surgery Federico II University, 80131 Naples, Italy; (P.R.); (M.G.); (V.M.); (I.C.); (M.N.D.D.M.); (G.I.)
| | - Gabriella Iannuzzo
- Department of Clinical Medicine, Surgery Federico II University, 80131 Naples, Italy; (P.R.); (M.G.); (V.M.); (I.C.); (M.N.D.D.M.); (G.I.)
| |
Collapse
|
46
|
Li M, Qi Z, Zhang J, Zhu K, Wang Y. Effect and Mechanism of Si-Miao-Yong-An on Vasa Vasorum Remodeling in ApoE -/- Mice with Atherosclerosis Vulnerable Plague. Front Pharmacol 2021; 12:634611. [PMID: 33935723 PMCID: PMC8080061 DOI: 10.3389/fphar.2021.634611] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 01/26/2021] [Indexed: 01/20/2023] Open
Abstract
Objective: To observe the effect of Si-Miao-Yong-An (SMYA) on atherosclerosis (AS) vulnerable plaques, and to further explore the mechanism by vasa vasorum (VV) angiogenesis and maturation as an entry point. Methods: SPF-class healthy male ApoE−/− mice were randomized into model group, simvastatin group and SMYA group, and C57BL/6 mice were used as the control group. After 8 weeks of intervention, the pathological morphology of plaque was observed by HE staining; the VV density in plaque and aortic adventitia were observed by immunohistochemistry; VV maturation was measured by double-labelling immunofluorescence; the critical proteins of HIF-1α-Apelin/APJ and Ang-1/Tie signal pathways were detected by western blotting. Results: SMYA decreased the plaque area and the ratio of plaque to lumen area; increased the minimum thickness of fibrous cap and its effect was greater than simvastatin. SMYA suppressed the VV neovascularization; promoted smooth muscle cells recruitment and VV maturation, which maintained plaque stability; its effect was obviously superior to simvastatin. SMYA deceased the expression of HIF-1α, Apelin, APJ, Phospho-MEK1/2 (Ser217/221), Phospho-p44/42 MAPK (Erk1/2) (Thr202/Tyr204), Phospho-p70 S6 Kinase (Thr421/Ser424), Ang-2 and Tie-2; it also increased the expression of Ang-1, Phospho-Akt (Ser473), Phospho-FOXO1 (Ser256) and Survivin. Conclusions: SMYA can decrease the AS plaque area in ApoE−/− mice, suppress the VV neovascularization and promote the VV maturation, and stabilize AS vulnerable plaque. The mechanism could be regulating the HIF-1α-Apelin/APJ and Ang-1/Tie signal pathways.
Collapse
Affiliation(s)
- Meng Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhongwen Qi
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Junping Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ke Zhu
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yueyao Wang
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
47
|
Chen Q, Song H, Yu J, Kim K. Current Development and Applications of Super-Resolution Ultrasound Imaging. SENSORS 2021; 21:s21072417. [PMID: 33915779 PMCID: PMC8038018 DOI: 10.3390/s21072417] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 03/22/2021] [Accepted: 03/24/2021] [Indexed: 02/07/2023]
Abstract
Abnormal changes of the microvasculature are reported to be key evidence of the development of several critical diseases, including cancer, progressive kidney disease, and atherosclerotic plaque. Super-resolution ultrasound imaging is an emerging technology that can identify the microvasculature noninvasively, with unprecedented spatial resolution beyond the acoustic diffraction limit. Therefore, it is a promising approach for diagnosing and monitoring the development of diseases. In this review, we introduce current super-resolution ultrasound imaging approaches and their preclinical applications on different animals and disease models. Future directions and challenges to overcome for clinical translations are also discussed.
Collapse
Affiliation(s)
- Qiyang Chen
- Department of Bioengineering, School of Engineering, University of Pittsburgh, Pittsburgh, PA 15261, USA;
- Center for Ultrasound Molecular Imaging and Therapeutics, Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Hyeju Song
- Department of Robotics Engineering, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu 42988, Korea;
| | - Jaesok Yu
- Department of Robotics Engineering, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu 42988, Korea;
- DGIST Robotics Research Center, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu 42988, Korea
- Correspondence: (J.Y.); (K.K.)
| | - Kang Kim
- Department of Bioengineering, School of Engineering, University of Pittsburgh, Pittsburgh, PA 15261, USA;
- Center for Ultrasound Molecular Imaging and Therapeutics, Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Division of Cardiology, Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
- McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Mechanical Engineering and Materials Science, School of Engineering, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Correspondence: (J.Y.); (K.K.)
