1
|
Herdiana Y, Levita J, Jiranusornkul S. Chitosan-Based Nanoparticles Targeted Delivery System: In Treatment Approach for Dyslipidemia. Int J Nanomedicine 2025; 20:6611-6636. [PMID: 40438188 PMCID: PMC12118489 DOI: 10.2147/ijn.s517492] [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: 01/14/2025] [Accepted: 03/20/2025] [Indexed: 06/01/2025] Open
Abstract
Hyperlipidemia, characterized by abnormally high lipid levels in the bloodstream, is a significant risk factor for cardiovascular diseases. Conventional treatments have limitations in efficacy and may lead to side effects. Nanotechnology offers unique advantages in drug delivery, including improved drug stability, prolonged circulation time, and enhanced tissue targeting. Using nanoparticles as carriers, therapeutic agents can be precisely delivered to the target site, such as the liver or arterial walls, where lipid metabolism occurs. Chitosan nanoparticles represent an advanced approach engineered with precision to target atherosclerotic plaques. They have dual functionalities, serving therapeutic and diagnostic purposes in managing atherosclerosis. Targeting strategies involve coating nanoparticles with ligands or antibodies that recognize specific receptors overexpressed in hyperlipidemic conditions. This selective uptake maximizes the therapeutic effect while minimizing off-target effects, making it a promising alternative to traditional treatments. The review provides an overview of recent research developments for managing dyslipidemia based on the molecular target pathway of dyslipidemia, focusing on Chitosan-based delivery systems that allow controlled drug release, targeting, and enhancing patient compliance.
Collapse
Affiliation(s)
- Yedi Herdiana
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, West Java, 45363, Indonesia
| | - Jutti Levita
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, West Java, 45363, Indonesia
| | - Supat Jiranusornkul
- Department of Pharmaceutical Science, Faculty of Pharmacy, Chiang Mai University, Chiang Mai, Thailand
- Clinical Research Center for Food and Herbal Product Trials and Development, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| |
Collapse
|
2
|
Sakamoto A, Grogan A, Kawakami R, Finn A, Shah P, Nair D, Batra K, Bailen C, Sakamoto M, Virmani R, Finn AV. Role of Hemoglobin-Stimulated Macrophages and Intraplaque Hemorrhage in the Development of Vascular Diseases. Arterioscler Thromb Vasc Biol 2025. [PMID: 40308195 DOI: 10.1161/atvbaha.125.321439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2025]
Abstract
Intraplaque hemorrhage plays a critical role in the life of advancing atherosclerotic plaques, not only by triggering an acute increase in lesion size but also by attracting macrophages to the site. Lysis of erythrocytes in these areas is thought to be caused by oxidative stress, which induces the release of free Hb (hemoglobin), which is quickly bound by haptoglobin to form Hb-haptoglobin complexes. Macrophages are the only cells in the body capable of scavenging these complexes through the CD (cluster of differentiation) 163 scavenger receptor, which mediates Hb-haptoglobin ingestion, driving their differentiation. Emerging data suggest that these Hb-stimulated macrophages play an essential role in responding to intraplaque hemorrhage through mediating iron metabolism and influencing other cell types, including endothelial and smooth muscle cells. This review focuses on the role of Hb-stimulated macrophages in promoting atherogenesis through their effects on (1) endothelial activation, neoangiogenesis, and vascular permeability; (2) endothelial-to-mesenchymal cell transition and subsequent apoptosis; and (3) the prevention of smooth muscle cell osteogenic transformation and calcification. These functions may also be relevant to other vascular diseases where erythrocyte accumulation drives the formation of Hb-stimulated macrophages, which is a fundamental response to hemorrhage no matter the clinical setting.
Collapse
Affiliation(s)
- Atsushi Sakamoto
- CVPath Institute, Inc, Gaithersburg, MD (A.S., A.G., R.K., A.F., P.S., D.N., K.B., C.B., M.S., R.V., A.V.F.)
- Hamamatsu University School of Medicine, Shizuoka, Japan (A.S.)
| | - Alyssa Grogan
- CVPath Institute, Inc, Gaithersburg, MD (A.S., A.G., R.K., A.F., P.S., D.N., K.B., C.B., M.S., R.V., A.V.F.)
| | - Rika Kawakami
- CVPath Institute, Inc, Gaithersburg, MD (A.S., A.G., R.K., A.F., P.S., D.N., K.B., C.B., M.S., R.V., A.V.F.)
| | - Alexandra Finn
- CVPath Institute, Inc, Gaithersburg, MD (A.S., A.G., R.K., A.F., P.S., D.N., K.B., C.B., M.S., R.V., A.V.F.)
| | - Palak Shah
- CVPath Institute, Inc, Gaithersburg, MD (A.S., A.G., R.K., A.F., P.S., D.N., K.B., C.B., M.S., R.V., A.V.F.)
| | - Diya Nair
- CVPath Institute, Inc, Gaithersburg, MD (A.S., A.G., R.K., A.F., P.S., D.N., K.B., C.B., M.S., R.V., A.V.F.)
| | - Krish Batra
- CVPath Institute, Inc, Gaithersburg, MD (A.S., A.G., R.K., A.F., P.S., D.N., K.B., C.B., M.S., R.V., A.V.F.)
| | - Caroline Bailen
- CVPath Institute, Inc, Gaithersburg, MD (A.S., A.G., R.K., A.F., P.S., D.N., K.B., C.B., M.S., R.V., A.V.F.)
| | - Mirai Sakamoto
- CVPath Institute, Inc, Gaithersburg, MD (A.S., A.G., R.K., A.F., P.S., D.N., K.B., C.B., M.S., R.V., A.V.F.)
| | - Renu Virmani
- CVPath Institute, Inc, Gaithersburg, MD (A.S., A.G., R.K., A.F., P.S., D.N., K.B., C.B., M.S., R.V., A.V.F.)
| | - Aloke V Finn
- CVPath Institute, Inc, Gaithersburg, MD (A.S., A.G., R.K., A.F., P.S., D.N., K.B., C.B., M.S., R.V., A.V.F.)
