1
|
Patil S, Kata R, Teichner E, Subtirelu R, Ghonim M, Ghonim M, Al-Daoud O, Ismoilov M, Herpin L, Ayubcha C, Werner T, Høilund-Carlsen PF, Alavi A. Associations of subclinical microcalcification and inflammation with carotid atheroma development: a dual-tracer PET/CT study. Eur J Nucl Med Mol Imaging 2025; 52:2502-2512. [PMID: 39939531 PMCID: PMC12119761 DOI: 10.1007/s00259-025-07127-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 01/29/2025] [Indexed: 02/14/2025]
Abstract
PURPOSE Carotid artery atherosclerosis, a significant manifestation of cardiovascular disease (CVD) and leading cause of stroke, develops through a gradual process of arterial inflammation and calcification. This study explores the relationship between arterial inflammation (18 F-FDG PET/CT) and vascular calcification (18 F-NaF PET/CT) in the left and right common carotid arteries (LCC/RCC) and their association with CVD and thromboembolic risk in patients with subclinical atherosclerosis. METHODS A cohort of 115 subjects (73 healthy volunteers, 42 at-risk for CVD) underwent 18 F-NaF and 18 F-FDG PET/CT imaging. Radiotracer uptake was quantitatively assessed by measuring the average blood-pool-corrected mean standardized uptake value (aSUVmean). RESULTS Relative to healthy volunteers, at-risk subjects had greater uptake of NaF and FDG (10-22% and 16-27% higher, respectively, in both arteries, p < 0.05). On multivariate regression, NaF aSUVmean correlated with age and BMI (p < 0.01), and FDG aSUVmean correlated with BMI (p ≤ 0.01), fibrinogen (p < 0.01 in LCC only), and total cholesterol (p = 0.02 in RCC only). NaF aSUVmean increased with elevated 10-year CVD risk (p = 0.003 in LCC only), while no significant trend was seen for FDG. NaF and FDG aSUVmean increased with elevated thromboembolic risk in both arteries (p < 0.05). No correlations between NaF and FDG aSUVmean were observed (p > 0.05). CONCLUSION 18 F-NaF PET/CT may serve as a prognostic tool for carotid microcalcification and subclinical atherosclerosis, while the utility of 18 F-FDG PET/CT remains uncertain. CLINICAL TRIAL REGISTRATION "Cardiovascular Molecular Calcification Assessed by 18F-NaF PET CT (CAMONA)", NCT01724749, https://clinicaltrials.gov/study/NCT01724749 .
Collapse
Affiliation(s)
- Shiv Patil
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Rithvik Kata
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Eric Teichner
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Robert Subtirelu
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Mohanad Ghonim
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Radiology, Ain Shams University, Cairo, Egypt
| | - Mohamed Ghonim
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Radiology, Ain Shams University, Cairo, Egypt
| | - Omar Al-Daoud
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Miraziz Ismoilov
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Lancelot Herpin
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | | | - Thomas Werner
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | | | - Abass Alavi
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
2
|
Tamboline M, Collins J, Jackson W, Gu W, Worssam M, Cheng P, David J, Taschereau R, Chatziioannou AF, Jackson S, Xu S, Ikotun OF. Preclinical evaluation of high-resolution CT, 18F-FDG, and 18F-NaF PET imaging for longitudinal monitoring of atherosclerosis. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07276-1. [PMID: 40289041 DOI: 10.1007/s00259-025-07276-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 04/07/2025] [Indexed: 04/29/2025]
Abstract
RATIONALE Detection of atherosclerosis is essential to the management and prevention of life-threatening cardiovascular events. Although non-invasive imaging modalities, such as 18F-sodium fluoride (18F-NaF), 18F-fluorodeoxyglucose (18F-FDG) PET, and CT, visualize distinct hallmarks of atherosclerosis, there has yet to be a singular multi-cohort interrogation of their strengths and limitations. Thus, we focused on identifying the optimal approach for visualizing atherosclerosis at different stages of disease progression. METHODS In this study, 6-week-old, male, ApoE deficient mice (Apoe-/-) were placed on a high cholesterol diet for 12-20 weeks to induce calcific atherosclerotic disease. Age-matched, male, wildtype (WT) C57BL/6 mice fed with regular chow served as the control group. Mice were imaged at 12, 15, 18, and 20 weeks after starting their respective diets. To follow the progression of calcified atherosclerotic lesions, at each time point, in vivo, 18F-NaF microPET/CT images were acquired 1 h and 3 h post tracer i.v. injection. In a separate cohort, in vivo 18F-FDG PET/CT images were acquired at 3 and 5 h post tracer i.v. injection to follow inflammation as a result of progressive atherosclerotic lesion formation. High-resolution microCT images were acquired for all mice to visualize aorta calcification. After each imaging session, a subset (n = 3) was euthanized from each group and histological analysis of the aorta was performed to confirm disease progression. RESULTS In this comparative study, within the same cohort, 18F-NaF PET detected atherosclerotic calcification earlier than microCT. At both 1 and 3 h post-injection (p.i.), calcified lesions were clearly detected by 18F-NaF with a six-fold higher signal in Apoe-/- compared to WT mice. Interestingly, 18F-NaF signal peaked at week 18, whereas aortic CT signal progressively increased with a 13-, 16-, and 29-fold at 15, 18, and 20 weeks, respectively. 18F-FDG arortic accumulation at weeks 12 and 15, were significantly greater in Apoe -/- mice than WT control when images were acquired at 5 h but not at 3 h p.i.. In contrast to histological analysis, at ≥ 16 weeks where inflammation is significantly elevated, 18F-FDG was equivalent in Apoe-/- and WT control mice and significantly reduced with disease progression. CONCLUSIONS Our results show that 18F-NaF PET and 18F-FDG PET are sensitive imaging modalities for the early detection of atherosclerotic lesions. However, both 18F-NaF PET and high-resolution microCT prove to be effective methods for monitoring late-stage and progressive disease.
Collapse
Affiliation(s)
- Mikayla Tamboline
- Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, 90025, USA
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90025, USA
| | - Jeffrey Collins
- Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, 90025, USA
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90025, USA
| | - William Jackson
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Wenduo Gu
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Matthew Worssam
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Paul Cheng
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University, Stanford, CA, 94305, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, 94305, USA
| | - John David
- Cardiovascular, Amgen Inc., Thousand Oaks, CA, 91320, USA
| | - Richard Taschereau
- Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, 90025, USA
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90025, USA
| | - Arion F Chatziioannou
- Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, 90025, USA
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90025, USA
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90025, USA
| | - Simon Jackson
- Cardiovascular, Amgen Inc., Thousand Oaks, CA, 91320, USA
| | - Shili Xu
- Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, 90025, USA.
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90025, USA.
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90025, USA.
- , Los Angeles, USA.
| | - Oluwatayo F Ikotun
- Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, 90025, USA.
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90025, USA.
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90025, USA.
- , Los Angeles, USA.
| |
Collapse
|
3
|
Tapley JK, Doyle BJ, Bellinge JW, Caddy HT, Blom DC, Churack T, Newby DE, Schultz CJ, Kelsey LJ. Low endothelial shear stress is associated with increased coronary atherosclerotic plaque activity in patients that presented with acute coronary syndrome. J Cardiovasc Comput Tomogr 2025:S1934-5925(25)00063-2. [PMID: 40280791 DOI: 10.1016/j.jcct.2025.04.003] [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: 11/29/2024] [Revised: 03/28/2025] [Accepted: 04/04/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Both coronary atherosclerotic plaque activity and low endothelial shear stress (ESS) are predictive of adverse cardiovascular events. We aimed to investigate their association and relationship with high-risk plaque features. METHODS Coronary computed tomography angiography (CCTA) based flow simulations were used to compute ESS in patients presenting with acute coronary syndrome proceeding percutaneous coronary intervention. Associations between ESS, CCTA plaque features and coronary plaque activity, measured by 18F-sodium fluoride (18F-NaF) positron emission tomography (PET), were investigated at the coronary segment and vessel level. RESULTS ESS and coronary plaque activity were both analyzed in 330 coronary segments and 123 vessels. The area of low ESS (<0.4 Pa), termed low shear area (LSA), was larger in 18F-NaF positive regions increasing from median 11.7 mm2 (IQR: 4.6-27.4) to 29.0 mm2 (IQR: 14.1-55.2) at the segment level (P < 0.0001) and from median 27.3 mm2 (IQR: 8.6-65.3) to 57.8 mm2 (26.6-108.2) at the vessel level (P = 0.0049). The maximum tissue-to-background ratio of 18F-NaF activity positively correlated with LSA at the segment level (rs = 0.27; P < 0.0001) and at the vessel level (rs = 0.38; P < 0.0001). LSA was associated with spotty calcification at both the segment (P <0.0001) and vessel level (P = 0.0042) and positive remodeling at the vessel level (P = 0.025). CONCLUSIONS In patients with acute coronary syndrome, LSA is associated with increased coronary atherosclerotic plaque activity, as measured by 18F-NaF PET.
Collapse
Affiliation(s)
- Jonathan K Tapley
- Vascular Engineering Laboratory, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and the UWA Centre for Medical Research, The University of Western Australia, Perth, Australia; Navier Medical Ltd., Perth, Australia; Royal Perth Hospital, Perth, Australia.
| | - Barry J Doyle
- Vascular Engineering Laboratory, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and the UWA Centre for Medical Research, The University of Western Australia, Perth, Australia; Navier Medical Ltd., Perth, Australia; School of Engineering, The University of Western Australia, Perth, Australia
| | - Jamie W Bellinge
- School of Medicine, The University of Western Australia, Perth, Australia; Department of Cardiology, Royal Perth Hospital, Perth, Australia; Department of Nuclear Medicine, Sir Charles Gairdner Hospital, Perth, Australia
| | - Harrison T Caddy
- Vascular Engineering Laboratory, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and the UWA Centre for Medical Research, The University of Western Australia, Perth, Australia; School of Engineering, The University of Western Australia, Perth, Australia; School of Human Sciences, The University of Western Australia, Perth, Australia
| | - Dirk C Blom
- Vascular Engineering Laboratory, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and the UWA Centre for Medical Research, The University of Western Australia, Perth, Australia; Curtin Medical School, Curtin University, Perth, Australia
| | | | - David E Newby
- British Heart Foundation Centre of Research Excellence, University of Edinburgh, Edinburgh, United Kingdom
| | - Carl J Schultz
- School of Medicine, The University of Western Australia, Perth, Australia; Department of Cardiology, Royal Perth Hospital, Perth, Australia
| | - Lachlan J Kelsey
- Vascular Engineering Laboratory, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and the UWA Centre for Medical Research, The University of Western Australia, Perth, Australia; Navier Medical Ltd., Perth, Australia; School of Engineering, The University of Western Australia, Perth, Australia
| |
Collapse
|
4
|
Rubin J, Cao Q, Sakai Y, Arnett N, Phi HQ, Hu AC, Cucchiara BL, Bos D, Saba L, Zee J, Song JW. Carotid Plaque Calcification Attenuation Characteristics are Associated with Intraplaque Hemorrhage Volumes: A 3D Segmentation-Based Analysis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.08.25325406. [PMID: 40297431 PMCID: PMC12036392 DOI: 10.1101/2025.04.08.25325406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Background and Purpose Despite the high prevalence of plaque calcifications in carotid atherosclerosis, the association between morphologic and attenuation features of calcifications and intraplaque hemorrhage (IPH) remains unclear. Methods Carotid bifurcation calcific plaques were identified on neck CTAs from patients with unilateral anterior circulation ischemic stroke consistent with embolic stroke of undetermined source. Plaque calcifications were manually segmented using 3D Slicer to measure volume, surface area, shape, and attenuation (Hounsfield Units (HU)) characteristics. IPH volume (IPHvol) was quantified using a semi-automated software. A linear mixed regression model evaluated associations between calcification features and IPHvol, adjusting for sex, age, and cardiovascular risk factors. An interaction term between calcification volume and attenuation was included after dichotomizing attenuation (>924HU) and volume (>30mm 3 ) as high versus low based on median values. Results From 70 patients (median age 68 years, 50% female), 116 calcific plaques containing 269 plaque calcifications were analyzed. Adjusting for age, cardiovascular risk factors and plaque calcification features, being female showed lower IPHvols compared to males (mean ratio 0.34, p=0.002). A significant interaction between calcification volume and attenuation emerged (p=0.042). Among plaques with low volumes (<30mm 3 ) of plaque calcifications, plaques with low-attenuation (<924HU) calcifications showed 5.53 times higher IPHvols than plaques with high-attenuation calcifications (p=0.003). Among plaques with high-attenuation calcifications (>924HU), plaques with high volumes of these calcifications showed 4.40 times higher IPHvols compared to low-volumes of high-attenuation calcifications (p=0.011). Conclusions Plaque calcification attenuation characteristics are associated with IPHvols. Beyond presence or volumes of plaque calcifications, calcification attenuation characteristics should be considered when evaluating unstable plaque components.
Collapse
|
5
|
Milovanovic P, Savic I, Popovic A, Grajic M. Ectopic calcifications in the musculoskeletal field: the basis for preventive and curative pharmacological strategies. Clin Rheumatol 2025; 44:869-886. [PMID: 39853559 DOI: 10.1007/s10067-025-07335-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 01/02/2025] [Accepted: 01/12/2025] [Indexed: 01/26/2025]
Abstract
Ectopic calcifications occur in tendons, ligaments, entheses, muscles, and fasciae, and are often associated with pain and inflammation. In clinical settings, these calcifications are commonly treated by physical therapy and/or surgical interventions. However, there is not enough understanding of pharmacological treatments as primary cures, supportive therapy to physical or surgical treatment, or even preventive measures to avoid or diminish the development of ectopic calcifications. Here, we summarize preclinical and clinical evidence for pharmacological candidates for treatment/prevention of ectopic calcification in the context of painful syndromes in the musculoskeletal field. Specifically, we discuss the potential mechanisms of nonsteroidal anti-inflammatory drugs, corticosteroids, H2-receptor blockers, bisphosphonates, minocycline, biologics, ACTH analogues, colchicine, calcium channel blockers, vitamins K2 and D, magnesium, zinc, curcumin, and phytates. Given that ectopic calcification is sometimes paradoxically associated with reduced bone mineralization, it appears particularly reasonable to employ strategies that can both inhibit ectopic calcification and promote bone mineralization, such as bisphosphonates and the combination of vitamin K2 and vitamin D, along with other supplements such as magnesium and zinc. Future studies need to test whether differential therapeutic approaches are needed in different phases of the disease and whether different mechanisms of ectopic calcification require different therapeutic strategies. A precondition for such approaches is further clinical and/or imaging delineation and differentiation of various types and phases of calcific diseases. Finally, it is essential to ensure that anti-calcification effects of new treatment strategies do not harm bone formation and skeletal mineralization.
Collapse
Affiliation(s)
- Petar Milovanovic
- Center of Bone Biology, Institute of Anatomy, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Ivana Savic
- Institute of Pathology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Ana Popovic
- Center for Physical Medicine and Rehabilitation, University Clinical Center of Serbia, Belgrade, Serbia
| | - Mirko Grajic
- Center for Physical Medicine and Rehabilitation, University Clinical Center of Serbia, Belgrade, Serbia.
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia.
| |
Collapse
|
6
|
Luo J, Wang Q, Liu W, Liao H, Qing W, Zhang M, Tang D, Luo G, Zhao H. Computed tomography provides a "one-stop-shop" targeted analysis for coronary artery calcification and osteoporosis: a review. Front Endocrinol (Lausanne) 2025; 16:1356831. [PMID: 40093749 PMCID: PMC11906312 DOI: 10.3389/fendo.2025.1356831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 02/05/2025] [Indexed: 03/19/2025] Open
Abstract
The global trend towards longer lifespans has led to an aging population and a rise in the prevalence of diseases that predominantly affect elderly people. Coronary artery calcification (CAC) and osteoporosis (OP) are common in elderly populations. CT scans provide a reliable method to assess and monitor the progression of these diseases. In this review, the relationship between OP and CAC in terms of pathophysiological mechanism, comorbidity risk factors and clinical manifestations is reviewed, with a focus on the advancements in CT imaging, clinical applications and the possibility for "one-stop-shop" for examination.