| |
Collapse
|
48
|
Wang X, Fu Y, Xie Z, Cao M, Qu W, Xi X, Zhong S, Piao M, Peng X, Jia Y, Meng L, Tian J. Establishment of a Novel Mouse Model for Atherosclerotic Vulnerable Plaque. Front Cardiovasc Med 2021; 8:642751. [PMID: 33796572 PMCID: PMC8007762 DOI: 10.3389/fcvm.2021.642751] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 02/23/2021] [Indexed: 12/23/2022] Open
Abstract
Background and Aims: Acute coronary syndrome (ACS) is a group of clinical syndromes characterized by rupture or erosion of atherosclerotic unstable plaques. Effective intervention for vulnerable plaques (VP) is of great significance to reduce adverse cardiovascular events. Methods: Fbn1C1039G+/− mice were crossbred with LDLR−/− mice to obtain a novel model for atherosclerotic VP. After the mice were fed with a high-fat diet (HFD) for 12 or 24 weeks, pathological staining and immunohistochemistry analyses were employed to evaluate atherosclerotic lesions. Results: Compared to control mice, Fbn1C1039G+/−LDLR−/− mice developed more severe atherosclerotic lesions, and the positive area of oil red O staining in the aortic sinus was significantly increased after 12 weeks (21.7 ± 2.0 vs. 6.3 ± 2.1) and 24 weeks (32.6 ± 2.5 vs. 18.7 ± 2.6) on a HFD. Additional vulnerable plaque characteristics, including significantly larger necrotic cores (280 ± 19 vs. 105 ± 7), thinner fiber caps (14.0 ± 2.8 vs. 32.6 ± 2.7), apparent elastin fiber fragmentation and vessel dilation (3,010 ± 67 vs. 1,465 ± 49), a 2-fold increase in macrophage number (8.5 ± 1.0 vs. 5.0 ± 0.6), obviously decreased smooth muscle cell number (0.6 ± 0.1 vs. 2.1 ± 0.2) and an ~25% decrease in total collagen content (33.6 ± 0.3 vs. 44.9 ± 9.1) were observed in Fbn1C1039G+/−LDLR−/− mice compared with control mice after 24 weeks. Furthermore, spontaneous plaque rupture, neovascularization, and intraplaque hemorrhage were detected in the model mouse plaque regions but not in those of the control mice. Conclusions: Plaques in Fbn1C1039G+/−LDLR−/− mice fed a HFD show many features of human advanced atherosclerotic unstable plaques. These results suggest that the Fbn1C1039G+/−LDLR−/− mouse is a novel model for investigating the pathological and physiological mechanisms of advanced atherosclerotic unstable plaques.
Collapse
Affiliation(s)
- Xueyu Wang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Yahong Fu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Zulong Xie
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Muhua Cao
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Wenbo Qu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Xiangwen Xi
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Shan Zhong
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Minghui Piao
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Xiang Peng
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Ying Jia
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Lingbo Meng
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jinwei Tian
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
49
|
Li L, Dmytriw AA, Krings T, Feng Y, Jiao L. Visualization of the Human Intracranial Vasa Vasorum In Vivo Using Optical Coherence Tomography. JAMA Neurol 2021; 77:903-905. [PMID: 32282013 DOI: 10.1001/jamaneurol.2020.0546] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Long Li
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Adam A Dmytriw
- Division of Neuroradiology, Toronto Western Hospital, Department of Medical Imaging, University of Toronto, Toronto, Ontario, Canada
| | - Timo Krings
- Division of Neuroradiology, Toronto Western Hospital, Department of Medical Imaging, University of Toronto, Toronto, Ontario, Canada
| | - Yiding Feng
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Liqun Jiao
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
50
|
Feng Y, Dmytriw AA, Yang B, Jiao L. Neovascularization in Human Intracranial Atherosclerotic In-Stent Restenosis. Diagnostics (Basel) 2021; 11:diagnostics11020322. [PMID: 33671117 PMCID: PMC7922829 DOI: 10.3390/diagnostics11020322] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/13/2021] [Accepted: 02/13/2021] [Indexed: 11/16/2022] Open
Abstract
Optical coherence tomography (OCT) has seen widespread use in cardiovascular and interventional endovascular imaging. While scattered reports of intracranial usage have been reported for the assessment of atherosclerotic stenosis, nutrifying neovasculature supplying plaque and neointima have not been demonstrated until now. We report the first in-vivo illustration of this phenomenon, which is a high-resolution depiction of a critical pathway for in-stent restenosis.
Collapse
Affiliation(s)
- Yiding Feng
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; (Y.F.); (B.Y.)
- Department of Interventional Neuroradiology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Adam A. Dmytriw
- Neuroradiology & Neurointervention Service, Brigham and Women’s Hospital, Boston, MA 02215, USA;
| | - Bin Yang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; (Y.F.); (B.Y.)
- Department of Interventional Neuroradiology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Liqun Jiao
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; (Y.F.); (B.Y.)
- Department of Interventional Neuroradiology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Correspondence:
| |
Collapse
|