- University of Maryland School of Medicine, Baltimore (A.V.F.)
| |
Collapse
|
3
|
Mori K, Umeno T, Kawashima T, Shuto T, Iwai R, Teng L, Tajikawa T, Nakayama Y, Miyamoto S. Six-Month Patency of Long Carotid Bypass Grafts Constructed with In-Body Tissue Architecture-Induced Small-Diameter Biotubes in a Goat Model. Bioengineering (Basel) 2025; 12:260. [PMID: 40150724 PMCID: PMC11939719 DOI: 10.3390/bioengineering12030260] [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: 02/09/2025] [Revised: 03/02/2025] [Accepted: 03/03/2025] [Indexed: 03/29/2025] Open
Abstract
This study investigated the long-term patency of regenerative Biotube grafts and discusses their feasibility as an alternative to autologous vein grafts for peripheral artery disease. Six Biotubes with a diameter of 4 mm were autologously fabricated in recipients using in vivo tissue engineering (in-body tissue architecture) technology and implanted as carotid artery bypass grafts in a goat model. All six grafts remained patent at 6 months despite exceeding 10 cm in length, demonstrating their biocompatibility and durability. Histological analysis revealed neointima formation, endothelialization, and minimal inflammation. However, in one goat, a graft developed stenosis, while another showed dilatation. These findings demonstrate the use of Biotubes as a viable option for peripheral vascular reconstruction as tissue-engineered vascular grafts. However, further optimization is needed to address emerging issues with their use, such as stenosis and aneurysm formation, to improve long-term patency.
Collapse
Affiliation(s)
- Kazuki Mori
- Department of Cardiovascular Surgery, Oita University, Yufu 879-5593, Japan; (T.U.); (T.K.); (T.S.); (S.M.)
| | - Tadashi Umeno
- Department of Cardiovascular Surgery, Oita University, Yufu 879-5593, Japan; (T.U.); (T.K.); (T.S.); (S.M.)
| | - Takayuki Kawashima
- Department of Cardiovascular Surgery, Oita University, Yufu 879-5593, Japan; (T.U.); (T.K.); (T.S.); (S.M.)
| | - Takashi Shuto
- Department of Cardiovascular Surgery, Oita University, Yufu 879-5593, Japan; (T.U.); (T.K.); (T.S.); (S.M.)
| | - Ryosuke Iwai
- Institute of Frontier Science and Technology, Okayama University of Science, Okayama 700-0005, Japan; (R.I.); (L.T.)
| | - Lupeng Teng
- Institute of Frontier Science and Technology, Okayama University of Science, Okayama 700-0005, Japan; (R.I.); (L.T.)
| | - Tsutomu Tajikawa
- Department of Mechanical Engineering, Faculty of Engineering Science, Kansai University, Osaka 564-8680, Japan;
| | | | - Shinji Miyamoto
- Department of Cardiovascular Surgery, Oita University, Yufu 879-5593, Japan; (T.U.); (T.K.); (T.S.); (S.M.)
| |
Collapse
|
4
|
Wang L, Zhang Y, Ji F, Si Z, Liu C, Wu X, Wang C, Chang H. Identification of crucial genes for polycystic ovary syndrome and atherosclerosis through comprehensive bioinformatics analysis and machine learning. Int J Gynaecol Obstet 2025. [PMID: 39981695 DOI: 10.1002/ijgo.70014] [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: 09/26/2024] [Revised: 01/23/2025] [Accepted: 02/03/2025] [Indexed: 02/22/2025]
Abstract
OBJECTIVE To identify potential biomarkers in patients with polycystic ovary syndrome (PCOS) and atherosclerosis, and to explore the common pathologic mechanisms between these two diseases in response to the increased risk of cardiovascular diseases in patients with PCOS. METHODS PCOS and atherosclerosis data sets were downloaded from the GEO database, and their differentially expressed genes were identified. Weighted gene co-expression network analysis was used to obtain intersection genes, and then protein-protein interaction and functional enrichment analysis were performed. Machine learning algorithms were used to select the key genes, which were then validated through external data sets. We constructed a nomogram to predict the risk of atherosclerosis in women with PCOS. Finally, the CIBERSORT algorithm was used to analyze the infiltration of immune cells in the atherosclerosis group. RESULTS We identified six hub genes (CD163, LAPTM5, TNFSF13B, MS4A4A, FGR, and IRF1) that exhibited excellent diagnostic value in validation data sets. Gene ontology terms and KEGG signaling pathway analysis revealed that key genes were associated with immune responses and inflammatory reactions. Abnormal immune cell infiltration was also found in the atherosclerosis group and was correlated with the six hub genes. CONCLUSION Common therapeutic targets of PCOS and atherosclerosis were preliminarily identified through bioinformatics analysis and machine learning techniques. These findings provide new treatment ideas for reducing the risk that PCOS will develop into atherosclerosis.