Collapse
Affiliation(s)
- Jing Luo
- Department of Radiology, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Qian Wang
- Department of Radiology, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
- Department of Radiology, Hong’an County People’s Hospital, Huanggang, Hubei, China
| | - Wenhong Liu
- Department of Radiology, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Huazhi Liao
- Department of Radiology, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Weipeng Qing
- Department of Radiology, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Minyi Zhang
- Major in Medical Imaging, The University of South China, Hengyang, Hunan, China
| | - Deqiu Tang
- Department of Radiology, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Guanghua Luo
- Department of Radiology, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Heng Zhao
- Department of Radiology, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| |
Collapse
|
7
|
Yap NAL, Khan Z, He X, Lee JG, Maung S, Morgan KR, Zhou T, Precht H, Serruys PW, Garcia-Garcia HM, Onuma Y, Hynes S, Kelle S, Mathur A, Baumbach A, Bourantas CV. What have we learnt from histology about the efficacy of coronary imaging modalities in assessing plaque composition? Front Cardiovasc Med 2025; 12:1507892. [PMID: 39925979 PMCID: PMC11802506 DOI: 10.3389/fcvm.2025.1507892] [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: 10/08/2024] [Accepted: 01/06/2025] [Indexed: 02/11/2025] Open
Abstract
Accurate evaluation of coronary artery pathology is essential for risk stratification and tailoring appropriate treatment. Intravascular imaging was introduced for this purpose 40 years ago enabling for the first time in vivo plaque characterization. Since then, several studies have evaluated the efficacy of the existing intravascular imaging modalities in assessing plaque pathology and composition and their potential in guiding intervention and predicting vulnerable plaques. Today it is known that intravascular imaging is an indispensable tool in percutaneous coronary intervention planning, but the existing modalities have a limited efficacy in predicting lesion vulnerability; a fact that should be attributed to their advantages and limitations in accurately assessing morpho-pathological features that are common in advanced atherosclerotic plaques. This review aims to provide a comprehensive evaluation of the performance of intravascular imaging in characterizing plaque phenotypes using histology as a reference standard; it summarizes the studies comparing the available invasive imaging techniques against histology, discusses the findings and limitations of these studies and highlights the potential of novel intravascular imaging approaches that were introduced for a more complete and comprehensive evaluation of plaque pathobiology.
Collapse
Affiliation(s)
- Nathan Angelo Lecaros Yap
- Device and Innovation Centre, William Harvey Research Institute Queen Mary University, London, United Kingdom
| | - Zahid Khan
- Barts Heart Centre, Barts Health NHS Trust, London, United Kingdom
- Medical Education, University of South Wales, Wales and University of Buckingham, Buckingham, United Kingdom
| | - Xingwei He
- Device and Innovation Centre, William Harvey Research Institute Queen Mary University, London, United Kingdom
| | - Jae-Geun Lee
- Device and Innovation Centre, William Harvey Research Institute Queen Mary University, London, United Kingdom
| | - Soe Maung
- Device and Innovation Centre, William Harvey Research Institute Queen Mary University, London, United Kingdom
| | - Kimberley R. Morgan
- Device and Innovation Centre, William Harvey Research Institute Queen Mary University, London, United Kingdom
| | - Tingquan Zhou
- Device and Innovation Centre, William Harvey Research Institute Queen Mary University, London, United Kingdom
| | - Helle Precht
- Conrad Research Center, Radiography Education, University College Lillebælt, Odense, Denmark
| | - Patrick W. Serruys
- Faculty of Medicine, National Heart & Lung Institute, Imperial College London, United Kingdom
- Department of Cardiology, National University of Ireland, Galway, Ireland
| | - Hector M. Garcia-Garcia
- Interventional Cardiology Department, MedStar Washington Hospital Center, Washington, DC, United States
| | - Yoshinobu Onuma
- Department of Cardiology, National University of Ireland, Galway, Ireland
| | - Sean Hynes
- Department of Pathology, National University of Ireland, Galway, Ireland
| | - Sebastian Kelle
- Department of Cardiology, Radiology and Intensive Care Medicine, Deutsches Herzzentrum Der Charite, Berlin, Germany
| | - Anthony Mathur
- Device and Innovation Centre, William Harvey Research Institute Queen Mary University, London, United Kingdom
- Barts Heart Centre, Barts Health NHS Trust, London, United Kingdom
| | - Andreas Baumbach
- Device and Innovation Centre, William Harvey Research Institute Queen Mary University, London, United Kingdom
- Barts Heart Centre, Barts Health NHS Trust, London, United Kingdom
| | - Christos V. Bourantas
- Device and Innovation Centre, William Harvey Research Institute Queen Mary University, London, United Kingdom
- Barts Heart Centre, Barts Health NHS Trust, London, United Kingdom
| |
Collapse
|
8
|
Wang W, Li Y, Zhu M, Xu Q, Cui J, Liu Y, Liu Y. Danlian-Tongmai formula improves diabetic vascular calcification by regulating CCN3/NOTCH signal axis to inhibit inflammatory reaction. Front Pharmacol 2025; 15:1510030. [PMID: 39834821 PMCID: PMC11743396 DOI: 10.3389/fphar.2024.1510030] [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: 10/12/2024] [Accepted: 12/04/2024] [Indexed: 01/22/2025] Open
Abstract
Background Vascular calcification (VC) commonly occurs in diabetes and is associated with cardiovascular disease incidence and mortality. Currently, there is no drug treatment for VC. The Danlian-Tongmai formula (DLTM) is a traditional Chinese medicine (TCM) prescription used for diabetic VC (DVC), but its mechanisms of action remain unclear. This study aims to elucidate the effects of DLTM on DVC and explore the underlying mechanisms of action. Methods Ultra-high-performance liquid chromatography-mass spectrometry (UHPLC-MS) was used to identify the metabolites of DLTM. A DVC rat model was established using streptozotocin (STZ) combined with vitamin D3 (VitD3). The effects of DLTM on DVC were evaluated through alizarin red staining, calcium deposition, and changes in osteogenic and contractile markers. The specific molecular mechanism of DLTM in treating diabetic VC was comprehensively analyzed by transcriptomics, molecular docking and in vivo experimental verification. Results We identified 108 major metabolites of DLTM. In vivo, high-dose DLTM significantly alleviated VC in diabetic rats. Transcriptomic analysis showed that DLTM treatment markedly altered the transcriptomic profile of rat aortas, which was associated with regulating the CCN3/NOTCH signaling pathway, promoting vascular smooth muscle contraction, and inhibiting the inflammatory responses. Molecular docking and molecular dynamics simulation demonstrated strong binding interactions between DLTM metabolites and key molecules within the CCN3/NOTCH pathway, including NOTCH1, DLL1, DLL4, hes1, and hey1. In vivo experiments confirmed that DLTM could upregulate CCN3, inhibit the activation of NOTCH signaling ligands DLL1 and downstream transcription factors hes1 and hey1, and reduce the release of inflammatory cytokines IL6, IL1β, and TNFα. Conclusion DLTM alleviates DVC by regulating the CCN3/NOTCH signaling axis to inhibit inflammatory responses. Our research provides experimental basis for clinical treatment and drug transformation of diabetic VC.
Collapse
Affiliation(s)
- Wenting Wang
- National Clinical Research Center for TCM Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yiwen Li
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Mengmeng Zhu
- National Clinical Research Center for TCM Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qian Xu
- National Clinical Research Center for TCM Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jing Cui
- National Clinical Research Center for TCM Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yanfei Liu
- National Clinical Research Center for TCM Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- The Second Department of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yue Liu
- National Clinical Research Center for TCM Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
9
|
Cui X, Zhang L, Lin L, Hu Y, Zhang M, Sun B, Zhang Z, Lu M, Guan X, Hao J, Li Y, Li C. Notoginsenoside R1-Protocatechuic aldehyde reduces vascular inflammation and calcification through increasing the release of nitric oxide to inhibit TGFβR1-YAP/TAZ pathway in vascular smooth muscle cells. Int Immunopharmacol 2024; 143:113574. [PMID: 39520961 DOI: 10.1016/j.intimp.2024.113574] [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: 09/19/2024] [Revised: 11/01/2024] [Accepted: 11/03/2024] [Indexed: 11/16/2024]
Abstract
Vascular calcification is a significant factor contributing to the rupture of vulnerable atherosclerotic plaques, ultimately leading to cardiovascular disease. However, no effective treatments are currently available to slow the progression of vascular calcification. Notoginsenoside R1 (R1) and protocatechuic aldehyde (PCAD), primary active components extracted from Panax notoginseng and Salvia miltiorrhiza Burge, have shown potential in mitigating endothelial injury and atherosclerosis. This study investigated the effects of R1-PCAD on nitric oxide (NO) production in endothelial cells (ECs) and its role in counteracting vascular calcification and inflammation. Additionally, it explored the mechanisms underlying these effects. To simulate atherosclerotic calcification, apolipoprotein E-deficient (ApoE-/-) mice were fed a high-fat diet and given intraperitoneal injections of vitamin D3. Treatment with the R1-PCAD combination improved endothelial function, reduced inflammation in the aorta, and lowered calcium deposition. Mechanistically, R1-PCAD enhanced eNOS-Ser1177 phosphorylation by activating the AMPKα/Akt pathway, which stimulated NO production and eNOS activation in ECs. In an in vitro co-culture model involving vascular smooth muscle cells (VSMCs) and ECs, R1-PCAD similarly reduced inflammation and calcification in VSMCs triggered by β-glycerophosphate, with these effects partially dependent on NO levels and EC functionality. Further investigation revealed that R1-PCAD facilitated NO release from ECs, which subsequently inhibited TGFβR1 activation in VSMCs. This inhibition reduced Smad2/3 activation and nuclear translocation of YAP/TAZ, thereby diminishing inflammation and calcification in VSMCs. These findings suggest that R1-PCAD alleviates vascular inflammation and calcification primarily via the NO-TGFβR1-YAP/TAZ signaling pathway. This study presents a promising new approach for treating vascular calcification by targeting intercellular signaling pathways.
Collapse
MESH Headings
- Animals
- Nitric Oxide/metabolism
- Ginsenosides/pharmacology
- Ginsenosides/therapeutic use
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/metabolism
- Mice
- Signal Transduction/drug effects
- Catechols/pharmacology
- Catechols/therapeutic use
- Benzaldehydes/pharmacology
- Benzaldehydes/therapeutic use
- Vascular Calcification/drug therapy
- Vascular Calcification/pathology
- Vascular Calcification/metabolism
- Mice, Inbred C57BL
- Male
- Receptor, Transforming Growth Factor-beta Type I/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Adaptor Proteins, Signal Transducing/metabolism
- Humans
- Transcription Factors/metabolism
- Anti-Inflammatory Agents/pharmacology
- Anti-Inflammatory Agents/therapeutic use
- Atherosclerosis/drug therapy
- Atherosclerosis/pathology
- Atherosclerosis/metabolism
- Cells, Cultured
- YAP-Signaling Proteins
Collapse
Affiliation(s)
- Xinhai Cui
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Lei Zhang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Lin Lin
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Yuanlong Hu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Muxin Zhang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Bowen Sun
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Zhiyuan Zhang
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Mengkai Lu
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Xiuya Guan
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Jiaqi Hao
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Yunlun Li
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| | - Chao Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| |
Collapse
|
10
|
Klauzen P, Basovich L, Shishkova D, Markova V, Malashicheva A. Macrophages in Calcific Aortic Valve Disease: Paracrine and Juxtacrine Disease Drivers. Biomolecules 2024; 14:1547. [PMID: 39766254 PMCID: PMC11673549 DOI: 10.3390/biom14121547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/27/2024] [Accepted: 11/29/2024] [Indexed: 01/11/2025] Open
Abstract
A significant role in the pathogenesis of CAVD is played by innate immunity cells, such as macrophages. In stenotic valves, macrophages have enhanced inflammatory activity, and the population's balance is shifted toward pro-inflammatory ones. Pro-inflammatory macrophages release cytokines, chemokines, and microRNA, which can directly affect the resident valvular cells and cause valve calcification. In CAVD patients, macrophages may have more pronounced pro-inflammatory properties, enhanced not only by paracrine signals but also by juxtacrine Notch signaling and epigenetic factors, which influence the maturation of macrophages' progenitors. In this review, we observe the accumulated data on the involvement of macrophages in CAVD development via paracrine and juxtacrine interactions.
Collapse
Affiliation(s)
- Polina Klauzen
- Laboratory of Regenerative Biomedicine, Institute of Cytology, Russian Academy of Sciences, Saint-Petersburg 194064, Russia.; (L.B.)
| | - Liubov Basovich
- Laboratory of Regenerative Biomedicine, Institute of Cytology, Russian Academy of Sciences, Saint-Petersburg 194064, Russia.; (L.B.)
| | - Daria Shishkova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo 650002, Russia; (D.S.); (V.M.)
| | - Victoria Markova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo 650002, Russia; (D.S.); (V.M.)
| | - Anna Malashicheva
- Laboratory of Regenerative Biomedicine, Institute of Cytology, Russian Academy of Sciences, Saint-Petersburg 194064, Russia.; (L.B.)
| |
Collapse
|
11
|
Behzadi P, St Hilaire C. Metabolites and metabolism in vascular calcification: links between adenosine signaling and the methionine cycle. Am J Physiol Heart Circ Physiol 2024; 327:H1361-H1375. [PMID: 39453431 PMCID: PMC11588312 DOI: 10.1152/ajpheart.00267.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 10/21/2024] [Accepted: 10/21/2024] [Indexed: 10/26/2024]
Abstract
The global population of individuals with cardiovascular disease is expanding, and a key risk factor for major adverse cardiovascular events is vascular calcification. The pathogenesis of cardiovascular calcification is complex and multifaceted, with external cues driving epigenetic, transcriptional, and metabolic changes that promote vascular calcification. This review provides an overview of some of the lesser understood molecular processes involved in vascular calcification and discusses the links between calcification pathogenesis and aspects of adenosine signaling and the methionine pathway; the latter of which salvages the essential amino acid methionine, but also provides the substrate critical for methylation, a modification that regulates the function and activity of DNA and proteins. We explore the complex and dynamic nature of osteogenic reprogramming underlying intimal atherosclerotic calcification and medial arterial calcification (MAC). Atherosclerotic calcification is more widely studied; however, emerging studies now show that MAC is a significant pathology independent from atherosclerosis. Furthermore, we emphasize metabolite and metabolic-modulating factors that influence vascular calcification pathogenesis. Although the contributions of these mechanisms are more well-define in relation to atherosclerotic intimal calcification, understanding these pathways may provide crucial mechanistic insights into MAC and inform future therapeutic approaches. Herein, we highlight the significance of adenosine and methyltransferase pathways as key regulators of vascular calcification pathogenesis.
Collapse
Affiliation(s)
- Parya Behzadi
- Division of Cardiology, Department of Medicine, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Cynthia St Hilaire
- Division of Cardiology, Department of Medicine, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| |
Collapse
|
12
|
Viegas C, Carreira J, Maia TM, Macedo AL, Matos AP, Neves J, Simes D. Gla Rich Protein (GRP) Mediates Vascular Smooth Muscle Cell (VSMC) Osteogenic Differentiation, Extracellular Vesicle (EV) Calcification Propensity, and Immunomodulatory Properties. Int J Mol Sci 2024; 25:12406. [PMID: 39596469 PMCID: PMC11594964 DOI: 10.3390/ijms252212406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 11/08/2024] [Accepted: 11/16/2024] [Indexed: 11/28/2024] Open
Abstract
Vascular calcification (VC) is a complex process involving vascular smooth muscle cell (VSMC) osteogenic differentiation, inflammation, and extracellular vesicle (EV) calcification and communication networks. Gla rich protein (GRP) is a calcification inhibitor involved in most of these processes. However, the molecular mechanism of GRP in VC and the specific characteristics, cargo, and functionality of calcifying EVs require further elucidation. Here, we use a combination of human ex vivo aortic fragments and primary vascular smooth muscle cell (VSMC) models to obtain new information on GRP function in VC and EVs released by VSMCs. We demonstrate that GRP inhibits VSMC osteogenic differentiation through downregulation of bone-related proteins and upregulation of mineralization inhibitors, with decreased mineral crystallinity in EVs deposited into the tissue extracellular matrix (ECM). EVs isolated by ultracentrifugation at 30K and 100K from the cell media (CM) and deposited in the ECM from control (CTR) and mineralizing (MM) VSMCs were biochemically, physically, and proteomically characterized. Four different EV populations were identified with shared markers commonly present in all EVs but with unique protein cargo and specific molecular profiles. Comparative proteomics identified several regulated proteins specifically loaded into MM EV populations associated with multiple processes involved in VC. Functional analysis demonstrated that 30K and 100K ECM-MM EVs with higher calcium and lower GRP levels induced macrophage inflammation. Our findings reinforce the functional relevance of GRP in multiple VC processes and suggest that ECM EVs released under calcification stress function as a new signaling axis on the calcification-inflammation cycle.
Collapse
Affiliation(s)
- Carla Viegas
- Centre of Marine Sciences (CCMAR/CIMAR LA), University of Algarve, 8005-139 Faro, Portugal; (J.C.); (D.S.)
- GenoGla Diagnostics, Centre of Marine Sciences (CCMAR), University of Algarve, 8005-139 Faro, Portugal
| | - Joana Carreira
- Centre of Marine Sciences (CCMAR/CIMAR LA), University of Algarve, 8005-139 Faro, Portugal; (J.C.); (D.S.)
| | - Teresa M. Maia
- VIB Center for Medical Biotechnology, Technologiepark-Zwijnaarde 75, 9052 Ghent, Belgium;
- Department of Biomolecular Medicine, Ghent University, Technologiepark-Zwijnaarde 75, 9052 Ghent, Belgium
- VIB Proteomics Core, 9052 Ghent, Belgium
| | - Anjos L. Macedo
- UCIBIO, Department of Chemistry, and Associate Laboratory i4HB—Institute for Health and Bioeconomy, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal;
| | - António P. Matos
- Egas Moniz Center for Interdisciplinary Research (CiiEM), Egas Moniz School of Health & Science, 2829-511 Caparica, Portugal;
| | - José Neves
- Service of Cardiothoracic Surgery, Santa Cruz Hospital, Centro Hospitalar de Lisboa Ocidental, 2790-134 Carnaxide, Portugal;
| | - Dina Simes
- Centre of Marine Sciences (CCMAR/CIMAR LA), University of Algarve, 8005-139 Faro, Portugal; (J.C.); (D.S.)