Collapse
Affiliation(s)
- Lirong Wang
- The First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yanli Zhang
- The First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Fan Ji
- The First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Zhenmin Si
- The First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Chengdong Liu
- Department of Traditional Chinese Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xiaoke Wu
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
- Department of Obstetrics and Gynecology, Heilongjiang Provincial Hospital, Harbin, China
| | - Chichiu Wang
- Department of Obstetrics and Gynecology, The Chinese University of Hong Kong, Hong Kong, China
| | - Hui Chang
- The First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
5
|
Roh T, Ju S, Park SY, Ahn Y, Chung J, Nakano M, Ryu G, Kim YJ, Kim G, Choi H, Lee SG, Kim IS, Lee SI, Chung C, Shimizu T, Miyoshi E, Jung SS, Park C, Yamasaki S, Park SY, Jo EK. Fucosylated haptoglobin promotes inflammation via Mincle in sepsis: an observational study. Nat Commun 2025; 16:1342. [PMID: 39904983 PMCID: PMC11794430 DOI: 10.1038/s41467-025-56524-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 01/22/2025] [Indexed: 02/06/2025] Open
Abstract
Haptoglobin (Hp) scavenges cell-free hemoglobin and correlates with the prognosis of human sepsis, a life-threatening systemic inflammatory condition. Despite extensive research on Hp glycosylation as a glyco-biomarker for cancers, understanding glycosylated modifications of Hp in sepsis patients (SPs) remains limited. Our study reveals elevated levels of terminal fucosylation at Asn207 and Asn211 of Hp in SP plasma, along with heightened inflammatory responses, compared to healthy controls (trial registration NCT05911711). Fucosylated (Fu)-Hp purified from SPs upregulates inflammatory cytokines and chemokines, along with NLRP3 inflammasome activation. Single-cell RNA sequencing identifies a distinct macrophage-like cell population with increased expressions of inflammatory mediators and FUT4 in response to Fu-Hp. Additionally, Mincle, a C-type lectin receptor, interacts with Fu-Hp to amplify the inflammatory responses and signaling. Moreover, the Hp fucosylation (AAL) level significantly correlates with the levels of inflammatory cytokines in sepsis patients, suggesting that Fu-Hp is clinically relevant. Finally, Fu-Hp treatment significantly enhances the levels of inflammatory cytokines in the plasma and various tissues of mice. Together, our findings reveal a role of Fu-Hp, derived from sepsis patients, in driving inflammation, and suggest that targeting Fu-Hp could serve as a promising intervention for combating sepsis. Trial registration NCT05911711.
Collapse
Affiliation(s)
- Taylor Roh
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
- Brain Korea 21 FOUR Project for Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Sungeun Ju
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - So Young Park
- Division of Pulmonary, Allergy and Critical Care Medicine, Kangdong Sacred Heart Hospital, Seoul, Republic of Korea.
| | - Yeonghwan Ahn
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Jiyun Chung
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Miyako Nakano
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan
| | - Gyoungah Ryu
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Young Jae Kim
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
- Brain Korea 21 FOUR Project for Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Geumseo Kim
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
- Brain Korea 21 FOUR Project for Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Hyewon Choi
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Sung-Gwon Lee
- Section of Genetics and Physiology, Laboratory of Molecular and Cellular Biology, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, MD, USA
| | - In Soo Kim
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
- Brain Korea 21 FOUR Project for Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Song-I Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chungnam National University Hospital, Daejeon, Republic of Korea
| | - Chaeuk Chung
- Division of Pulmonology and Critical Care Medicine, Department of Internal Medicine, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Takashi Shimizu
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
- Laboratory of Molecular Immunology, Immunology Frontier Research Center (IFReC), Osaka University, Suita, Osaka, Japan
| | - Eiji Miyoshi
- Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Sung-Soo Jung
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Chungoo Park
- School of Biological Sciences and Technology, Chonnam National University, Gwangju, Republic of Korea
| | - Sho Yamasaki
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
- Laboratory of Molecular Immunology, Immunology Frontier Research Center (IFReC), Osaka University, Suita, Osaka, Japan
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, Osaka, Japan
| | - Seung-Yeol Park
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea.
| | - Eun-Kyeong Jo
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon, Republic of Korea.
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea.
| |
Collapse
|
6
|
Yan W, Li Y, Wang G, Huang Y, Xie P. Clinical application and immune infiltration landscape of stemness-related genes in heart failure. ESC Heart Fail 2025; 12:250-270. [PMID: 39275894 PMCID: PMC11769652 DOI: 10.1002/ehf2.15055] [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: 12/31/2023] [Revised: 07/17/2024] [Accepted: 08/21/2024] [Indexed: 09/16/2024] Open
Abstract
BACKGROUND Heart failure (HF) is the leading cause of morbidity and mortality worldwide. Stemness refers to the self-renewal and differentiation ability of cells. However, little is known about the heart's stemness properties. Thus, the current study aims to identify putative stemness-related biomarkers to construct a viable prediction model of HF and characterize the immune infiltration features of HF. METHODS HF datasets from the Gene Expression Omnibus (GEO) database were adopted as the training and validation cohorts while stemness-related genes were obtained from GeneCards and previously published papers. Feature selection was performed using two machine learning algorithms. Nomogram models were then constructed to predict HF risk based on the selected key genes. Moreover, the biological functions of the key genes were evaluated using Gene Ontology (GO) and Kyoto Encyclopedia of Genes Genomes (KEGG) pathway analyses, and gene set variation analysis (GSVA) and enrichment analysis (GSEA) were performed between the high- and low-risk groups. The immune infiltration landscape in HF was investigated, and the interaction network of key genes was analysed to predict potential targets and molecular mechanisms. RESULTS Seven key genes, namely SMOC2, LUM, FNDC1, SCUBE2, CD163, BLM and S1PR3, were included in the proposed nomogram. This nomogram showed good predictive performance for HF diagnosis in the training and validation sets. GO and KEGG analyses revealed that the key genes were primarily associated with ageing, inflammatory processes and DNA oxidation. GSEA and GSVA identified various inflammatory and immune signalling pathways that were enriched between the high- and low-risk groups. The infiltration of 15 immune cell subsets suggests that adaptive immunity has an important role in HF. CONCLUSIONS Our study identified a clinically significant stemness-related signature for predicting HF risk, with the potential to improve early disease diagnosis, optimize risk stratification and provide new strategies for treating patients with HF.
Collapse
Affiliation(s)
- Wenting Yan
- Gansu University of Traditional Chinese MedicineLanzhouChina
| | - Yanling Li
- Department of CardiologyGansu Provincial HospitalLanzhouChina
| | - Gang Wang
- First Clinical Medical College of Lanzhou UniversityLanzhouChina
| | - Yuan Huang
- Gansu University of Traditional Chinese MedicineLanzhouChina
| | - Ping Xie
- Department of CardiologyGansu Provincial HospitalLanzhouChina
| |
Collapse
|
7
|
Gastanadui MG, Margaroli C, Litovsky S, Richter RP, Wang D, Xing D, Wells JM, Gaggar A, Nanda V, Patel RP, Payne GA. Spatial Transcriptomic Approach to Understanding Coronary Atherosclerotic Plaque Stability. Arterioscler Thromb Vasc Biol 2024; 44:e264-e276. [PMID: 39234691 PMCID: PMC11499036 DOI: 10.1161/atvbaha.123.320330] [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: 10/26/2023] [Accepted: 08/08/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND Coronary atherosclerotic plaques susceptible to acute coronary syndrome have traditionally been characterized by their surrounding cellular architecture. However, with the advent of intravascular imaging, novel mechanisms of coronary thrombosis have emerged, challenging our contemporary understanding of acute coronary syndrome. These intriguing findings underscore the necessity for a precise molecular definition of plaque stability. Considering this, our study aimed to investigate the vascular microenvironment in patients with stable and unstable plaques using spatial transcriptomics. METHODS Autopsy-derived coronary arteries were preserved and categorized by plaque stability (n=5 patients per group). We utilized the GeoMx spatial profiling platform and Whole Transcriptome Atlas to link crucial histological morphology markers in coronary lesions with differential gene expression in specific regions of interest, thereby mapping the vascular transcriptome. This innovative approach allowed us to conduct cell morphological and spatially resolved transcriptional profiling of atherosclerotic plaques while preserving crucial intercellular signaling. RESULTS We observed intriguing spatial and cell-specific transcriptional patterns in stable and unstable atherosclerotic plaques, showcasing regional variations within the intima and media. These regions exhibited differential expression of proinflammatory molecules (eg, IFN-γ [interferon-γ], MHC [major histocompatibility complex] class II, proinflammatory cytokines) and prothrombotic signaling pathways. By using lineage tracing through spatial deconvolution of intimal CD68+ (cluster of differentiation 68) cells, we characterized unique, intraplaque subpopulations originating from endothelial, smooth muscle, and myeloid lineages with distinct regional locations associated with plaque instability. In addition, unique transcriptional signatures were observed in vascular smooth muscle and CD68+ cells among plaques exhibiting coronary calcification. CONCLUSIONS Our study illuminates distinct cell-specific and regional transcriptional alterations present in unstable plaques. Furthermore, we characterize spatially resolved, in situ evidence supporting cellular transdifferentiation and intraplaque plasticity as significant contributors to plaque instability in human coronary atherosclerosis. Our results provide a powerful resource for the identification of novel mediators of acute coronary syndrome, opening new avenues for preventative and therapeutic treatments.