- GenoGla Diagnostics, Centre of Marine Sciences (CCMAR), University of Algarve, 8005-139 Faro, Portugal
| |
Collapse
|
13
|
Li H, Li R, Gong C, Wu Z, Jia Q. The relationship between dietary vitamin B1 intake and severe abdominal aortic calcification among the general population in the United States. Front Cardiovasc Med 2024; 11:1415151. [PMID: 39606188 PMCID: PMC11599191 DOI: 10.3389/fcvm.2024.1415151] [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: 04/10/2024] [Accepted: 10/29/2024] [Indexed: 11/29/2024] Open
Abstract
Background Vitamin B1 deficiency is closely associated with vascular system damage, but the relationship between dietary vitamin B1 intake and abdominal aortic calcification (AAC) remains unclear and warrants further investigation. Methods 2,640 participants from the National Health and Nutrition Examination Survey (NHANES) 2013-2014 were included in the study. Severe AAC was defined as Kauppila score >5. Multivariable logistic regression analysis and restricted cubic splines (RCS) were used to examine the relationship between dietary vitamin B1 and severe AAC. Results The increase in dietary intake of vitamin B1 is significantly correlated with a decrease in the risk of severe AAC (OR: 0.601, 95% CI: 0.406, 0.892). Compared to the first quartile of dietary vitamin B1 intake, the fourth quartile had a significantly reduced risk of severe AAC (OR: 0.358, 95% CI: 0.172, 0.744). RCS indicated a decreasing trend in the risk of severe AAC with increasing dietary vitamin B1 intake. Conclusion Our research findings indicate that the increase in dietary intake of vitamin B1 is significantly associated with a decrease in the risk of severe AAC. Thus, increasing dietary vitamin B1 intake appropriately may reduce the risk of severe AAC.
Collapse
Affiliation(s)
- Hanbo Li
- Department of Vascular Surgery, The First Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ruihua Li
- Department of General Surgery, Vascular Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Changle Gong
- The First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhe Wu
- The First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qiang Jia
- Institute of Pharmaceutical Research, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
14
|
Kinoshita D, Suzuki K, Fujimoto D, Niida T, Usui E, Minami Y, Dey D, Lee H, McNulty I, Ako J, Ferencik M, Kakuta T, Jang IK. Relationship between plaque burden and plaque vulnerability: Acute coronary syndromes versus chronic coronary syndrome. J Cardiovasc Comput Tomogr 2024; 18:559-566. [PMID: 39278792 DOI: 10.1016/j.jcct.2024.09.002] [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: 07/04/2024] [Revised: 08/25/2024] [Accepted: 09/06/2024] [Indexed: 09/18/2024]
Abstract
BACKGROUND The relationship between plaque burden and microscopic characterization of plaque features as it pertains to clinical presentation has not been fully investigated. The aim of this study was to compare the relationship between plaque burden and plaque vulnerability in patients with acute coronary syndromes (ACS) versus chronic coronary syndrome (CCS). METHODS Patients who underwent both coronary computed tomography angiography (CTA) and optical coherence tomography (OCT) before coronary intervention were enrolled. All plaques were detected in culprit vessels using CTA, and total plaque volume (TPV) and OCT features were assessed at the corresponding sites. All plaques were divided into three groups according to the tertile levels of TPV (low TPV: <96.5 mm3, moderate TPV: 96.5-164.7 mm3, high TPV: ≥164.8 mm3). RESULTS A total of 990 plaques were imaged by OCT in 419 patients: 445 plaques in 190 (45.3%) patients with ACS and 545 in 229 (54.7%) with CCS. Macrophage was more prevalent in plaques with greater TPV in patients who presented with ACS but not in those who presented with CCS (low vs. moderate vs. high TPV group: macrophage 57.4% vs. 71.8% vs. 82.4% in ACS; 63.4% vs. 67.8% vs. 66.7% in CCS; interaction P = 0.004). Lipid arc increased as TPV increased, especially in patients who presented with ACS. Conversely, the layer index increased as TPV increased in patients with CCS. CONCLUSION Greater plaque burden was closely related to higher levels of plaque vulnerability in ACS and greater volume of layered plaque in CCS. TRIAL REGISTRATION clinicaltrials.gov Identifier: NCT04523194.
Collapse
Affiliation(s)
- Daisuke Kinoshita
- Gill Gray Research Laboratory, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Keishi Suzuki
- Gill Gray Research Laboratory, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Daichi Fujimoto
- Gill Gray Research Laboratory, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Takayuki Niida
- Gill Gray Research Laboratory, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Eisuke Usui
- Department of Cardiovascular Medicine, Tsuchiura Kyodo General Hospital, Tsuchiura, Ibaraki, Japan
| | - Yoshiyasu Minami
- Department of Cardiovascular Medicine, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Damini Dey
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Hang Lee
- Biostatistics Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Iris McNulty
- Gill Gray Research Laboratory, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Junya Ako
- Department of Cardiovascular Medicine, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Maros Ferencik
- Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR, USA
| | - Tsunekazu Kakuta
- Department of Cardiovascular Medicine, Tsuchiura Kyodo General Hospital, Tsuchiura, Ibaraki, Japan.
| | - Ik-Kyung Jang
- Gill Gray Research Laboratory, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
15
|
Kim KA, Jung HO, Kim MJ, Lee SY, Ahn Y, Jung MH, Chung WB, Lee DH, Youn HJ, Chang HJ. Higher serum phosphate within the normal range is associated with the development of calcified aortic valve disease. Front Cardiovasc Med 2024; 11:1450757. [PMID: 39399509 PMCID: PMC11467965 DOI: 10.3389/fcvm.2024.1450757] [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: 06/18/2024] [Accepted: 09/12/2024] [Indexed: 10/15/2024] Open
Abstract
Background Despite the essential role of ectopic osteogenic calcium-phosphate metabolism in the development of calcific aortic valve disease (CAVD), the implications of high serum phosphate levels in CAVD development are not fully understood. Methods Asymptomatic individuals who underwent health screening using serial cardiac computed tomography (CT) and echocardiography were selected from a multicenter registry. CAVD was identified and quantified on CT images using the aortic valve calcification (AVC) score. The associations between initial serum phosphate levels and the presence of baseline CAVD, development of new CAVD, and the AVC score progression rate were investigated using multivariable regression models. Results A total of 736 individuals were selected for analysis, and the median interscan duration was 36.4 months. On initial CT, 83 (13.7%) participants had baseline CAVD, while 52 (7.0%) individuals developed new CAVD during follow-up. Serum phosphate levels were not associated with a higher probability of baseline CAVD but were predictive of newly developed CAVD (odds ratio per 1 mg/dl, 1.05; 95% confidence interval, 1.01-1.10; p = 0.02). Higher phosphate levels were also associated with a faster AVC score progression in those with baseline CAVD (regression coefficient per 1 mg/dl, 15.55 Agatston units/year; 95% confidence interval, 6.02-25.07; p < 0.01), an association which remained significant when the analysis was extended to include newly developed CAVD. Conclusion Even slight elevations in serum phosphate are associated with accelerated CAVD progression from an early stage. Further studies are needed to investigate whether the regulation of phosphate metabolism can slow the progression of CAVD to aortic stenosis.
Collapse
Affiliation(s)
- Kyung An Kim
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul, Republic of Korea
- Division of Cardiology, Department of Internal Medicine, Incheon St. Mary’s Hospital, The Catholic University of Korea, Incheon, Republic of Korea
- Catholic Research Institute for Intractable Cardiovascular Disease, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hae-Ok Jung
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Mi-Jeong Kim
- Division of Cardiology, Department of Internal Medicine, Incheon St. Mary’s Hospital, The Catholic University of Korea, Incheon, Republic of Korea
- Catholic Research Institute for Intractable Cardiovascular Disease, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - So-Young Lee
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul, Republic of Korea
- Catholic Research Institute for Intractable Cardiovascular Disease, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yuran Ahn
- Catholic Research Institute for Intractable Cardiovascular Disease, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Division of Cardiology, Department of Internal Medicine, Uijeongbu St. Mary’s Hospital, The Catholic University of Korea, Uijeongbu, Republic of Korea
| | - Mi-Hyang Jung
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul, Republic of Korea
- Catholic Research Institute for Intractable Cardiovascular Disease, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Woo-Baek Chung
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Dong-Hyeon Lee
- Health Promotion Center, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Ho-Joong Youn
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hyuk-Jae Chang
- Division of Cardiology, Yonsei Cardiovascular Center, Yonsei University Health System, Seoul, Republic of Korea
| |
Collapse
|
16
|
Tóth A, Balogh E, Jeney V. In Vitro Models of Cardiovascular Calcification. Biomedicines 2024; 12:2155. [PMID: 39335668 PMCID: PMC11429067 DOI: 10.3390/biomedicines12092155] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 09/15/2024] [Accepted: 09/21/2024] [Indexed: 09/30/2024] Open
Abstract
Cardiovascular calcification, characterized by hydroxyapatite deposition in the arterial wall and heart valves, is associated with high cardiovascular morbidity and mortality. Cardiovascular calcification is a hallmark of aging but is frequently seen in association with chronic diseases, such as chronic kidney disease (CKD), diabetes, dyslipidemia, and hypertension in the younger population as well. Currently, there is no therapeutic approach to prevent or cure cardiovascular calcification. The pathophysiology of cardiovascular calcification is highly complex and involves osteogenic differentiation of various cell types of the cardiovascular system, such as vascular smooth muscle cells and valve interstitial cells. In vitro cellular and ex vivo tissue culture models are simple and useful tools in cardiovascular calcification research. These models contributed largely to the discoveries of the numerous calcification inducers, inhibitors, and molecular mechanisms. In this review, we provide an overview of the in vitro cell culture and the ex vivo tissue culture models applied in the research of cardiovascular calcification.
Collapse
Affiliation(s)
- Andrea Tóth
- MTA-DE Lendület Vascular Pathophysiology Research Group, Research Centre for Molecular Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Enikő Balogh
- MTA-DE Lendület Vascular Pathophysiology Research Group, Research Centre for Molecular Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Viktória Jeney
- MTA-DE Lendület Vascular Pathophysiology Research Group, Research Centre for Molecular Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| |
Collapse
|
17
|
Gao M, Wen W, Li H, Zheng Y, Yun M, Meng J, Wang S, Wang B, Hu B, Mou T, Yu Y, Zhang X, Li X. Coronary sodium [ 18F]fluoride activity predicts outcomes post-CABG: a comparative evaluation with conventional metrics. Eur J Nucl Med Mol Imaging 2024; 51:3235-3251. [PMID: 38730084 DOI: 10.1007/s00259-024-06736-4] [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: 01/11/2024] [Accepted: 04/26/2024] [Indexed: 05/12/2024]
Abstract
PURPOSE The value of preoperative multidisciplinary approach remains inadequately delineated in forecasting postoperative outcomes of patients undergoing coronary artery bypass grafting (CABG). Herein, we aimed to ascertain the efficacy of multi-modality cardiac imaging in predicting post-CABG cardiovascular outcomes. METHODS Patients with triple coronary artery disease underwent cardiac sodium [18F]fluoride ([18F]NaF) positron emission tomography/computed tomography (PET/CT), coronary angiography, and CT-based coronary artery calcium scoring before CABG. The maximum coronary [18F]NaF activity (target-to-blood ratio [TBR]max) and the global coronary [18F]NaF activity (TBRglobal) was determined. The primary endpoint was perioperative myocardial infarction (PMI) within 7-day post-CABG. Secondary endpoint included major adverse cardiac and cerebrovascular events (MACCEs) and recurrent angina. RESULTS This prospective observational study examined 101 patients for a median of 40 months (interquartile range: 19-47 months). Both TBRmax (odds ratio [OR] = 1.445; p = 0.011) and TBRglobal (OR = 1.797; P = 0.018) were significant predictors of PMI. TBRmax>3.0 (area under the curve [AUC], 0.65; sensitivity, 75.0%; specificity, 56.8%; p = 0.036) increased PMI risk by 3.661-fold, independent of external confounders. Kaplan-Meier test revealed a decrease in MACCE survival rate concomitant with an escalating TBRmax. TBRmax>3.6 (AUC, 0.70; sensitivity, 76.9%; specificity, 73.9%; p = 0.017) increased MACCEs risk by 5.520-fold. Both TBRmax (hazard ratio [HR], 1.298; p = 0.004) and TBRglobal (HR = 1.335; p = 0.011) were significantly correlated with recurrent angina. No significant associations were found between CAC and SYNTAX scores and between PMI occurrence and long-term MACCEs. CONCLUSION Quantification of coronary microcalcification activity via [18F]NaF PET displayed a strong ability to predict early and long-term post-CABG cardiovascular outcomes, thereby outperforming conventional metrics of coronary macrocalcification burden and stenosis severity. TRIAL REGISTRATION The trial was registered with the Chinese Clinical Trial Committee (number: ChiCTR1900022527; URL: www.chictr.org.cn/showproj.html?proj=37933 ).
Collapse
Affiliation(s)
- Mingxin Gao
- Department of Nuclear Medicine, Molecular Imaging Lab, Beijing Anzhen Hospital, Capital Medical University, No.2, Anzhen Road, Chaoyang District, Beijing, 10029, China
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, No. 2, Anzhen Road, Chaoyang District, Beijing, 100029, China
| | - Wanwan Wen
- Department of Nuclear Medicine, Molecular Imaging Lab, Beijing Anzhen Hospital, Capital Medical University, No.2, Anzhen Road, Chaoyang District, Beijing, 10029, China
| | - Haiyang Li
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, No. 2, Anzhen Road, Chaoyang District, Beijing, 100029, China
| | - Yaqi Zheng
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, No. 2, Anzhen Road, Chaoyang District, Beijing, 100029, China
| | - Mingkai Yun
- Department of Nuclear Medicine, Molecular Imaging Lab, Beijing Anzhen Hospital, Capital Medical University, No.2, Anzhen Road, Chaoyang District, Beijing, 10029, China
| | - Jingjing Meng
- Department of Nuclear Medicine, Molecular Imaging Lab, Beijing Anzhen Hospital, Capital Medical University, No.2, Anzhen Road, Chaoyang District, Beijing, 10029, China
| | - Shipan Wang
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, No. 2, Anzhen Road, Chaoyang District, Beijing, 100029, China
| | - Bolin Wang
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, No. 2, Anzhen Road, Chaoyang District, Beijing, 100029, China
| | - Biao Hu
- Department of Nuclear Medicine, Molecular Imaging Lab, Beijing Anzhen Hospital, Capital Medical University, No.2, Anzhen Road, Chaoyang District, Beijing, 10029, China
| | - Tiantian Mou
- Department of Nuclear Medicine, Molecular Imaging Lab, Beijing Anzhen Hospital, Capital Medical University, No.2, Anzhen Road, Chaoyang District, Beijing, 10029, China
| | - Yang Yu
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, No. 2, Anzhen Road, Chaoyang District, Beijing, 100029, China.
| | - Xiaoli Zhang
- Department of Nuclear Medicine, Molecular Imaging Lab, Beijing Anzhen Hospital, Capital Medical University, No.2, Anzhen Road, Chaoyang District, Beijing, 10029, China.
| | - Xiang Li
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Vienna General Hospital, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
18
|
Miao S, Yu F, Sheng R, Zhang X, Li Y, Qi Y, Lu S, Ji P, Fan J, Zhang X, Xu T, Wang Z, Liu Y, Yang G. Radiomics of pericoronary adipose tissue on computed tomography angiography predicts coronary heart disease in patients with type 2 diabetes mellitus. BMC Cardiovasc Disord 2024; 24:300. [PMID: 38867152 PMCID: PMC11167783 DOI: 10.1186/s12872-024-03970-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 06/04/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND Diabetes is a common chronic metabolic disease. The progression of the disease promotes vascular inflammation and the formation of atherosclerosis, leading to cardiovascular disease. The coronary artery perivascular adipose tissue attenuation index based on CCTA is a new noninvasive imaging biomarker that reflects the spatial changes in perivascular adipose tissue attenuation in CCTA images and the inflammation around the coronary arteries. In this study, a radiomics approach is proposed to extract a large number of image features from CCTA in a high-throughput manner and combined with clinical diagnostic data to explore the predictive ability of vascular perivascular adipose imaging data based on CCTA for coronary heart disease in diabetic patients. METHODS R language was used for statistical analysis to screen the variables with significant differences. A presegmentation model was used for CCTA vessel segmentation, and the pericoronary adipose region was screened out. PyRadiomics was used to calculate the radiomics features of pericoronary adipose tissue, and SVM, DT and RF were used to model and analyze the clinical data and radiomics data. Model performance was evaluated using indicators such as PPV, FPR, AAC, and ROC. RESULTS The results indicate that there are significant differences in age, blood pressure, and some biochemical indicators between diabetes patients with and without coronary heart disease. Among 1037 calculated radiomic parameters, 18.3% showed significant differences in imaging omics features. Three modeling methods were used to analyze different combinations of clinical information, internal vascular radiomics information and pericoronary vascular fat radiomics information. The results showed that the dataset of full data had the highest ACC values under different machine learning models. The support vector machine method showed the best specificity, sensitivity, and accuracy for this dataset. CONCLUSIONS In this study, the clinical data and pericoronary radiomics data of CCTA were fused to predict the occurrence of coronary heart disease in diabetic patients. This provides information for the early detection of coronary heart disease in patients with diabetes and allows for timely intervention and treatment.