Collapse
Affiliation(s)
- Maria G Gastanadui
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL, USA
- Cardiopulmonary Research Program, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Camilla Margaroli
- Department of Pathology, Division of Molecular & Cellular Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
- Program in Protease/Matrix Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Silvio Litovsky
- Department of Pathology, Division of Anatomic Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
- Comprehensive Cardiovascular Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Robert P. Richter
- Program in Protease/Matrix Biology, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Pediatrics, Division of Pediatric Critical Care, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Dezhi Wang
- Department of Pathology, Pathology Core Research Laboratory, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Dongqi Xing
- Cardiopulmonary Research Program, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Program in Protease/Matrix Biology, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Medicine, Division of Pulmonary, Allergy & Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Lung Health Center, University of Alabama at Birmingham, Birmingham, AL
- Vascular Biology and Hypertension Program, University of Alabama at Birmingham, Birmingham, AL, USA
| | - J. Michael Wells
- Cardiopulmonary Research Program, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Program in Protease/Matrix Biology, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Medicine, Division of Pulmonary, Allergy & Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Lung Health Center, University of Alabama at Birmingham, Birmingham, AL
- Vascular Biology and Hypertension Program, University of Alabama at Birmingham, Birmingham, AL, USA
- Medical Service at Birmingham VA Medical Center, Birmingham, AL
| | - Amit Gaggar
- Cardiopulmonary Research Program, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Program in Protease/Matrix Biology, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Medicine, Division of Pulmonary, Allergy & Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Lung Health Center, University of Alabama at Birmingham, Birmingham, AL
- Vascular Biology and Hypertension Program, University of Alabama at Birmingham, Birmingham, AL, USA
- Medical Service at Birmingham VA Medical Center, Birmingham, AL
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Vivek Nanda
- Department of Pathology, Division of Molecular & Cellular Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Rakesh P. Patel
- Cardiopulmonary Research Program, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Pathology, Division of Molecular & Cellular Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
- Program in Protease/Matrix Biology, University of Alabama at Birmingham, Birmingham, AL, USA
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Gregory A. Payne
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL, USA
- Cardiopulmonary Research Program, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Program in Protease/Matrix Biology, University of Alabama at Birmingham, Birmingham, AL, USA
- Comprehensive Cardiovascular Center, University of Alabama at Birmingham, Birmingham, AL, USA
- Vascular Biology and Hypertension Program, University of Alabama at Birmingham, Birmingham, AL, USA
- Medical Service at Birmingham VA Medical Center, Birmingham, AL
| |
Collapse
|
8
|
Consonni FM, Incerti M, Bertolotti M, Ballerini G, Garlatti V, Sica A. Heme catabolism and heme oxygenase-1-expressing myeloid cells in pathophysiology. Front Immunol 2024; 15:1433113. [PMID: 39611159 PMCID: PMC11604077 DOI: 10.3389/fimmu.2024.1433113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 10/01/2024] [Indexed: 11/30/2024] Open
Abstract
Although the pathological significance of myeloid cell heterogeneity is still poorly understood, new evidence indicates that distinct macrophage subsets are characterized by specific metabolic programs that influence disease onset and progression. Within this scenario, distinct subsets of macrophages, endowed with high rates of heme catabolism by the stress-responsive enzyme heme oxygenase-1 (HO-1), play critical roles in physiologic and pathological conditions. Of relevance, the substrates of HO-1 activity are the heme groups that derive from cellular catabolism and are converted into carbon monoxide (CO), biliverdin and Fe2+, which together elicit anti-apoptotic, anti-inflammatory activities and control oxidative damage. While high levels of expression of HO-1 enzyme by specialized macrophage populations (erythrophagocytes) guarantee the physiological disposal of senescent red blood cells (i.e. erythrocateresis), the action of HO-1 takes on pathological significance in various diseases, and abnormal CO metabolism has been observed in cancer, hematological diseases, hypertension, heart failure, inflammation, sepsis, neurodegeneration. Modulation of heme catabolism and CO production is therefore a feasible therapeutic opportunity in various diseases. In this review we discuss the role of HO-1 in different pathological contexts (i.e. cancer, infections, cardiovascular, immune-mediated and neurodegenerative diseases) and highlight new therapeutic perspectives on the modulation of the enzymatic activity of HO-1.
Collapse
Affiliation(s)
- Francesca Maria Consonni
- Department of Pharmaceutical Sciences, University of Piemonte Orientale “A. Avogadro”, Novara, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Martina Incerti
- Department of Pharmaceutical Sciences, University of Piemonte Orientale “A. Avogadro”, Novara, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Milena Bertolotti
- Navita S.r.l., University of Eastern Piedmont A. Avogadro, Novara, Italy
| | - Giulia Ballerini
- Department of Pharmaceutical Sciences, University of Piemonte Orientale “A. Avogadro”, Novara, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Valentina Garlatti
- Department of Pharmaceutical Sciences, University of Piemonte Orientale “A. Avogadro”, Novara, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Antonio Sica
- Department of Pharmaceutical Sciences, University of Piemonte Orientale “A. Avogadro”, Novara, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| |
Collapse
|
9
|
Obare LM, Bailin SS, Zhang X, Nthenge K, Priest S, Liu Q, Stolze LK, Sheng Q, Gangula R, Behrens M, Jenkins B, Prasad P, Neikirk K, Prakash P, Hogan M, Zhang L, Beasley HK, Shao J, Miller-Fleming TW, Actkins KV, Phillips MA, Hubert D, Malone J, Labeeb C, Gelbard A, Chaillon A, Mashayekhi M, Gabriel CL, Temu T, Olson L, Jones A, Beeri K, Baker P, Kawai K, Ghosh SKB, Guo L, Virmani R, Finn A, Shah P, Yang TS, Bick AG, Kirabo A, Su YR, Phillips EJ, Mallal S, Dash C, Koethe JR, Gianella S, McReynolds MR, Absi T, Hinton A, Wanjalla CN. HIV persists in late coronary atheroma and is associated with increased local inflammation and disease progression. RESEARCH SQUARE 2024:rs.3.rs-5125826. [PMID: 39483879 PMCID: PMC11527356 DOI: 10.21203/rs.3.rs-5125826/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Chronic inflammation contributes to the prevalence of cardiovascular disease in people living with HIV (PLWH). The immune mechanisms driving atherosclerosis progression in PLWH remain unclear. This study conducted comprehensive assessments of medium-sized coronary arteries and aorta from deceased PLWH and controls without HIV using DNA/RNA assays, spatial transcriptomics, and high-resolution mass spectrometry. Findings revealed more significant inflammation correlated with higher HIV copy numbers in late atheroma of PLWH. Enhanced CXCL12 and decreased ABCA1/ABCG1 expression in CD163+ macrophages were co-localized in coronaries of PLWH, suggesting a reduction in plasma lipoprotein clearance compared to controls. Spatial analyses identified potential therapeutic targets by revealing inflammatory changes in medium-sized arteries and the aorta. We examined the relationship between atherosclerotic phenotypes and inflammatory gene expression in Vanderbilts Biobank to study these findings in a larger clinical cohort. This established a significant association between ABCA1 and CXCL12 gene expressions with atherosclerosis, partly influenced by HIV.