Collapse
Affiliation(s)
- Shumei Miao
- School of Computer Science and Engineering, Southeast University, Sipailou 2, Nanjing, 210096, Jiangsu, China
- Department of Information, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Medical Informatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Feihong Yu
- Department of Ultrasonic Department, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Rongrong Sheng
- Department of Information, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaoliang Zhang
- Department of Information, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Medical Informatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yong Li
- Department of Cardiovascular Medicine Department, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yaolei Qi
- School of Computer Science and Engineering, Southeast University, Sipailou 2, Nanjing, 210096, Jiangsu, China
| | - Shan Lu
- Department of Nutritional Department, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Pei Ji
- Department of Information, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jiyue Fan
- Department of Information, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xin Zhang
- Department of Information, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Medical Informatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Tingyu Xu
- Department of Information, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Medical Informatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhongmin Wang
- Department of Information, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Medical Informatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yun Liu
- Department of Geriatrics endocrinology, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Rd 300, Nanjing, 210096, Jiangsu, China.
| | - Guanyu Yang
- School of Computer Science and Engineering, Southeast University, Sipailou 2, Nanjing, 210096, Jiangsu, China.
| |
Collapse
|
19
|
Fayad ZA, Robson PM, Fuster V. Rethinking Heart Attack Prevention: The Myth of the "Vulnerable Plaque" and Reality of Patient Risk. J Am Coll Cardiol 2024; 83:2145-2147. [PMID: 38811092 DOI: 10.1016/j.jacc.2024.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 04/10/2024] [Indexed: 05/31/2024]
Affiliation(s)
- Zahi A Fayad
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA.
| | - Philip M Robson
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Valentin Fuster
- Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| |
Collapse
|
20
|
Çakan F, Sunal AS, Adar A, Onalan O. Aortic Arch Calcification Observed on Chest X-Ray May Serve as an Independent Predictor for Recurrent Stroke. Arq Bras Cardiol 2024; 121:e20230805. [PMID: 39082580 PMCID: PMC12080711 DOI: 10.36660/abc.20230805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 03/12/2024] [Accepted: 04/03/2024] [Indexed: 05/17/2025] Open
Abstract
BACKGROUND Despite advances in diagnostic and treatment modalities, there is a need for predictive markers for recurrent strokes. OBJECTIVES This study aimed to investigate the relationship between aortic arch calcification (AAC) and stroke recurrence in stroke patients during a one-year follow-up. METHODS All stroke patients who experienced their first event were evaluated for participation in the study. Patients who experienced recurrent strokes during the one-year follow-up were recorded. AAC was assessed by chest radiography. Based on the occurrence of recurrent strokes the patients were divided into two groups. AAC was classified into four categories according to its degree, and the presence of AAC was included in the statistical analysis. The relationship between AAC and recurrent stroke was assessed using a receiver operating characteristic curve. A significance level of <0.05 was deemed acceptable for all statistical analyses. RESULTS A total of 203 patients were included in the study (46.8% female, mean age 69±12.3). Recurrent stroke was detected in 49 patients. AAC, hypertension, and atrial fibrillation were more frequent in patients with recurrent stroke. Patients with recurrent stroke had a lower glomerular filtration rate and a higher red cell distribution width (RDW). In multivariate regression analysis, AAC (hazard ratio [HR], 3.544; 95% CI:1.653-7.598, p=0.001) and RDW (HR,1.214; 95% CI:1.053-1.400, p=0.008) were identified as independent predictors of recurrent stroke. CONCLUSION The presence of AAC (≥ grade 1) and RDW were found to be significantly associated with the development of recurrent stroke within one year. These findings may have prognostic significance in the follow-up of stroke patients.
Collapse
Affiliation(s)
- Fahri Çakan
- Çerkezköy State HospitalTekirdağTurquiaÇerkezköy State Hospital, Tekirdağ – Turquia
| | - Asli Sert Sunal
- Çerkezköy State HospitalTekirdağTurquiaÇerkezköy State Hospital, Tekirdağ – Turquia
| | - Adem Adar
- Baskent University Faculty of MedicineAlanya Application And Research CenterAntalyaTurquiaBaskent University Faculty of Medicine – Alanya Application And Research Center, Antalya – Turquia
| | - Orhan Onalan
- Karabuk University Faculty of MedicineKarabukTurquiaKarabuk University Faculty of Medicine, Karabuk – Turquia
| |
Collapse
|
21
|
Elsaygh J, Zaher A, Parikh MA, Frishman WH, Peterson SJ. Nanotechnology: The Future for Diagnostic and Therapeutic Intervention in Cardiovascular Diseases is Here. Cardiol Rev 2024:00045415-990000000-00281. [PMID: 38814069 DOI: 10.1097/crd.0000000000000727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
With advances in technology and medicine over the last 3 decades, cardiovascular medicine has evolved tremendously. Nanotechnology provides a promising future in personalized precision medicine. In this review, we delve into the current and prospective applications of nanotechnology and nanoparticles in cardiology. Nanotechnology has allowed for point-of-care testing such as high-sensitivity troponins, as well as more precise cardiac imaging. This review is focused on 3 diseases within cardiology: coronary artery disease, heart failure, and valvular heart disease. The use of nanoparticles in coronary stents has shown success in preventing in-stent thrombosis, as well as using nanosized drug delivery medications to prevent neointimal proliferation in a way that spares systemic toxicity. In addition, by using nanoparticles as drug delivery systems, nanotechnology can be utilized in the delivery of goal-directed medical therapy in heart failure patients. It has also been shown to improve cell therapy in this patient population by helping in cell retention of grafts. Finally, the use of nanoparticles in the manufacturing of bioprosthetic valves provides a promising future for the longevity and success of cardiac valve repair and replacement.
Collapse
Affiliation(s)
- Jude Elsaygh
- From the Department of Medicine, New York Presbyterian Brooklyn Methodist Hospital, Brooklyn, NY
| | - Anas Zaher
- From the Department of Medicine, New York Presbyterian Brooklyn Methodist Hospital, Brooklyn, NY
| | - Manish A Parikh
- From the Department of Medicine, New York Presbyterian Brooklyn Methodist Hospital, Brooklyn, NY
- Weill Department of Medicine, Weill Cornell Medicine, New York, NY
| | | | - Stephen J Peterson
- From the Department of Medicine, New York Presbyterian Brooklyn Methodist Hospital, Brooklyn, NY
- Weill Department of Medicine, Weill Cornell Medicine, New York, NY
| |
Collapse
|
22
|
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
|
23
|
Liu C, Zhang H, Yang Y, Cao Y, Liang D. The association between vitamin C intake and the risk of abdominal aortic calcification: A population-based study. Clin Nutr ESPEN 2024; 60:254-260. [PMID: 38479919 DOI: 10.1016/j.clnesp.2024.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 01/07/2024] [Accepted: 02/12/2024] [Indexed: 04/13/2024]
Abstract
BACKGROUND The beneficial effects of dietary vitamin C intake on human health have received widespread attention from the population. However, the correlation between vitamin C intake and abdominal aortic calcification remains unclear. The authors aimed to investigate the relationship between dietary vitamin C intake and AAC in US adults. METHODS Our data for this study were obtained from the National Health and Nutrition Examination Survey (NHANES) 2013-2014, and participants had complete data on dietary vitamin C intake and AAC scores. We used weighted multivariate linear regression and multivariate logistic regression analyses to explore the independent relationship between vitamin C intake and AAC scores, along with subgroup analyses and restricted cubic splines. RESULTS A total of 2876 participants were enrolled in this study, with a mean AAC score of 1.47 ± 0.14 and a prevalence of severe AAC of 8.12%. We observed a 0.5 unit decrease in AAC scores in participants in the highest quartile compared to those in the lowest quartile of VitC intake. In contrast, there was no significant correlation between VitC intake and risk of severe AAC. Besides, subgroup analysis and interaction tests showed that there was no dependence of the association between VitC intake and AAC. CONCLUSION Dietary VitC intake was associated with reduced AAC scores, but there was no significant correlation between dietary VitC intake and risk of severe AAC.
Collapse
Affiliation(s)
- Chang Liu
- School of Medicine, Nankai University, Tianjin, China
| | - Hao Zhang
- The Eighth Medical Center, Chinese PLA (People's Liberation Army) General Hospital, Beijing, China
| | - Yuwei Yang
- The Eighth Medical Center, Chinese PLA (People's Liberation Army) General Hospital, Beijing, China
| | - Yan Cao
- The Eighth Medical Center, Chinese PLA (People's Liberation Army) General Hospital, Beijing, China
| | - Dan Liang
- Department of Endocrine, People's Hospital of Chongqing Liangjiang New Area, Chongqing, China.
| |
Collapse
|
24
|
Ben Dhaou C, Scott ML, Orr AW. Advances in Understanding Cardiovascular Disease Pathogenesis through Next-Generation Technologies. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:476-481. [PMID: 38519246 PMCID: PMC10988757 DOI: 10.1016/j.ajpath.2024.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 01/19/2024] [Accepted: 01/23/2024] [Indexed: 03/24/2024]
Affiliation(s)
- Cyrine Ben Dhaou
- Department of Pathology and Translational Pathobiology, LSU Health Shreveport, Shreveport, Louisiana
| | - Matthew L Scott
- Department of Pathology and Translational Pathobiology, LSU Health Shreveport, Shreveport, Louisiana
| | - A Wayne Orr
- Department of Pathology and Translational Pathobiology, LSU Health Shreveport, Shreveport, Louisiana; Department of Molecular and Cellular Physiology, LSU Health Shreveport, Shreveport, Louisiana; Department of Cell Biology and Anatomy, LSU Health Shreveport, Shreveport, Louisiana.
| |
Collapse
|
25
|
Jansen I, Cahalane R, Hengst R, Akyildiz A, Farrell E, Gijsen F, Aikawa E, van der Heiden K, Wissing T. The interplay of collagen, macrophages, and microcalcification in atherosclerotic plaque cap rupture mechanics. Basic Res Cardiol 2024; 119:193-213. [PMID: 38329498 PMCID: PMC11008085 DOI: 10.1007/s00395-024-01033-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 01/17/2024] [Accepted: 01/19/2024] [Indexed: 02/09/2024]
Abstract
The rupture of an atherosclerotic plaque cap overlying a lipid pool and/or necrotic core can lead to thrombotic cardiovascular events. In essence, the rupture of the plaque cap is a mechanical event, which occurs when the local stress exceeds the local tissue strength. However, due to inter- and intra-cap heterogeneity, the resulting ultimate cap strength varies, causing proper assessment of the plaque at risk of rupture to be lacking. Important players involved in tissue strength include the load-bearing collagenous matrix, macrophages, as major promoters of extracellular matrix degradation, and microcalcifications, deposits that can exacerbate local stress, increasing tissue propensity for rupture. This review summarizes the role of these components individually in tissue mechanics, along with the interplay between them. We argue that to be able to improve risk assessment, a better understanding of the effect of these individual components, as well as their reciprocal relationships on cap mechanics, is required. Finally, we discuss potential future steps, including a holistic multidisciplinary approach, multifactorial 3D in vitro model systems, and advancements in imaging techniques. The obtained knowledge will ultimately serve as input to help diagnose, prevent, and treat atherosclerotic cap rupture.
Collapse
Affiliation(s)
- Imke Jansen
- Department of Biomedical Engineering, Thorax Center Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Rachel Cahalane
- Mechanobiology and Medical Device Research Group (MMDRG), Biomedical Engineering, College of Science and Engineering, University of Galway, Galway, Ireland
- Division of Cardiovascular Medicine, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ranmadusha Hengst
- Department of Biomedical Engineering, Thorax Center Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Ali Akyildiz
- Department of Biomedical Engineering, Thorax Center Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Biomechanical Engineering, Technical University Delft, Delft, The Netherlands
| | - Eric Farrell
- Department of Oral and Maxillofacial Surgery, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Frank Gijsen
- Department of Biomedical Engineering, Thorax Center Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Biomechanical Engineering, Technical University Delft, Delft, The Netherlands
| | - Elena Aikawa
- Division of Cardiovascular Medicine, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kim van der Heiden
- Department of Biomedical Engineering, Thorax Center Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Tamar Wissing
- Department of Biomedical Engineering, Thorax Center Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.
| |
Collapse
|
26
|
Perez KA, Deppe DW, Filas A, Singh SA, Aikawa E. Multimodal Analytical Tools to Enhance Mechanistic Understanding of Aortic Valve Calcification. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:539-550. [PMID: 37517686 PMCID: PMC10988764 DOI: 10.1016/j.ajpath.2023.06.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 06/14/2023] [Accepted: 06/29/2023] [Indexed: 08/01/2023]
Abstract
This review focuses on technologies at the core of calcific aortic valve disease (CAVD) and drug target research advancement, including transcriptomics, proteomics, and molecular imaging. We examine how bulk RNA sequencing and single-cell RNA sequencing have engendered organismal genomes and transcriptomes, promoting the analysis of tissue gene expression profiles and cell subpopulations, respectively. We bring into focus how the field is also largely influenced by increasingly accessible proteome profiling techniques. In unison, global transcriptional and protein expression analyses allow for increased understanding of cellular behavior and pathogenic pathways under pathologic stimuli including stress, inflammation, low-density lipoprotein accumulation, increased calcium and phosphate levels, and vascular injury. We also look at how direct investigation of protein signatures paves the way for identification of targetable pathways for pharmacologic intervention. Here, we note that imaging techniques, once a clinical diagnostic tool for late-stage CAVD, have since been refined to address a clinical need to identify microcalcifications using positron emission tomography/computed tomography and even detect in vivo cellular events indicative of early stage CAVD and map the expression of identified proteins in animal models. Together, these techniques generate a holistic approach to CAVD investigation, with the potential to identify additional novel regulatory pathways.
Collapse
Affiliation(s)
- Katelyn A Perez
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Daniel W Deppe
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Aidan Filas
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Sasha A Singh
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
27
|
Luo Y, He F, Zhang Y, Li S, Lu R, Wei X, Huang J. Transcription Factor 21: A Transcription Factor That Plays an Important Role in Cardiovascular Disease. Pharmacology 2024; 109:183-193. [PMID: 38493769 DOI: 10.1159/000536585] [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: 12/05/2023] [Accepted: 01/18/2024] [Indexed: 03/19/2024]
Abstract
BACKGROUND According to the World Health Organisation's Health Report 2019, approximately 17.18 million people die from cardiovascular disease each year, accounting for more than 30% of all global deaths. Therefore, the occurrence of cardiovascular disease is still a global concern. The transcription factor 21 (TCF21) plays an important role in cardiovascular diseases. This article reviews the regulation mechanism of TCF21 expression and activity and focuses on its important role in atherosclerosis in order to contribute to the development of diagnosis and treatment of cardiovascular diseases. SUMMARY TCF21 is involved in the phenotypic regulation of vascular smooth muscle cells (VSMCs), promotes the proliferation and migration of VSMCs, and participates in the activation of inflammatory sequences. Increased proliferation and migration of VSMCs can lead to neointimal hyperplasia after vascular injury. Abnormal hyperplasia of neointima and inflammation are one of the main features of atherosclerosis. Therefore, targeting TCF21 may become a potential treatment for relieving atherosclerosis. KEY MESSAGES TCF21 as a member of basic helix-loop-helix transcription factors regulates cell growth and differentiation by modulating gene expression during the development of different organs and plays an important role in cardiovascular development and disease. VSMCs and cells derived from VSMCs constitute the majority of plaques in atherosclerosis. TCF21 plays a key role in regulation of VSMCs' phenotype, thus accelerating atherogenesis in the early stage. However, TCF21 enhances plaque stability in late-stage atherosclerosis. The dual role of TCF21 should be considered in the translational medicine.
Collapse
Affiliation(s)
- Yaqian Luo
- Department of Pathophysiology, Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Hengyang Medical School, University of South China, Hengyang, China,
| | - Fangzhou He
- Department of Anaesthesia, Chuanshan College, University of South China, Hengyang, China
| | - Yifang Zhang
- Department of Pathophysiology, Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Hengyang Medical School, University of South China, Hengyang, China
| | - Shufan Li
- Department of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, China
| | - Ruirui Lu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, China
| | - Xing Wei
- Department of Pathophysiology, Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Hengyang Medical School, University of South China, Hengyang, China
| | - Ji Huang
- Department of Pathophysiology, Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
28
|
Jo Y, Majumdar U, Bose S. Vitamin D3 Release from MgO Doped 3D Printed TCP Scaffolds for Bone Regeneration. ACS Biomater Sci Eng 2024; 10:1676-1685. [PMID: 38386843 PMCID: PMC11186521 DOI: 10.1021/acsbiomaterials.3c01779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Abstract
Regenerating bone tissue in critical-sized craniofacial bone defects remains challenging and requires the implementation of innovative bone implants with early stage osteogenesis and blood vessel formation. Vitamin D3 is incorporated into MgO-doped 3D-printed scaffolds for defect-specific and patient-specific implants in low load-bearing areas. This novel bone implant also promotes early stage osteogenesis and blood vessel development. Our results show that vitamin D3-loaded MgO-doped 3D-printed scaffolds enhance osteoblast cell proliferation 1.3-fold after being cultured for 7 days. Coculture studies on osteoblasts derived from human mesenchymal stem cells (hMSCs) and osteoclasts derived from monocytes show the upregulation of genes related to osteoblastogenesis and the downregulation of RANK-L, which is essential for osteoclastogenesis. Release of vitamin D3 also inhibits osteoclast differentiation by 1.9-fold after a 21-day culture. After 6 weeks, vitamin D3 release from MgO-doped 3D-printed scaffolds enhances the new bone formation, mineralization, and angiogenic potential. The multifunctional 3D-printed scaffolds can improve early stage osteogenesis and blood vessel formation in craniofacial bone defects.