Collapse
Affiliation(s)
- Laventa M. Obare
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Samuel S. Bailin
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Xiuqi Zhang
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kisyua Nthenge
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Stephen Priest
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Qi Liu
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lindsey K Stolze
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Quanhu Sheng
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Rama Gangula
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Madelaine Behrens
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Brenita Jenkins
- Department of Biochemistry and Molecular Biology, The Huck Institute of the Life Sciences, Pennsylvania State University, State College, PA, USA
| | - Praveena Prasad
- Department of Biochemistry and Molecular Biology, The Huck Institute of the Life Sciences, Pennsylvania State University, State College, PA, USA
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Prem Prakash
- The Center for AIDS Health Disparities Research, Meharry Medical College, Nashville, TN, USA
- Department of Microbiology, Immunology, and Physiology, Meharry Medical College, Nashville, TN, USA
- Department of Biochemistry, Cancer Biology, Pharmacology and Neuroscience, Meharry Medical College, Nashville, TN, USA
| | | | - Liang Zhang
- Central Microscopy Research Facility, University of Iowa, Iowa City, IA, USA
| | - Heather K. Beasley
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Jianqiang Shao
- Central Microscopy Research Facility, University of Iowa, Iowa City, IA, USA
| | | | - Kyera V. Actkins
- Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Mark A. Phillips
- Department of Integrative Biology, Oregon State University, Corvallis, OR, USA
| | - David Hubert
- Department of Integrative Biology, Oregon State University, Corvallis, OR, USA
| | - Jordan Malone
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Cassia Labeeb
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alexander Gelbard
- Department of Otolaryngology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Antoine Chaillon
- Division of Infectious Diseases, University of California, San Diego, CA, USA
| | - Mona Mashayekhi
- Division of Diabetes, Endocrinology, and Metabolism, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Curtis L. Gabriel
- Division of Gastroenterology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Tecla Temu
- Department of Pathology, Harvard Medical School, Boston, MA, USA
| | - Lana Olson
- VANTAGE, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Angela Jones
- VANTAGE, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Karen Beeri
- VANTAGE, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Paxton Baker
- VANTAGE, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kenji Kawai
- CVPath Institute, Gaithersburg, Maryland, USA
| | | | - Laing Guo
- CVPath Institute, Gaithersburg, Maryland, USA
| | | | - Aloke Finn
- CVPath Institute, Gaithersburg, Maryland, USA
| | - Palak Shah
- CVPath Institute, Gaithersburg, Maryland, USA
| | - Tzushan Sharon Yang
- Division of Comparative Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alexander G. Bick
- Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Annet Kirabo
- The Center for AIDS Health Disparities Research, Meharry Medical College, Nashville, TN, USA
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Yan R Su
- Department of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Elizabeth J. Phillips
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
- Institute for Immunology and Infectious Diseases, Murdoch University, WA, Western Australia
| | - Simon Mallal
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
- Institute for Immunology and Infectious Diseases, Murdoch University, WA, Western Australia
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN
| | - Chandravanu Dash
- Department of Microbiology, Immunology, and Physiology, Meharry Medical College, Nashville, TN, USA
- Department of Biochemistry, Cancer Biology, Pharmacology and Neuroscience, Meharry Medical College, Nashville, TN, USA
- NanoString Technologies, Inc., Seattle, WA
| | - John R. Koethe
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sara Gianella
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Melanie R. McReynolds
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Tarek Absi
- Department of Cardiac Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Antentor Hinton
- The Center for AIDS Health Disparities Research, Meharry Medical College, Nashville, TN, USA
| | - Celestine N. Wanjalla
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
10
|
Lu AL, Yin L, Huang Y, Islam ZH, Kanchetty R, Johnston C, Zhang K, Xie X, Park KH, Chalfant CE, Wang B. The role of 6-phosphogluconate dehydrogenase in vascular smooth muscle cell phenotypic switching and angioplasty-induced intimal hyperplasia. JVS Vasc Sci 2024; 5:100214. [PMID: 39318609 PMCID: PMC11420449 DOI: 10.1016/j.jvssci.2024.100214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 07/21/2024] [Indexed: 09/26/2024] Open
Abstract
Background Restenosis poses a significant challenge for individuals afflicted with peripheral artery diseases, often leading to considerable morbidity and necessitating repeated interventions. The primary culprit behind the pathogenesis of restenosis is intimal hyperplasia (IH), in which the hyperproliferative and migratory vascular smooth muscle cell (VSMC) accumulate excessively in the tunica intima. 6-Phosphogluconate dehydrogenase (6PGD), sometimes referred to as PGD, is one of the critical enzymes in pentose phosphate pathway (PPP). In this study, we sought to probe whether 6PGD is aberrantly regulated in IH and contributes to VSMC phenotypic switching. Methods We used clinical specimens of diseased human coronary arteries with IH lesions and observed robust upregulation of 6PGD at protein level in both the medial and intimal layers in comparison with healthy arterial segments. Results 6PGD activity and protein expression were profoundly stimulated upon platelet-derived growth factor-induced VSMC phenotypic switching. Using gain-of-function (dCas9-mediated transcriptional activation) and loss-of-function (small interfering RNA-mediated) silencing, we were able to demonstrate the pathogenic role of 6PGD in driving VSMC hyperproliferation, migration, dedifferentiation, and inflammation. Finally, we conducted a rat model of balloon angioplasty in the common carotid artery, with Pluronic hydrogel-assisted perivascular delivery of Physcion, a selective 6PGD inhibitor with poor systemic bioavailability, and observed effective mitigation of IH. Conclusions We contend that aberrant 6PGD expression and activity-indicative of a metabolic shift toward pentose phosphate pathway-could serve as a new disease-driving mechanism and, hence, an actionable target for the development of effective new therapies for IH and restenosis after endovascular interventions.