Collapse
Affiliation(s)
- Yongdeok Jo
- W. M. Keck Biomedical Materials Research Laboratory, School of Mechanical and Materials Engineering, Washington State University, Pullman, Washington 99164, United States
| | - Ujjayan Majumdar
- W. M. Keck Biomedical Materials Research Laboratory, School of Mechanical and Materials Engineering, Washington State University, Pullman, Washington 99164, United States
| | - Susmita Bose
- W. M. Keck Biomedical Materials Research Laboratory, School of Mechanical and Materials Engineering, Washington State University, Pullman, Washington 99164, United States
| |
Collapse
|
29
|
McIlwrath SL, Carroll-Portillo AC, Lin HC, Westlund KN. In vivo imaging of cathepsin B in activated glia in the brain after orofacial formalin test. Sci Rep 2024; 14:4517. [PMID: 38402255 PMCID: PMC10894209 DOI: 10.1038/s41598-024-52854-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 01/24/2024] [Indexed: 02/26/2024] Open
Abstract
PURPOSE Cathepsin B (Cat B) is a cysteine lysosomal protease that is upregulated in many inflammatory diseases and widely expressed in the brain. Here, we used a Cat B activatable near-infrared (NIR) imaging probe to measure glial activation in vivo in the formalin test, a standard orofacial inflammatory pain model. The probe's efficacy was quantified with immunohistochemical analysis of the somatosensory cortex. PROCEDURES Three different concentrations of Cat B imaging probe (30, 50, 100 pmol/200 g bodyweight) were injected intracisternally into the foramen magnum of rats under anesthesia. Four hours later formalin (1.5%, 50 μl) was injected into the upper lip and the animal's behaviors recorded for 45 min. Subsequently, animals were repeatedly scanned using the IVIS Spectrum (8, 10, and 28 h post imaging probe injection) to measure extracellular Cat B activity. Aldehyde fixed brain sections were immunostained with antibodies against microglial marker Iba1 or astrocytic GFAP and detected with fluorescently labeled secondary antibodies to quantify co-localization with the fluorescent probe. RESULTS The Cat B imaging probe only slightly altered the formalin test results. Nocifensive behavior was only reduced in phase 1 in the 100 pmol group. In vivo measured fluorescence efficiency was highest in the 100 pmol group 28 h post imaging probe injection. Post-mortem immunohistochemical analysis of the somatosensory cortex detected the greatest amount of NIR fluorescence localized on microglia and astrocytes in the 100 pmol imaging probe group. Sensory neuron neuropeptide and cell injury marker expression in ipsilateral trigeminal ganglia was not altered by the presence of fluorescent probe. CONCLUSIONS These data demonstrate a concentration- and time-dependent visualization of extracellular Cat B in activated glia in the formalin test using a NIR imaging probe. Intracisternal injections are well suited for extracellular CNS proteinase detection in conditions when the blood-brain barrier is intact.
Collapse
Affiliation(s)
| | - Amanda C Carroll-Portillo
- New Mexico VA Health Care System, Albuquerque, NM, 87108, USA
- Division of Gastroenterology and Hepatology, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Henry C Lin
- New Mexico VA Health Care System, Albuquerque, NM, 87108, USA
- Division of Gastroenterology and Hepatology, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Karin N Westlund
- New Mexico VA Health Care System, Albuquerque, NM, 87108, USA
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, 87106, USA
| |
Collapse
|
30
|
Riley RF, Patel MP, Abbott JD, Bangalore S, Brilakis ES, Croce KJ, Doshi D, Kaul P, Kearney KE, Kerrigan JL, McEntegart M, Maehara A, Rymer JA, Sutton NR, Shah B. SCAI Expert Consensus Statement on the Management of Calcified Coronary Lesions. JOURNAL OF THE SOCIETY FOR CARDIOVASCULAR ANGIOGRAPHY & INTERVENTIONS 2024; 3:101259. [PMID: 39132214 PMCID: PMC11307856 DOI: 10.1016/j.jscai.2023.101259] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
The prevalence of calcification in obstructive coronary artery disease is on the rise. Percutaneous coronary intervention of these calcified lesions is associated with increased short-term and long-term risks. To optimize percutaneous coronary intervention results, there is an expanding array of treatment modalities geared toward calcium modification prior to stent implantation. The Society for Cardiovascular Angiography and Interventions, herein, puts forth an expert consensus document regarding methods to identify types of calcified coronary lesions, a central algorithm to help guide use of the various calcium modification strategies, tips for when using each treatment modality, and a look at future studies and trials for treating this challenging lesion subset.
Collapse
Affiliation(s)
| | | | - J. Dawn Abbott
- Lifespan Cardiovascular Institute, Providence, Rhode Island
| | | | | | | | - Darshan Doshi
- Massachusetts General Hospital, Boston, Massachusetts
| | | | | | | | - Margaret McEntegart
- NewYork-Presbyterian Hospital/Columbia University Irving Medical Center, New York, New York
| | - Akiko Maehara
- NewYork-Presbyterian Hospital/Columbia University Irving Medical Center, New York, New York
- Cardiovascular Research Foundation, New York, New York
| | | | - Nadia R. Sutton
- Vanderbilt University Medical Center, Nashville, Tennessee
- Vanderbilt University, Nashville, Tennessee
| | - Binita Shah
- NYU Grossman School of Medicine, New York, New York
- VA New York Harbor Healthcare System, New York, New York
| |
Collapse
|
31
|
Los J, Mensink FB, Mohammadnia N, Opstal TSJ, Damman P, Volleberg RHJA, Peeters DAM, van Royen N, Garcia-Garcia HM, Cornel JH, El Messaoudi S, van Geuns RJM. Invasive coronary imaging of inflammation to further characterize high-risk lesions: what options do we have? Front Cardiovasc Med 2024; 11:1352025. [PMID: 38370159 PMCID: PMC10871865 DOI: 10.3389/fcvm.2024.1352025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 01/15/2024] [Indexed: 02/20/2024] Open
Abstract
Coronary atherosclerosis remains a leading cause of morbidity and mortality worldwide. The underlying pathophysiology includes a complex interplay of endothelial dysfunction, lipid accumulation and inflammatory pathways. Multiple structural and inflammatory features of the atherosclerotic lesions have become targets to identify high-risk lesions. Various intracoronary imaging devices have been developed to assess the morphological, biocompositional and molecular profile of the intracoronary atheromata. These techniques guide interventional and therapeutical management and allow the identification and stratification of atherosclerotic lesions. We sought to provide an overview of the inflammatory pathobiology of atherosclerosis, distinct high-risk plaque features and the ability to visualize this process with contemporary intracoronary imaging techniques.
Collapse
Affiliation(s)
- Jonathan Los
- Department of Cardiology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Frans B. Mensink
- Department of Cardiology, Radboud University Medical Center, Nijmegen, Netherlands
| | | | - Tjerk S. J. Opstal
- Department of Cardiology, Radboud University Medical Center, Nijmegen, Netherlands
- Department of Cardiology, Northwest Clinics, Alkmaar, Netherlands
| | - Peter Damman
- Department of Cardiology, Radboud University Medical Center, Nijmegen, Netherlands
| | | | - Denise A. M. Peeters
- Department of Cardiology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Niels van Royen
- Department of Cardiology, Radboud University Medical Center, Nijmegen, Netherlands
| | | | - Jan H. Cornel
- Department of Cardiology, Radboud University Medical Center, Nijmegen, Netherlands
- Department of Cardiology, Northwest Clinics, Alkmaar, Netherlands
- Dutch Network for Cardiovascular Research (WCN), Utrecht, Netherlands
| | - Saloua El Messaoudi
- Department of Cardiology, Radboud University Medical Center, Nijmegen, Netherlands
| | | |
Collapse
|
32
|
Rahoual G, Zeitouni M, Charpentier E, Ritvo PG, Rouanet S, Procopi N, Boukhelifa S, Charleux P, Guedeney P, Kerneis M, Barthélémy O, Silvain J, Montalescot G, Redheuil A, Collet JP. Phenotyping coronary plaque by computed tomography in premature coronary artery disease. Eur Heart J Cardiovasc Imaging 2024; 25:257-266. [PMID: 37597177 DOI: 10.1093/ehjci/jead212] [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: 05/10/2023] [Revised: 07/06/2023] [Accepted: 08/09/2023] [Indexed: 08/21/2023] Open
Abstract
AIMS Premature coronary artery disease (CAD) is an aggressive disease with multiple recurrences mostly related to new coronary lesions. This study aimed to compare coronary plaque characteristics of individuals with premature CAD with those of incidental plaques found in matched individuals free of overt cardiovascular disease, using coronary computed tomography angiography (CCTA). METHODS AND RESULTS Of 1552 consecutive individuals who underwent CCTA, 106 individuals with history of acute or stable obstructive CAD ≤45 years were matched by age, sex, smoking status, cardiovascular heredity, and dyslipidaemia with 106 controls. CCTA were analysed for Coronary Artery Disease Reporting and Data System score, plaque composition, and high-risk plaque (HRP) features, including spotty calcification, positive remodelling, low attenuation, and napkin-ring sign. The characteristics of 348 premature CAD plaques were compared with those of 167 incidental coronary plaques of matched controls. The prevalence of non-calcified plaques was higher among individuals with premature CAD (65.1 vs. 30.2%, P < 0.001), as well as spotty calcification (42.5 vs. 17.9%, P < 0.001), positive remodelling (41.5 vs. 9.4%, P < 0.001), low attenuation (24.5 vs. 3.8%, P < 0.001), and napkin-ring sign (1.9 vs. 0.0%). They exhibited an average of 2.2 (2.7) HRP, while the control group displayed 0.4 (0.8) HRP (P < 0.001). Within a median follow-up of 24 (16, 34) months, individuals with premature CAD and ischaemic recurrence (n = 24) had more HRP [4.3 (3.9)] than those without ischaemic recurrence [1.5 (1.9)], mostly non-calcified with low attenuation and positive remodelling. CONCLUSION Coronary atherosclerosis in individuals with premature CAD is characterized by a high and predominant burden of non-calcified plaque and unusual high prevalence of HRP, contributing to disease progression with multiple recurrences. A comprehensive qualitative CCTA assessment of plaque characteristics may further risk stratify our patients, beyond cardiovascular risk factors.
Collapse
Affiliation(s)
- Ghilas Rahoual
- ACTION Study Group, INSERM UMRS 1166, Institut de Cardiologie, Hôpital Pitié-Salpêtrière (AP-HP), Sorbonne Université, 47-83 boulevard de l'Hôpital, Paris 75013, France
| | - Michel Zeitouni
- ACTION Study Group, INSERM UMRS 1166, Institut de Cardiologie, Hôpital Pitié-Salpêtrière (AP-HP), Sorbonne Université, 47-83 boulevard de l'Hôpital, Paris 75013, France
| | - Etienne Charpentier
- INSERM UMRS 1146, CNRS, Institute of Cardiometabolism and Nutrition, unité d'Imagerie Cardiovasculaire et Thoracique, Hôpital Pitié-Salpêtrière (AP-HP), Sorbonne Université, Paris 75013, France
| | - Paul-Gydeon Ritvo
- INSERM UMRS 1146, CNRS, Institute of Cardiometabolism and Nutrition, unité d'Imagerie Cardiovasculaire et Thoracique, Hôpital Pitié-Salpêtrière (AP-HP), Sorbonne Université, Paris 75013, France
| | - Stéphanie Rouanet
- Statistician Unit, StatEthic, ACTION Study Group, Levallois-Perret, France
| | - Niki Procopi
- ACTION Study Group, INSERM UMRS 1166, Institut de Cardiologie, Hôpital Pitié-Salpêtrière (AP-HP), Sorbonne Université, 47-83 boulevard de l'Hôpital, Paris 75013, France
| | - Sena Boukhelifa
- ACTION Study Group, INSERM UMRS 1166, Institut de Cardiologie, Hôpital Pitié-Salpêtrière (AP-HP), Sorbonne Université, 47-83 boulevard de l'Hôpital, Paris 75013, France
| | - Pierre Charleux
- ACTION Study Group, INSERM UMRS 1166, Institut de Cardiologie, Hôpital Pitié-Salpêtrière (AP-HP), Sorbonne Université, 47-83 boulevard de l'Hôpital, Paris 75013, France
| | - Paul Guedeney
- ACTION Study Group, INSERM UMRS 1166, Institut de Cardiologie, Hôpital Pitié-Salpêtrière (AP-HP), Sorbonne Université, 47-83 boulevard de l'Hôpital, Paris 75013, France
| | - Mathieu Kerneis
- ACTION Study Group, INSERM UMRS 1166, Institut de Cardiologie, Hôpital Pitié-Salpêtrière (AP-HP), Sorbonne Université, 47-83 boulevard de l'Hôpital, Paris 75013, France
| | - Olivier Barthélémy
- ACTION Study Group, INSERM UMRS 1166, Institut de Cardiologie, Hôpital Pitié-Salpêtrière (AP-HP), Sorbonne Université, 47-83 boulevard de l'Hôpital, Paris 75013, France
| | - Johanne Silvain
- ACTION Study Group, INSERM UMRS 1166, Institut de Cardiologie, Hôpital Pitié-Salpêtrière (AP-HP), Sorbonne Université, 47-83 boulevard de l'Hôpital, Paris 75013, France
| | - Gilles Montalescot
- ACTION Study Group, INSERM UMRS 1166, Institut de Cardiologie, Hôpital Pitié-Salpêtrière (AP-HP), Sorbonne Université, 47-83 boulevard de l'Hôpital, Paris 75013, France
| | - Alban Redheuil
- INSERM UMRS 1146, CNRS, Institute of Cardiometabolism and Nutrition, unité d'Imagerie Cardiovasculaire et Thoracique, Hôpital Pitié-Salpêtrière (AP-HP), Sorbonne Université, Paris 75013, France
| | - Jean-Philippe Collet
- ACTION Study Group, INSERM UMRS 1166, Institut de Cardiologie, Hôpital Pitié-Salpêtrière (AP-HP), Sorbonne Université, 47-83 boulevard de l'Hôpital, Paris 75013, France
| |
Collapse
|
33
|
Yang H, Liu Y, Chen G, Zhou B, Xu G, Li Q, Zhu L. Caspase-3/gasdermin-E axis facilitates the progression of coronary artery calcification by inducing the release of high mobility group box protein 1. Int Immunopharmacol 2024; 127:111454. [PMID: 38159554 DOI: 10.1016/j.intimp.2023.111454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 12/18/2023] [Accepted: 12/24/2023] [Indexed: 01/03/2024]
Abstract
Coronary artery calcification (CAC) is commonly observed in atherosclerotic plaques, which is a pathogenic factor for severe coronary artery disease (CAD). The phenotype changes of vascular smooth muscle cells (VSMCs) are found to participate in CAC progression, which is mainly induced by vascular inflammation and oxidative stress (OS). HMGB1, a critical inflammatory cytokine, is recently reported to induce arterial calcification, which is regulated by the Caspase-3/gasdermin-E (GSDME) axis. However, the function of the Caspase-3/GSDME axis in CAC is unknown. Herein, the involvement of the Caspase-3/GSDME axis in CAC was studied to explore the possible targets for CAC. CAC model was constructed in mice, which was verified by red cytoplasm in coronary artery tissues, increased macrophage infiltration, aggravated inflammation, and enhanced RAGE signaling, accompanied by an increased release of HMGB1 and an activated Caspase-3/ GSDME axis. In β-GP-treated MOVAS-1 cells, calcification, the ROS accumulation, enhanced LDH and HMGB1 release, enlarged macrophage production, aggravated inflammation, and activated RAGE signaling were observed, which were markedly abolished by the transfection of si-HMGB1 and si-GSDME. Moreover, the calcification deposition, the activity of Caspase-3/ GSDME axis, release of HMGB1, macrophage infiltration, cytokine production, and RAGE signaling in CAC mice were notably alleviated by VSMCs-specific GSDME knockdown, not by hematopoietic stem cells (HSCs)-specific GSDME knockdown. Collectively, Caspase-3/GSDME axis facilitated the progression of CAC by inducing the release of HMGB1.