Collapse
Affiliation(s)
- Amy L. Lu
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA
- Division of Hematology & Oncology, Department of Medicine, University of Virginia, Charlottesville, VA
| | - Li Yin
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA
- Department of Vascular Surgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang, China
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Yitao Huang
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA
| | - Zain Husain Islam
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA
| | - Rohan Kanchetty
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA
| | - Campbell Johnston
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA
| | - Kaijie Zhang
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA
- Department of Vascular Surgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang, China
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Xiujie Xie
- Division of Hematology & Oncology, Department of Medicine, University of Virginia, Charlottesville, VA
| | - Ki Ho Park
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA
| | - Charles E. Chalfant
- Division of Hematology & Oncology, Department of Medicine, University of Virginia, Charlottesville, VA
- Research Service, Richmond Veterans Administration Medical Center, Richmond, VA
| | - Bowen Wang
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL
| |
Collapse
|
11
|
Sun X, Jia X, Tan Z, Fan D, Chen M, Cui N, Liu A, Liu D. Oral Nanoformulations in Cardiovascular Medicine: Advances in Atherosclerosis Treatment. Pharmaceuticals (Basel) 2024; 17:919. [PMID: 39065770 PMCID: PMC11279631 DOI: 10.3390/ph17070919] [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: 05/31/2024] [Revised: 07/01/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Atherosclerosis (AS) is the formation of atherosclerotic plaques on the walls of the arteries, causing them to narrow. If this occurs in the coronary arteries, the blood vessels may be completely blocked, resulting in myocardial infarction; if it occurs in the blood vessels of the brain, the blood vessels may be blocked, resulting in cerebral infarction, i.e., stroke. Studies have shown that the pathogenesis of atherosclerosis involves the processes of inflammation, lipid infiltration, oxidative stress, and endothelial damage, etc. SIRT, as a key factor regulating the molecular mechanisms of oxidative stress, inflammation, and aging, has an important impact on the pathogenesis of plaque formation, progression, and vulnerability. Statistics show that AS accounts for about 50 per cent of deaths in Western countries. Currently, oral medication is the mainstay of AS treatment, but its development is limited by side effects, low bioavailability and other unfavourable factors. In recent years, with the rapid development of nano-preparations, researchers have combined statins and natural product drugs within nanopreparations to improve their bioavailability. Based on this, this paper summarises the main pathogenesis of AS and also proposes new oral nanoformulations such as liposomes, nanoparticles, nanoemulsions, and nanocapsules to improve their application in the treatment of AS.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Aidong Liu
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (X.S.); (X.J.); (Z.T.); (D.F.); (M.C.); (N.C.)
| | - Da Liu
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (X.S.); (X.J.); (Z.T.); (D.F.); (M.C.); (N.C.)
| |
Collapse
|
12
|
Kawai K, Kawakami R, Finn AV, Virmani R. Differences in Stable and Unstable Atherosclerotic Plaque. Arterioscler Thromb Vasc Biol 2024; 44:1474-1484. [PMID: 38924440 DOI: 10.1161/atvbaha.124.319396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2024]
Affiliation(s)
- Kenji Kawai
- Department of Pathology, CVPath Institute, Gaithersburg, MD (K.K., R.K., A.V.F., R.V.)
| | - Rika Kawakami
- Department of Pathology, CVPath Institute, Gaithersburg, MD (K.K., R.K., A.V.F., R.V.)
| | - Aloke V Finn
- Department of Pathology, CVPath Institute, Gaithersburg, MD (K.K., R.K., A.V.F., R.V.)