Collapse
Affiliation(s)
- Honghui Yang
- Department of Cardiology, Zhengzhou University, Central China Fuwai Hospital, Zhengzhou 451464, PR China.
| | - Yingying Liu
- Department of Cardiology, Zhengzhou University, Central China Fuwai Hospital, Zhengzhou 451464, PR China
| | - Gengyu Chen
- Department of Cardiology, Zhengzhou University, Central China Fuwai Hospital, Zhengzhou 451464, PR China
| | - Botong Zhou
- Department of Cardiology, Zhengzhou University, Central China Fuwai Hospital, Zhengzhou 451464, PR China
| | - Guian Xu
- Department of Cardiology, Zhengzhou University, Central China Fuwai Hospital, Zhengzhou 451464, PR China
| | - Qingman Li
- Department of Cardiology, Zhengzhou University, Central China Fuwai Hospital, Zhengzhou 451464, PR China
| | - Lijie Zhu
- Department of Cardiology, Zhengzhou University, Central China Fuwai Hospital, Zhengzhou 451464, PR China
| |
Collapse
|
34
|
Kotsovilis S, Salagianni M, Varela A, Davos CH, Galani IE, Andreakos E. Comprehensive Analysis of 1-Year-Old Female Apolipoprotein E-Deficient Mice Reveals Advanced Atherosclerosis with Vulnerable Plaque Characteristics. Int J Mol Sci 2024; 25:1355. [PMID: 38279355 PMCID: PMC10816800 DOI: 10.3390/ijms25021355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 01/14/2024] [Accepted: 01/15/2024] [Indexed: 01/28/2024] Open
Abstract
Apolipoprotein E-knockout (Apoe-/-) mice constitute the most widely employed animal model of atherosclerosis. Deletion of Apoe induces profound hypercholesterolemia and promotes the development of atherosclerosis. However, despite its widespread use, the Apoe-/- mouse model remains incompletely characterized, especially at late time points and advanced disease stages. Thus, it is unclear how late atherosclerotic plaques compare to earlier ones in terms of lipid deposition, calcification, macrophage accumulation, smooth muscle cell presence, or plaque necrosis. Additionally, it is unknown how cardiac function and hemodynamic parameters are affected at late disease stages. Here, we used a comprehensive analysis based on histology, fluorescence microscopy, and Doppler ultrasonography to show that in normal chow diet-fed Apoe-/- mice, atherosclerotic lesions at the level of the aortic valve evolve from a more cellular macrophage-rich phenotype at 26 weeks to an acellular, lipid-rich, and more necrotic phenotype at 52 weeks of age, also marked by enhanced lipid deposition and calcification. Coronary artery atherosclerotic lesions are sparse at 26 weeks but ubiquitous and extensive at 52 weeks; yet, left ventricular function was not significantly affected. These findings demonstrate that atherosclerosis in Apoe-/- mice is a highly dynamic process, with atherosclerotic plaques evolving over time. At late disease stages, histopathological characteristics of increased plaque vulnerability predominate in combination with frequent and extensive coronary artery lesions, which nevertheless may not necessarily result in impaired cardiac function.
Collapse
Affiliation(s)
- Sotirios Kotsovilis
- Laboratory of Immunobiology, Center for Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation, Academy of Athens, GR 11527 Athens, Greece; (S.K.); (M.S.); (I.E.G.)
| | - Maria Salagianni
- Laboratory of Immunobiology, Center for Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation, Academy of Athens, GR 11527 Athens, Greece; (S.K.); (M.S.); (I.E.G.)
| | - Aimilia Varela
- Cardiovascular Research Laboratory, Center for Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation, Academy of Athens, GR 11527 Athens, Greece; (A.V.); (C.H.D.)
| | - Constantinos H. Davos
- Cardiovascular Research Laboratory, Center for Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation, Academy of Athens, GR 11527 Athens, Greece; (A.V.); (C.H.D.)
| | - Ioanna E. Galani
- Laboratory of Immunobiology, Center for Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation, Academy of Athens, GR 11527 Athens, Greece; (S.K.); (M.S.); (I.E.G.)
| | - Evangelos Andreakos
- Laboratory of Immunobiology, Center for Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation, Academy of Athens, GR 11527 Athens, Greece; (S.K.); (M.S.); (I.E.G.)
| |
Collapse
|
35
|
Qin Z, Yu L, Zhang Y, Xu Q, Li C, Zhao S, Xi X, Tian Y, Wang Z, Tian J, Yu B. Coronary artery calcification and plaque stability: an optical coherence tomography study. Heliyon 2023; 9:e23191. [PMID: 38149191 PMCID: PMC10750051 DOI: 10.1016/j.heliyon.2023.e23191] [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: 01/19/2023] [Revised: 10/12/2023] [Accepted: 11/29/2023] [Indexed: 12/28/2023] Open
Abstract
Background Coronary artery calcification (CAC), a surrogate of atherosclerosis, is related to stent underexpansion and adverse cardiac events. However, the effect of CAC on plaque stability is still controversial and the morphological significance of CAC has yet to be elucidated. Methods A retrospective series of 419 patients with acute coronary syndrome (ACS) who underwent optical coherence tomography (OCT) were enrolled. Patients were classified into three groups based on the calcification size in culprit plaques and the features of the culprit and non-culprit plaques among these groups were compared. Logistic regression was used to analyze independent risk factors for culprit plaque rupture and the nonlinear relationship between calcification parameters and culprit plaque rupture. Furthermore, we compared the detailed calcification parameters of different kinds of plaques. Results A total of 419 culprit plaques and 364 non-culprit plaques were identified. The incidence of calcification was 53.9 % in culprit plaques and 50.3 % in non-culprit plaques. Compared with culprit plaques without calcification, plaque rupture, macrophages and cholesterol crystals were more frequently observed in the spotty calcification group, and the lipid length was longer; the incidence of macrophages and cholesterol crystals was higher in the macrocalcification group. Calcification tended to be smaller in ruptured plaques than in non-ruptured plaques. Moreover, the arc and length of calcification were greater in culprit plaques than in non-culprit plaques. Conclusions Vulnerable features were more frequently observed in culprit plaques with spotty calcification, whereas the presence of macrocalcification calcifications did not significantly increase plaque vulnerability. Calcification tends to be larger in culprit plaques than in non-culprit plaques.
Collapse
Affiliation(s)
- Zhifeng Qin
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, China
| | - Li Yu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, China
| | - Yanwen Zhang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, China
| | - Qinglu Xu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, China
| | - Chao Li
- School of Electronic Science and Engineering, University of Electronic Science and Technology of China, Chengdu, China
| | - Suhong Zhao
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, China
| | - Xiangwen Xi
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, China
| | - Yanan Tian
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, China
| | - Zhao Wang
- School of Electronic Science and Engineering, University of Electronic Science and Technology of China, Chengdu, China
| | - Jinwei Tian
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, China
| | - Bo Yu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, China
| |
Collapse
|
36
|
Kitagawa T, Sasaki K, Fujii Y, Ikegami Y, Tatsugami F, Awai K, Hirokawa Y, Nakano Y. 18F-sodium fluoride positron emission tomography following coronary computed tomography angiography in predicting long-term coronary events: a 5-year follow-up study. J Nucl Cardiol 2023; 30:2365-2378. [PMID: 37127726 DOI: 10.1007/s12350-023-03277-5] [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: 01/12/2023] [Accepted: 03/31/2023] [Indexed: 05/03/2023]
Abstract
PURPOSE The predictive value of 18F-sodium fluoride (18F-NaF) positron emission tomography (PET) in combination with coronary computed tomography (CT) angiography (CCTA) for future coronary events has attracted interest. We evaluated the potential of 18F-NaF PET/CT following CCTA to predict major coronary events (MACE) during a 5-year follow-up period. METHODS Forty patients with coronary atherosclerotic lesions detected on CCTA underwent 18F-NaF PET/CT examination. Each lesion was evaluated for luminal stenosis and high-risk plaque (HRP) with < 30 Hounsfield units and a > 1.1 remodeling index on CCTA. Focal 18F-NaF uptake in each lesion was quantified using the maximum tissue-to-background ratio (TBRmax), and the maximum TBRmax per patient (M-TBRmax) was determined. We followed MACE (cardiac death, acute coronary syndrome, and/or coronary revascularization > 6 months after 18F-NaF PET/CT) for 5 years. RESULTS In total, 142 coronary lesions were analyzed. Eleven patients experienced any MACE. Patients with MACE showed a higher M-TBRmax than those without (1.40 ± .19 vs. 1.18 ± .18, P = .0011), and the optimal M-TBRmax cutoff to predict MACE was 1.29. Patients with M-TBRmax of ≥ 1.29 had a higher risk of MACE than those with lower values (P = .012, log-rank test), whereas patients with obstructive stenosis and those with HRP did not. Multivariate Cox proportional analysis adjusted for age, sex, coronary risk factors, and CCTA findings showed that M-TBRmax of ≥ 1.29 remained an independent predictor of 5-year MACE (hazard ratio, 5.4; 95% confidence interval, 1.1-25.4; P = .034). CONCLUSION 18F-NaF PET/CT following CCTA provides useful strategies to predict 5-year MACE.
Collapse
Affiliation(s)
- Toshiro Kitagawa
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, 1-2-3 Kasumi, Minami-Ku, Hiroshima, 734-8551, Japan.
| | - Ko Sasaki
- Hiroshima Heiwa Clinic, Hiroshima, Japan
| | - Yuto Fujii
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, 1-2-3 Kasumi, Minami-Ku, Hiroshima, 734-8551, Japan
| | - Yuki Ikegami
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, 1-2-3 Kasumi, Minami-Ku, Hiroshima, 734-8551, Japan
| | - Fuminari Tatsugami
- Department of Diagnostic Radiology, Hiroshima University Hospital, Hiroshima, Japan
| | - Kazuo Awai
- Department of Diagnostic Radiology, Hiroshima University Hospital, Hiroshima, Japan
| | | | - Yukiko Nakano
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, 1-2-3 Kasumi, Minami-Ku, Hiroshima, 734-8551, Japan
| |
Collapse
|
37
|
Dong Y, Liu Y, Cheng P, Liao H, Jiang C, Li Y, Liu S, Xu X. Lower limb arterial calcification and its clinical relevance with peripheral arterial disease. Front Cardiovasc Med 2023; 10:1271100. [PMID: 38075978 PMCID: PMC10710292 DOI: 10.3389/fcvm.2023.1271100] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 11/08/2023] [Indexed: 09/14/2024] Open
Abstract
Lower limb arterial calcification (LLAC) is associated with an increased risk of mortality and it predicts poor outcomes after endovascular interventions in patients with peripheral artery disease (PAD). Detailed histological analysis of human lower artery specimens pinpointed the presence of LLAC in two distinct layers: the intima and the media. Intimal calcification has been assumed to be an atherosclerotic pathology and it is associated with smoking and obesity. It becomes instrumental in lumen stenosis, thereby playing a crucial role in disease progression. On the contrary, medial calcification is a separate process, systematically regulated and linked with age advancement, diabetes, and chronic kidney disease. It prominently interacts with vasodilation and arterial stiffness. Given that both types of calcifications frequently co-exist in PAD patients, it is vital to understand their respective mechanisms within the context of PAD. Calcification can be easily identifiable entity on imaging scans. Considering the highly improved abilities of novel imaging technologies in differentiating intimal and medial calcification within the lower limb arteries, this review aimed to describe the distinct histological and imaging features of the two types of LLAC. Additionally, it aims to provide in-depth insight into the risk factors, the effects on hemodynamics, and the clinical implications of LLAC, either occurring in the intimal or medial layers.
Collapse
Affiliation(s)
- Yue Dong
- Department of Radiology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuankang Liu
- Department of Radiology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Panpan Cheng
- Department of Radiology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongli Liao
- Department of Radiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cuiping Jiang
- Department of Radiology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Li
- Department of Radiology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuhua Liu
- Department of Burns, Tongren Hospital of Wuhan University, Wuhan, China
| | - Xiangyang Xu
- Department of Radiology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
38
|
Jia J, Zhang J, He Q, Wang M, Liu Q, Wang T, Chen X, Wang W, Xu H. Association between dietary vitamin C and abdominal aortic calcification among the US adults. Nutr J 2023; 22:58. [PMID: 37964312 PMCID: PMC10647183 DOI: 10.1186/s12937-023-00889-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 10/30/2023] [Indexed: 11/16/2023] Open
Abstract
BACKGROUND Cardiovascular disease (CVD) is the leading cause of mortality, and vascular calcification has been highly correlated with CVD events. Abdominal aortic calcification (AAC) has been shown to predict subclinical CVD and incident CVD events. However, the relationship between vitamin C and abdominal aortic calcification remains unclear. OBJECTIVE To investigate the relationship of dietary vitamin C with AAC among the adult population in the US. METHODS The National Health and Nutrition Examination Survey (NHANES) 2013-2014 provided the data for the cross-sectional study. 2297 subjects (1089 males) were included in the study. Two scoring systems, AAC 24-point scale (Kauppila) and AAC 8-point scale (Schousboe), were used for the measurement of AAC score. Dietary vitamin C intake was calculated as the average of two rounds of 24-h interview recall data and classified in tertiles for analysis. We applied weighted multiple regression analyses to assess the relationship of dietary vitamin C with AAC score and the risk of having AAC. To ensure the robustness of the findings, subgroup and sensitivity analyses were performed. Additionally, smooth curve fittings, using generalized additive models (GAM) were employed to visualize potential nonlinear relationships. Furthermore, an exploratory analysis on the relationship of vitamin C supplements with AAC was also conducted. RESULTS The results showed that higher dietary vitamin C intake was related to a reduction in AAC score (AAC-24: β = -0.338, 95% confidence interval [CI] -0.565, -0.111, P = 0.004; AAC-8: β = -0.132, 95%CI -0.217, -0.047, P = 0.002), and lower risk of AAC (odds ratio [OR] = 0.807, 95%CI 0.659, 0.989, P = 0.038). However, the relationship of vitamin C supplements with AAC was not identified. CONCLUSIONS The study revealed that higher intake of dietary vitamin C rather than vitamin C supplements was related to reduced AAC score and lower risk of AAC, indicating that diets rich in vitamin C are recommended due to its potential benefits for protecting against vascular calcification and CVD among the adult population in the US.
Collapse
Affiliation(s)
- Jundi Jia
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Academy of Chinese Medical Sciences, Beijing, China
| | - Jie Zhang
- National Integrated Traditional and Western Medicine Center for Cardiovascular Disease, China-Japan Friendship Hospital, Beijing, China
| | - Qiao He
- Clinical Epidemiology and Evidence-Based Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Mingqi Wang
- Clinical Epidemiology and Evidence-Based Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Qiyu Liu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, China
| | - Tongxin Wang
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Academy of Chinese Medical Sciences, Beijing, China
| | - Xuanye Chen
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Academy of Chinese Medical Sciences, Beijing, China
| | - Wen Wang
- Clinical Epidemiology and Evidence-Based Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China.
| | - Hao Xu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
39
|
Sun X, Zheng Y, Xie L, Zhou Y, Liu R, Ma Y, Zhao M, Liu Y. Autophagy reduces aortic calcification in diabetic mice by reducing matrix vesicle body-mediated IL-1β release. Exp Cell Res 2023; 432:113803. [PMID: 37774764 DOI: 10.1016/j.yexcr.2023.113803] [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: 08/04/2023] [Revised: 09/24/2023] [Accepted: 09/26/2023] [Indexed: 10/01/2023]
Abstract
Vascular calcification (VC) is a common pathological process of cardiovascular disease that occurs in patients with type 2 diabetes mellitus (T2DM). However, the molecular basis of VC progression remains unknown. A GEO dataset (GSE146638) was analyzed to show that microbodies and IL-1β may play important roles in the pathophysiology of VC. The release of matrix vesicle bodies (MVBs) and IL-1β and the colocalization of IL-1β with MVBs or autophagosomes were studied by immunofluorescence in an in vivo diabetes mouse model with aortic calcification and an in vitro high glucose cell calcification model. MVB numbers, IL-1β levels and autophagy were increased in calcified mouse aortas and calcified vascular smooth muscle cells (VSMCs). IL-1β colocalized with MVBs and autophagosomes. The MVBs from calcified VSMCs induced the calcification of normal recipient VSMCs, and this effect was alleviated by silencing IL-1β. The autophagy inducer rapamycin reduced IL-1β expression and calcification in VSMCs, while these processes were induced by the autophagy inhibitor chloroquine. In conclusion, our results suggested that MVBs could carry IL-1β out of cells and induce VC in normal VSMCs, and these processes could be counteracted by autophagy. These results suggested that MVB-mediated IL-1β release may be an effective target for treating vascular calcification.
Collapse
Affiliation(s)
- Xiaolei Sun
- Department of General Surgery (Vascular Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China; Department of Interventional Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China; Laboratory of Nucleic Acids in Medicine for National High-Level Talents, Nucleic Acid Medicine of Luzhou Key Laboratory, Southwest Medical University, Luzhou, 646000, China; Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China; Cardiovascular and Metabolic Diseases Key Laboratory of Sichuan, Luzhou, 646000, China.
| | - Yang Zheng
- Department of General Surgery (Vascular Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China; Department of Vascular and Interventional, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Linzhuo Xie
- Department of General Surgery (Vascular Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Yuanqun Zhou
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China; State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion of Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Runyu Liu
- Department of General Surgery (Vascular Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Yarong Ma
- Department of Ophthalmology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Ming Zhao
- Department of Gastroenterology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, China.
| | - Yong Liu
- Department of General Surgery (Vascular Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
40
|
Nelles G, Abdelwahed YS, Alyaqoob A, Seppelt C, Stähli BE, Meteva D, Kränkel N, Haghikia A, Skurk C, Dreger H, Knebel F, Trippel TD, Krisper M, Sieronski L, Gerhardt T, Zanders L, Klotsche J, Landmesser U, Joner M, Leistner DM. Spotty calcium deposits within acute coronary syndrome (ACS)-causing culprit lesions impact inflammatory vessel-wall interactions and are associated with higher cardiovascular event rates at one year follow-up: Results from the prospective translational OPTICO-ACS study program. Atherosclerosis 2023; 385:117284. [PMID: 37871405 DOI: 10.1016/j.atherosclerosis.2023.117284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 08/31/2023] [Accepted: 09/06/2023] [Indexed: 10/25/2023]
Abstract
BACKGROUND AND AIMS Spotty calcium deposits (SCD) represent a vulnerable plaque feature which seems to result - as based on recent invitro studies - from inflammatory vessel-wall interactions. SCD can be reliably assessed by optical coherence tomography (OCT). Their prognostic impact is yet unknown. Therefore, the aims of this translational study were to comprehensively characterize different plaque calcification patterns, to analyze the associated inflammatory mechanisms in the microenvironment of acute coronary syndrome (ACS)-causing culprit lesions (CL) and to investigate the prognostic significance of SCD in a large cohort of ACS-patients. METHODS CL of the first 155 consecutive ACS-patients from the translational OPTICO-ACS-study program were investigated by OCT-characterization of the calcium phenotype at ACS-causing culprit lesions. Simultaneous immunophenotyping by flow-cytometric analysis and cytokine bead array technique across the CL gradient (ratio local/systemic levels) was performed and incidental major adverse cardiovascular events plus (MACE+) at 12 months after ACS were assessed. RESULTS SCD were observed within 45.2% of all analyzed ACS-causing culprit lesions (CL). Culprits containing spotty calcium were characterized by an increased culprit ratio of innate effector cytokines interleukin (IL)-8 [2.04 (1.24) vs. 1.37 (1.10) p < 0.05], as well as TNF (tumor necrosis factor)-α [1.17 (0.93) vs. 1.06 (0.89); p < 0.05)] and an increased ratio of circulating neutrophils [0.96 (0.85) vs. 0.91 (0.77); p < 0.05] as compared to culprit plaques without SCD. Total monocyte levels did not differ between the two groups (p = n.s.). However, SCD-containing CLs were characterized by an increased culprit ratio of intermediate monocytes [(1.15 (0.81) vs. 0.96 (0.84); p < 0.05)] with an enhanced surface expression of the integrin receptor CD49d as compared to intermediate monocytes derived from SCD-free CLs [(1.06 (0.94) vs. 0.97 (0.91)] p < 0.05. Finally, 12 months rates of MACE+ were higher in patients with, as compared to patients without SCD at CL (16.4% vs. 5.3%; p < 0.05). CONCLUSIONS This study for the first time identified a specific inflammatory profile of CL with SCD, with a predominance of neutrophils, intermediate monocytes and their corresponding effector molecules. Hence, this study advances our understanding of ACS-causing CL and provides the basis for future personalized anti-inflammatory, therapeutic approaches to ACS.
Collapse
Affiliation(s)
- Gregor Nelles
- Department of Cardiology Charité University Medicine Berlin, Campus Benjamin-Franklin, 12203, Berlin, Germany; DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, 12203, Berlin, Germany; Department of Medicine, Cardiology/Angiology, Goethe University Hospital, Frankfurt, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Frankfurt Rhine-Main, Frankfurt, Germany
| | - Youssef S Abdelwahed
- Department of Cardiology Charité University Medicine Berlin, Campus Benjamin-Franklin, 12203, Berlin, Germany; DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, 12203, Berlin, Germany
| | - Aseel Alyaqoob
- Department of Cardiology and ISAR Research Centre, German Heart Centre, 80636, Munich, Germany; DZHK (German Centre for Cardiovascular Research) Partner Site Munch, 80636, Munich, Germany
| | - Claudio Seppelt
- Department of Cardiology Charité University Medicine Berlin, Campus Benjamin-Franklin, 12203, Berlin, Germany; DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, 12203, Berlin, Germany; Department of Medicine, Cardiology/Angiology, Goethe University Hospital, Frankfurt, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Frankfurt Rhine-Main, Frankfurt, Germany
| | - Barbara E Stähli
- Department of Cardiology Charité University Medicine Berlin, Campus Benjamin-Franklin, 12203, Berlin, Germany; DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, 12203, Berlin, Germany; Department of Cardiology, Universitäres Herzzentrum, Universitätsspital Zürich, Zurich, Switzerland
| | - Denitsa Meteva
- Department of Cardiology Charité University Medicine Berlin, Campus Benjamin-Franklin, 12203, Berlin, Germany; DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, 12203, Berlin, Germany
| | - Nicolle Kränkel
- Department of Cardiology Charité University Medicine Berlin, Campus Benjamin-Franklin, 12203, Berlin, Germany; DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, 12203, Berlin, Germany
| | - Arash Haghikia
- Department of Cardiology Charité University Medicine Berlin, Campus Benjamin-Franklin, 12203, Berlin, Germany; DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, 12203, Berlin, Germany
| | - Carsten Skurk
- Department of Cardiology Charité University Medicine Berlin, Campus Benjamin-Franklin, 12203, Berlin, Germany; DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, 12203, Berlin, Germany
| | - Henryk Dreger
- DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, 12203, Berlin, Germany; Department of Cardiology Charité University Medicine Berlin, Campus Mitte, 10117, Germany
| | - Fabian Knebel
- DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, 12203, Berlin, Germany; Department of Cardiology Charité University Medicine Berlin, Campus Mitte, 10117, Germany; Department of Cardiology, Sana Clinic Lichtenberg, 10365, Berlin, Germany
| | - Tobias D Trippel
- DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, 12203, Berlin, Germany; Department of Cardiology, Charité University Medicine, Campus Virchow, 13353, Berlin, Germany
| | - Maximilian Krisper
- Department of Cardiology, Charité University Medicine, Campus Virchow, 13353, Berlin, Germany
| | - Lara Sieronski
- Department of Cardiology Charité University Medicine Berlin, Campus Benjamin-Franklin, 12203, Berlin, Germany; DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, 12203, Berlin, Germany
| | - Teresa Gerhardt
- Department of Cardiology Charité University Medicine Berlin, Campus Benjamin-Franklin, 12203, Berlin, Germany; DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, 12203, Berlin, Germany; Berlin Institute of Health (BIH), 10117, Berlin, Germany; Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, USA
| | - Lukas Zanders
- Department of Cardiology Charité University Medicine Berlin, Campus Benjamin-Franklin, 12203, Berlin, Germany; DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, 12203, Berlin, Germany
| | - Jens Klotsche
- German Rheumatism Research Centre Berlin, Institute for Social Medicine, Epidemiology und Heath Economy, Charité University Medicine Berlin, Campus Mitte, 10117, Berlin, Germany
| | - Ulf Landmesser
- Department of Cardiology Charité University Medicine Berlin, Campus Benjamin-Franklin, 12203, Berlin, Germany; DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, 12203, Berlin, Germany; Berlin Institute of Health (BIH), 10117, Berlin, Germany
| | - Michael Joner
- Department of Cardiology and ISAR Research Centre, German Heart Centre, 80636, Munich, Germany; DZHK (German Centre for Cardiovascular Research) Partner Site Munch, 80636, Munich, Germany
| | - David M Leistner
- Department of Cardiology Charité University Medicine Berlin, Campus Benjamin-Franklin, 12203, Berlin, Germany; DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, 12203, Berlin, Germany; Department of Medicine, Cardiology/Angiology, Goethe University Hospital, Frankfurt, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Frankfurt Rhine-Main, Frankfurt, Germany; Berlin Institute of Health (BIH), 10117, Berlin, Germany.
| |
Collapse
|
41
|
Ho LC, Chen YH, Wu TY, Kao LZ, Hung SY, Liou HH, Chen PC, Tsai PJ, Lin HK, Lee YC, Wang HH, Tsai YS. Phosphate burden induces vascular calcification through a NLRP3-caspase-1-mediated pyroptotic pathway. Life Sci 2023; 332:122123. [PMID: 37742736 DOI: 10.1016/j.lfs.2023.122123] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 09/01/2023] [Accepted: 09/21/2023] [Indexed: 09/26/2023]
Abstract
AIMS The aim of this study is to clarify the role of NLRP3 inflammasome in phosphate burden-induced vascular smooth muscle cell (VSMC) calcification. MAIN METHODS VSMC calcification was induced using a high concentration of inorganic phosphate. After pharmacological inhibition or genetic silencing of the NLRP3 inflammasome, pyroptosis, or potassium efflux, the cells were examined by RT-qPCR, immunofluorescence, and western blotting to identify the NLRP3-mediated pathway for VSMC calcification. KEY FINDINGS Calcified VSMCs with α-smooth muscle actin (α-SMA) disarray presented features of pyroptosis, including caspase-1 maturation, cleaved gasdermin D (GSDMD), and a high supernatant level of lactate dehydrogenase A. Pharmacological inhibitions of caspase-1 and pyroptosis attenuated VSMC calcification, whereas interleukin-1β receptor antagonism did not. Unlike canonical NLRP3 activation, osteogenic VSMCs did not upregulate NLRP3 expression. However, NLRP3 genetic silencing or inhibitions, which targets different domains of the NLRP3 protein, could ameliorate VSMC calcification by aborting caspase-1 and GSDMD activation. Furthermore, potassium efflux through the inward-rectifier potassium channel, and not through the P2X7 receptor, triggered NLRP3 inflammasome activation and VSMC calcification. SIGNIFICANCE In the present study, we identified a potassium efflux-triggered NLRP3-caspase-1-mediated pyroptotic pathway for VSMC calcification that is unique and different from the canonical NLRP3 inflammasome activation. Therefore, targeting this pathway may serve as a novel therapeutic strategy for vascular calcification.
Collapse
Affiliation(s)
- Li-Chun Ho
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan; Division of General Medicine, Department of Internal Medicine, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan.
| | - Yu-Hsin Chen
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ting-Yun Wu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ling-Zhen Kao
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Shih-Yuan Hung
- Division of Nephrology, Department of Internal Medicine, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Hung-Hsiang Liou
- Division of Nephrology, Department of Internal Medicine, Hsin-Jen Hospital, New Taipei City, Taiwan
| | - Pei-Chun Chen
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Pei-Jane Tsai
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston Salem, NC, USA
| | - Hui-Kuan Lin
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston Salem, NC, USA
| | - Yi-Che Lee
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Hsi-Hao Wang
- Division of Nephrology, Department of Internal Medicine, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Yau-Sheng Tsai
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston Salem, NC, USA; Clinical Medicine Research Center, National Cheng Kung University Hospital, Tainan, Taiwan.
| |
Collapse
|
42
|
Ballester-Servera C, Alonso J, Cañes L, Vázquez-Sufuentes P, Puertas-Umbert L, Fernández-Celis A, Taurón M, Rodríguez-Sinovas A, López-Andrés N, Rodríguez C, Martínez-González J. Lysyl oxidase-dependent extracellular matrix crosslinking modulates calcification in atherosclerosis and aortic valve disease. Biomed Pharmacother 2023; 167:115469. [PMID: 37729730 DOI: 10.1016/j.biopha.2023.115469] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/30/2023] [Accepted: 09/07/2023] [Indexed: 09/22/2023] Open
Abstract
Extracellular matrix (ECM) is an active player in cardiovascular calcification (CVC), a major public health issue with an unmet need for effective therapies. Lysyl oxidase (LOX) conditions ECM biomechanical properties; thus, we hypothesized that LOX might impact on mineral deposition in calcific aortic valve disease (CAVD) and atherosclerosis. LOX was upregulated in calcified valves from two cohorts of CAVD patients. Strong LOX immunostaining was detected surrounding calcified foci in calcified human valves and atherosclerotic lesions colocalizing with RUNX2 on valvular interstitial cells (VICs) or vascular smooth muscle cells (VSMCs). Both LOX secretion and organized collagen deposition were enhanced in calcifying VICs exposed to osteogenic media. β-aminopropionitrile (BAPN), an inhibitor of LOX, attenuated collagen deposition and calcification. VICs seeded onto decellularized matrices from BAPN-treated VICs calcified less than cells cultured onto control scaffolds; instead, VICs exposed to conditioned media from cells over-expressing LOX or cultured onto LOX-crosslinked matrices calcified more. Atherosclerosis was induced in WT and transgenic mice that overexpress LOX in VSMC (TgLOXVSMC) by AAV-PCSK9D374Y injection and high-fat feeding. In atherosclerosis-challenged TgLOXVSMC mice both atherosclerosis burden and calcification assessed by near-infrared fluorescence (NIRF) imaging were higher than in WT mice. These animals also exhibited larger calcified areas in atherosclerotic lesions from aortic arches and brachiocephalic arteries. Moreover, LOX transgenesis exacerbated plaque inflammation, and increased VSMC cellularity, the rate of RUNX2-positive cells and both connective tissue content and collagen cross-linking. Our findings highlight the relevance of LOX in CVC and postulate this enzyme as a potential therapeutic target for CVC.
Collapse
Affiliation(s)
- Carme Ballester-Servera
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona, Spain; CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain; Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Judith Alonso
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona, Spain; CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain; Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Laia Cañes
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona, Spain; CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain; Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Paula Vázquez-Sufuentes
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona, Spain; Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Lídia Puertas-Umbert
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain; Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain; Institut de Recerca Hospital de la Santa Creu i Sant Pau (IRHSCSP), Barcelona, Spain
| | - Amaya Fernández-Celis
- Cardiovascular Translational Research, Navarrabiomed, IdiSNA, UPNA, Hospital Universitario de Navarra (HUN), Pamplona, Spain
| | - Manel Taurón
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain; Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain; Departamento de Cirugía Cardíaca, Hospital de la Santa Creu i Sant Pau-Universitat Autònoma de Barcelona (HSCSP-UAB), Barcelona, Spain
| | - Antonio Rodríguez-Sinovas
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain; Cardiovascular Diseases Research Group, Vall d'Hebron University Hospital and Research Institute, Barcelona, Spain
| | - Natalia López-Andrés
- Cardiovascular Translational Research, Navarrabiomed, IdiSNA, UPNA, Hospital Universitario de Navarra (HUN), Pamplona, Spain
| | - Cristina Rodríguez
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain; Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain; Institut de Recerca Hospital de la Santa Creu i Sant Pau (IRHSCSP), Barcelona, Spain.
| | - José Martínez-González
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona, Spain; CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain; Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain.
| |
Collapse
|
43
|
Alfaidi M, Evans PC, Pickering JG. Editorial: Endothelial-to-mesenchymal transition in cardiovascular disease. Front Cardiovasc Med 2023; 10:1290050. [PMID: 37900559 PMCID: PMC10602815 DOI: 10.3389/fcvm.2023.1290050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 09/22/2023] [Indexed: 10/31/2023] Open
Affiliation(s)
- Mabruka Alfaidi
- Department of Internal Medicine, Division of Cardiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LS, United States
| | - Paul C. Evans
- Biochemical Pharmacology, William Harvey Research Institute, Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - J. Geoffrey Pickering
- Departments of Medicine, Biochemistry, and Medical Biophysics, Western University, London, ON, Canada
| |
Collapse
|
44
|
Nouizi F, Kwong TC, Turong B, Nikkhah D, Sampathkumaran U, Gulsen G. Fast ICCD-based temperature modulated fluorescence tomography. APPLIED OPTICS 2023; 62:7420-7430. [PMID: 37855510 PMCID: PMC11396546 DOI: 10.1364/ao.499281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 09/06/2023] [Indexed: 10/20/2023]
Abstract
Fluorescence tomography (FT) has become a powerful preclinical imaging modality with a great potential for several clinical applications. Although it has superior sensitivity and utilizes low-cost instrumentation, the highly scattering nature of bio-tissue makes FT in thick samples challenging, resulting in poor resolution and low quantitative accuracy. To overcome the limitations of FT, we previously introduced a novel method, termed temperature modulated fluorescence tomography (TMFT), which is based on two key elements: (1) temperature-sensitive fluorescent agents (ThermoDots) and (2) high-intensity focused ultrasound (HIFU). The fluorescence emission of ThermoDots increases up to hundredfold with only several degree temperature elevation. The exceptional and reversible response of these ThermoDots enables their modulation, which effectively allows their localization using the HIFU. Their localization is then used as functional a priori during the FT image reconstruction process to resolve their distribution with higher spatial resolution. The last version of the TMFT system was based on a cooled CCD camera utilizing a step-and-shoot mode, which necessitated long total imaging time only for a small selected region of interest (ROI). In this paper, we present the latest version of our TMFT technology, which uses a much faster continuous HIFU scanning mode based on an intensified CCD (ICCD) camera. This new, to the best of our knowledge, version can capture the whole field-of-view (FOV) of 50×30m m 2 at once and reduces the total imaging time down to 30 min, while preserving the same high resolution (∼1.3m m) and superior quantitative accuracy (<7% error) as the previous versions. Therefore, this new method is an important step toward utilization of TMFT for preclinical imaging.
Collapse
|
45
|
Hosoda H, Kataoka Y, Nicholls SJ, Puri R, Murai K, Kitahara S, Mitsui K, Sugane H, Sawada K, Iwai T, Matama H, Honda S, Takagi K, Fujino M, Yoneda S, Otsuka F, Takamisawa I, Nishihira K, Asaumi Y, Kawai K, Noguchi T. Calcified plaque harboring lipidic materials associates with no-reflow phenomenon after PCI in stable CAD. Int J Cardiovasc Imaging 2023; 39:1927-1941. [PMID: 37378706 PMCID: PMC10589149 DOI: 10.1007/s10554-023-02905-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 06/19/2023] [Indexed: 06/29/2023]
Abstract
Calcified atheroma has been viewed conventionally as stable lesion which less likely increases no-reflow phenomenon. Given that lipidic materials triggers the formation of calcification, lipidic materials could exist within calcified lesion, which may cause no-reflow phenomenon after PCI. The REASSURE-NIRS registry (NCT04864171) employed near-infrared spectroscopy and intravascular ultrasound imaging to evaluate maximum 4-mm lipid-core burden index (maxLCBI4mm) at target lesions containing small (maximum calcification arc < 180°: n = 272) and large calcification (maximum calcification arc ≥ 180°: n = 189) in stable CAD patients. The associations of maxLCBI4mm with corrected TIMI frame count (CTFC) and no-reflow phenomenon after PCI were analyzed in patients with target lesions containing small and large calcification, respectively. No-reflow phenomenon occurred in 8.0% of study population. Receiver-operating characteristics curve analyses revealed that optimal cut-off values of maxLCBI4mm for predicting no-reflow phenomenon were 585 at small calcification (AUC = 0.72, p < 0.001) and 679 at large calcification (AUC = 0.76, p = 0.001). Target lesions containing small calcification with maxLCBI4mm ≥ 585 more likely exhibited a greater CTFC (p < 0.001). In those with large calcification, 55.6% of them had maxLCBI4mm ≥ 400 [vs. 56.2% (small calcification), p = 0.82]. Furthermore, a higher CTFC (p < 0.001) was observed in association with maxLCBI4mm ≥ 679 at large calcification. On multivariable analysis, maxLCBI4mm at large calcification still independently predicted no-reflow phenomenon (OR = 1.60, 95%CI = 1.32-1.94, p < 0.001). MaxLCBI4mm at target lesions exhibiting large calcification elevated a risk of no-reflow phenomenon after PCI. Calcified plaque containing lipidic materials is not necessarily stable lesion, but could be active and high-risk one causing no-reflow phenomenon.
Collapse
Affiliation(s)
- Hayato Hosoda
- Department of Cardiovascular Medicine, Chikamori Hospital, Kochi, India
| | - Yu Kataoka
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, 6-1, Kishibe-Shimmachi, Suita, Osaka, 564-8565, Japan.
| | | | - Rishi Puri
- Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Kota Murai
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, 6-1, Kishibe-Shimmachi, Suita, Osaka, 564-8565, Japan
| | - Satoshi Kitahara
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, 6-1, Kishibe-Shimmachi, Suita, Osaka, 564-8565, Japan
| | - Kentaro Mitsui
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, 6-1, Kishibe-Shimmachi, Suita, Osaka, 564-8565, Japan
| | - Hiroki Sugane
- Department of Cardiovascular Medicine, Chikamori Hospital, Kochi, India
| | - Kenichiro Sawada
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, 6-1, Kishibe-Shimmachi, Suita, Osaka, 564-8565, Japan
| | - Takamasa Iwai
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, 6-1, Kishibe-Shimmachi, Suita, Osaka, 564-8565, Japan
| | - Hideo Matama
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, 6-1, Kishibe-Shimmachi, Suita, Osaka, 564-8565, Japan
| | - Satoshi Honda
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, 6-1, Kishibe-Shimmachi, Suita, Osaka, 564-8565, Japan
| | - Kensuke Takagi
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, 6-1, Kishibe-Shimmachi, Suita, Osaka, 564-8565, Japan
| | - Masashi Fujino
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, 6-1, Kishibe-Shimmachi, Suita, Osaka, 564-8565, Japan
| | - Shuichi Yoneda
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, 6-1, Kishibe-Shimmachi, Suita, Osaka, 564-8565, Japan
| | - Fumiyuki Otsuka
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, 6-1, Kishibe-Shimmachi, Suita, Osaka, 564-8565, Japan
| | - Itaru Takamisawa
- Department of Cardiovascular Medicine, Sakakibara Heart Institute, Fuchyu, Tokyo, Japan
| | - Kensaku Nishihira
- Department of Cardiology, Miyazaki Medical Association Hospital, Miyazaki, Japan
| | - Yasuhide Asaumi
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, 6-1, Kishibe-Shimmachi, Suita, Osaka, 564-8565, Japan
| | - Kazuya Kawai
- Department of Cardiovascular Medicine, Chikamori Hospital, Kochi, India
| | - Teruo Noguchi
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, 6-1, Kishibe-Shimmachi, Suita, Osaka, 564-8565, Japan
| |
Collapse
|
46
|
Jansen I, Crielaard H, Wissing T, Bouten C, Gijsen F, Akyildiz AC, Farrell E, van der Heiden K. A tissue-engineered model of the atherosclerotic plaque cap: Toward understanding the role of microcalcifications in plaque rupture. APL Bioeng 2023; 7:036120. [PMID: 37786532 PMCID: PMC10541963 DOI: 10.1063/5.0168087] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 09/18/2023] [Indexed: 10/04/2023] Open
Abstract
Rupture of the cap of an atherosclerotic plaque can lead to thrombotic cardiovascular events. It has been suggested, through computational models, that the presence of microcalcifications in the atherosclerotic cap can increase the risk of cap rupture. However, the experimental confirmation of this hypothesis is still lacking. In this study, we have developed a novel tissue-engineered model to mimic the atherosclerotic fibrous cap with microcalcifications and assess the impact of microcalcifications on cap mechanics. First, human carotid plaque caps were analyzed to determine the distribution, size, and density of microcalcifications in real cap tissue. Hydroxyapatite particles with features similar to real cap microcalcifications were used as microcalcification mimics. Injected clusters of hydroxyapatite particles were embedded in a fibrin gel seeded with human myofibroblasts which deposited a native-like collagenous matrix around the particles, during the 21-day culture period. Second harmonic multiphoton microscopy imaging revealed higher local collagen fiber dispersion in regions of hydroxyapatite clusters. Tissue-engineered caps with hydroxyapatite particles demonstrated lower stiffness and ultimate tensile stress than the control group samples under uniaxial tensile loading, suggesting increased rupture risk in atherosclerotic plaques with microcalcifications. This model supports previous computational findings regarding a detrimental role for microcalcifications in cap rupture risk and can further be deployed to elucidate tissue mechanics in pathologies with calcifying soft tissues.
Collapse
Affiliation(s)
- Imke Jansen
- Department of Biomedical Engineering, Thorax Center Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Hanneke Crielaard
- Department of Biomedical Engineering, Thorax Center Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Tamar Wissing
- Department of Biomedical Engineering, Thorax Center Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | | | | | | | - Eric Farrell
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Kim van der Heiden
- Department of Biomedical Engineering, Thorax Center Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
47
|
Zhao XK, Zhu MM, Wang SN, Zhang TT, Wei XN, Wang CY, Zheng J, Zhu WY, Jiang MX, Xu SW, Yang XX, Duan YJ, Zhang BC, Han JH, Miao QR, Hu H, Chen YL. Transcription factor 21 accelerates vascular calcification in mice by activating the IL-6/STAT3 signaling pathway and the interplay between VSMCs and ECs. Acta Pharmacol Sin 2023; 44:1625-1636. [PMID: 36997664 PMCID: PMC10374894 DOI: 10.1038/s41401-023-01077-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 03/13/2023] [Indexed: 03/31/2023]
Abstract
Vascular calcification is caused by the deposition of calcium salts in the intimal or tunica media layer of the aorta, which increases the risk of cardiovascular events and all-cause mortality. However, the mechanisms underlying vascular calcification are not fully clarified. Recently it has been shown that transcription factor 21 (TCF21) is highly expressed in human and mouse atherosclerotic plaques. In this study we investigated the role of TCF21 in vascular calcification and the underlying mechanisms. In carotid artery atherosclerotic plaques collected from 6 patients, we found that TCF21 expression was upregulated in calcific areas. We further demonstrated TCF21 expression was increased in an in vitro vascular smooth muscle cell (VSMC) osteogenesis model. TCF21 overexpression promoted osteogenic differentiation of VSMC, whereas TCF21 knockdown in VSMC attenuated the calcification. Similar results were observed in ex vivo mouse thoracic aorta rings. Previous reports showed that TCF21 bound to myocardin (MYOCD) to inhibit the transcriptional activity of serum response factor (SRF)-MYOCD complex. We found that SRF overexpression significantly attenuated TCF21-induced VSMC and aortic ring calcification. Overexpression of SRF, but not MYOCD, reversed TCF21-inhibited expression of contractile genes SMA and SM22. More importantly, under high inorganic phosphate (3 mM) condition, SRF overexpression reduced TCF21-induced expression of calcification-related genes (BMP2 and RUNX2) as well as vascular calcification. Moreover, TCF21 overexpression enhanced IL-6 expression and downstream STAT3 activation to facilitate vascular calcification. Both LPS and STAT3 could induce TCF21 expression, suggesting that the inflammation and TCF21 might form a positive feedback loop to amplify the activation of IL-6/STAT3 signaling pathway. On the other hand, TCF21 induced production of inflammatory cytokines IL-1β and IL-6 in endothelial cells (ECs) to promote VSMC osteogenesis. In EC-specific TCF21 knockout (TCF21ECKO) mice, VD3 and nicotine-induced vascular calcification was significantly reduced. Our results suggest that TCF21 aggravates vascular calcification by activating IL-6/STAT3 signaling and interplay between VSMC and EC, which provides new insights into the pathogenesis of vascular calcification. TCF21 enhances vascular calcification by activating the IL-6-STAT3 signaling pathway. TCF21 inhibition may be a new potential therapeutic strategy for the prevention and treatment of vascular calcification.
Collapse
Affiliation(s)
- Xiao-Kang Zhao
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Meng-Meng Zhu
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Sheng-Nan Wang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Ting-Ting Zhang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Xiao-Ning Wei
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Cheng-Yi Wang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Juan Zheng
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Wen-Ya Zhu
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Mei-Xiu Jiang
- The Institute of Translational Medicine, the National Engineering Research Center for Bioengineering Drugs and the Technologies, Nanchang University, Nanchang, 330031, China
| | - Suo-Wen Xu
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, China
- School of Pharmacy, Bengbu Medical College, Bengbu, 233000, China
| | - Xiao-Xiao Yang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Ya-Jun Duan
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, China
| | - Bu-Chun Zhang
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, China
| | - Ji-Hong Han
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
- College of Life Sciences, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, 300071, China
| | - Qing R Miao
- Diabetes and Obesity Research Center, New York University Long Island School of Medicine, New York, NY, USA
| | - Hao Hu
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, China.
| | - Yuan-Li Chen
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China.
| |
Collapse
|
48
|
Caiazzo G, Di Mario C, Kedhi E, De Luca G. Current Management of Highly Calcified Coronary Lesions: An Overview of the Current Status. J Clin Med 2023; 12:4844. [PMID: 37510959 PMCID: PMC10381772 DOI: 10.3390/jcm12144844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/16/2023] [Accepted: 07/21/2023] [Indexed: 07/30/2023] Open
Abstract
The amount of coronary calcium strongly correlates with the degree of atherosclerosis and, therefore, with the rate of future cardiac events. Calcified coronary lesions still represent a challenge for interventional cardiologists, bringing not only a higher risk of immediate complications during percutaneous coronary interventions (PCI), but also a higher risk of late stent failure due to under-expansion and/or malapposition, and therefore, have a relevant prognostic impact. Accurate identification of the calcified plaques together with the analysis of their distribution pattern within the vessel wall by intracoronary imaging is important to improve the successful treatment of these lesions. The aim of this review is to guide readers through the assessment of the calcified plaque distribution using intracoronary imaging in order to select the best devices and strategies for plaque debulking and lesion preparation.
Collapse
Affiliation(s)
- Gianluca Caiazzo
- ICCU, San Giuseppe Moscati Hospital, ASL CE, 81031 Aversa, Italy
| | - Carlo Di Mario
- Structural Interventional Cardiology, Careggi University Hospital, 50134 Florence, Italy
| | - Elvin Kedhi
- Erasmus Hospital, Université libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Giuseppe De Luca
- Division of Cardiology, AOU Policlinico G Martino, 98124 Messina, Italy
- IRCCS Galeazzi-Sant'Ambrogio Hospital, 20157 Milan, Italy
| |
Collapse
|
49
|
Witz A, Effertz D, Goebel N, Schwab M, Franke UFW, Torzewski M. Pro-Calcifying Role of Enzymatically Modified LDL (eLDL) in Aortic Valve Sclerosis via Induction of IL-6 and IL-33. Biomolecules 2023; 13:1091. [PMID: 37509127 PMCID: PMC10377083 DOI: 10.3390/biom13071091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 06/25/2023] [Accepted: 07/03/2023] [Indexed: 07/30/2023] Open
Abstract
One of the contributors to atherogenesis is enzymatically modified LDL (eLDL). eLDL was detected in all stages of aortic valve sclerosis and was demonstrated to trigger the activation of p38 mitogen-activated protein kinase (p38 MAPK), which has been identified as a pro-inflammatory protein in atherosclerosis. In this study, we investigated the influence of eLDL on IL-6 and IL-33 induction, and also the impact of eLDL on calcification in aortic valve stenosis (AS). eLDL upregulated phosphate-induced calcification in valvular interstitial cells (VICs)/myofibroblasts isolated from diseased aortic valves, as demonstrated by alizarin red staining. Functional studies demonstrated activation of p38 MAPK as well as an altered gene expression of osteogenic genes known to be involved in vascular calcification. In parallel with the activation of p38 MAPK, eLDL also induced upregulation of the cytokines IL-6 and IL-33. The results suggest a pro-calcifying role of eLDL in AS via induction of IL-6 and IL-33.
Collapse
Affiliation(s)
- Annemarie Witz
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376 Stuttgart, Germany
| | - Denise Effertz
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376 Stuttgart, Germany
| | - Nora Goebel
- Department of Cardiovascular Surgery, Robert-Bosch-Hospital, 70376 Stuttgart, Germany
| | - Matthias Schwab
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376 Stuttgart, Germany
- Department of Clinical Pharmacology, University of Tuebingen, 72076 Tuebingen, Germany
- Department of Biochemistry and Pharmacy, University of Tuebingen, 72076 Tuebingen, Germany
| | - Ulrich F W Franke
- Department of Cardiovascular Surgery, Robert-Bosch-Hospital, 70376 Stuttgart, Germany
| | - Michael Torzewski
- Department of Laboratory Medicine and Hospital Hygiene, Robert-Bosch-Hospital, 70376 Stuttgart, Germany
| |
Collapse
|
50
|
Neels JG, Gollentz C, Chinetti G. Macrophage death in atherosclerosis: potential role in calcification. Front Immunol 2023; 14:1215612. [PMID: 37469518 PMCID: PMC10352763 DOI: 10.3389/fimmu.2023.1215612] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 06/20/2023] [Indexed: 07/21/2023] Open
Abstract
Cell death is an important aspect of atherosclerotic plaque development. Insufficient efferocytosis of death cells by phagocytic macrophages leads to the buildup of a necrotic core that impacts stability of the plaque. Furthermore, in the presence of calcium and phosphate, apoptotic bodies resulting from death cells can act as nucleation sites for the formation of calcium phosphate crystals, mostly in the form of hydroxyapatite, which leads to calcification of the atherosclerotic plaque, further impacting plaque stability. Excessive uptake of cholesterol-loaded oxidized LDL particles by macrophages present in atherosclerotic plaques leads to foam cell formation, which not only reduces their efferocytosis capacity, but also can induce apoptosis in these cells. The resulting apoptotic bodies can contribute to calcification of the atherosclerotic plaque. Moreover, other forms of macrophage cell death, such as pyroptosis, necroptosis, parthanatos, and ferroptosis can also contribute by similar mechanisms to plaque calcification. This review focuses on macrophage death in atherosclerosis, and its potential role in calcification. Reducing macrophage cell death and/or increasing their efferocytosis capacity could be a novel therapeutic strategy to reduce the formation of a necrotic core and calcification and thereby improving atherosclerotic plaque stability.
Collapse
Affiliation(s)
- Jaap G. Neels
- Université Côte d’Azur, Institut national de la santé et de la recherche médicale (INSERM), Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France
| | - Claire Gollentz
- Université Côte d’Azur, Centre Hospitalier Universitaire (CHU), Institut national de la santé et de la recherche médicale (NSERM), Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France
| | - Giulia Chinetti
- Université Côte d’Azur, Centre Hospitalier Universitaire (CHU), Institut national de la santé et de la recherche médicale (NSERM), Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France
| |
Collapse
|