- University of Maryland School of Medicine, Baltimore (A.V.F.)
| | - Renu Virmani
- Department of Pathology, CVPath Institute, Gaithersburg, MD (K.K., R.K., A.V.F., R.V.)
| |
Collapse
|
13
|
Li B, Shaikh F, Zamzam A, Abdin R, Qadura M. Inflammatory Biomarkers to Predict Major Adverse Cardiovascular Events in Patients with Carotid Artery Stenosis. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:997. [PMID: 38929614 PMCID: PMC11205582 DOI: 10.3390/medicina60060997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 06/13/2024] [Accepted: 06/15/2024] [Indexed: 06/28/2024]
Abstract
Background and Objectives: Inflammatory proteins and their prognostic value in patients with carotid artery stenosis (CAS) have not been adequately studied. Herein, we identified CAS-specific biomarkers from a large pool of inflammatory proteins and assessed the ability of these biomarkers to predict adverse events in individuals with CAS. Materials and Methods: Samples of blood were prospectively obtained from 336 individuals (290 with CAS and 46 without CAS). Plasma concentrations of 29 inflammatory proteins were determined at recruitment, and the patients were followed for 24 months. The outcome of interest was a major adverse cardiovascular event (MACE; composite of stroke, myocardial infarction, or death). The differences in plasma protein concentrations between patients with vs. without a 2-year MACE were determined using the independent t-test or Mann-Whitney U test to identify CAS-specific prognostic biomarkers. Kaplan-Meier and Cox proportional hazards analyses with adjustment for baseline demographic and clinical characteristics were performed to assess the prognostic value of differentially expressed inflammatory proteins in predicting a 2-year MACE in patients with CAS. Results: The mean age of the cohort was 68.8 (SD 10.2) years and 39% were female. The plasma concentrations of two inflammatory proteins were significantly higher in individuals with a 2-year MACE relative to those without a 2-year MACE: IL-6 (5.07 (SD 4.66) vs. 3.36 (SD 4.04) pg/mL, p = 0.03) and CD163 (233.825 (SD 230.306) vs. 159.673 (SD 175.669) pg/mL, p = 0.033). Over a follow-up period of 2 years, individuals with elevated levels of IL-6 were more likely to develop MACE (HR 1.269 (95% CI 1.122-1.639), p = 0.042). Similarly, over a 2-year period, patients with high levels of CD163 were more likely to develop MACE (HR 1.413 (95% CI 1.022-1.954), p = 0.036). Conclusions: The plasma levels of inflammatory proteins IL-6 and CD163 are independently associated with adverse outcomes in individuals with CAS. These CAS-specific prognostic biomarkers may assist in the risk stratification of patients at an elevated risk of a MACE and subsequently guide further vascular evaluation, specialist referrals, and aggressive medical/surgical management, thereby improving outcomes for patients with CAS.
Collapse
Affiliation(s)
- Ben Li
- Department of Surgery, University of Toronto, Toronto, ON M5S 1A1, Canada;
- Division of Vascular Surgery, St. Michael’s Hospital, University of Toronto, Toronto, ON M5S 1A1, Canada; (F.S.); (A.Z.)
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A1, Canada
- Temerty Centre for Artificial Intelligence Research and Education in Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Farah Shaikh
- Division of Vascular Surgery, St. Michael’s Hospital, University of Toronto, Toronto, ON M5S 1A1, Canada; (F.S.); (A.Z.)
| | - Abdelrahman Zamzam
- Division of Vascular Surgery, St. Michael’s Hospital, University of Toronto, Toronto, ON M5S 1A1, Canada; (F.S.); (A.Z.)
| | - Rawand Abdin
- Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada;
| | - Mohammad Qadura
- Department of Surgery, University of Toronto, Toronto, ON M5S 1A1, Canada;
- Division of Vascular Surgery, St. Michael’s Hospital, University of Toronto, Toronto, ON M5S 1A1, Canada; (F.S.); (A.Z.)
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A1, Canada
- Li Ka Shing Knowledge Institute, St. Michael’s Hospital, University of Toronto, Toronto, ON M5S 1A1, Canada
| |
Collapse
|
14
|
Li D, Fan C, Li X, Zhao L. The role of macrophage polarization in vascular calcification. Biochem Biophys Res Commun 2024; 710:149863. [PMID: 38579535 DOI: 10.1016/j.bbrc.2024.149863] [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: 12/30/2023] [Revised: 03/25/2024] [Accepted: 03/28/2024] [Indexed: 04/07/2024]
Abstract
Vascular calcification is an important factor in the high morbidity and mortality of Cardiovascular and cerebrovascular diseases. Vascular damage caused by calcification of the intima or media impairs the physiological function of the vascular wall. Inflammation is a central factor in the development of vascular calcification. Macrophages are the main inflammatory cells. Dynamic changes of macrophages with different phenotypes play an important role in the occurrence, progression and stability of calcification. This review focuses on macrophage polarization and the relationship between macrophages of different phenotypes and calcification environment, as well as the mechanism of interaction, it is considered that macrophages can promote vascular calcification by releasing inflammatory mediators and promoting the osteogenic transdifferentiation of smooth muscle cells and so on. In addition, several therapeutic strategies aimed at macrophage polarization for vascular calcification are described, which are of great significance for targeted treatment of vascular calcification.
Collapse
Affiliation(s)
- Dan Li
- The Second Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan City, Shandong Province, China
| | - Chu Fan
- Department of Cardiology, Beijing AnZhen Hospital, Capital Medical University, Beijing City, China
| | - Xuepeng Li
- Department of Cardiology, Beijing AnZhen Hospital, Capital Medical University, Beijing City, China
| | - Lin Zhao
- The Second Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan City, Shandong Province, China; Department of Cardiology, Beijing AnZhen Hospital, Capital Medical University, Beijing City, China.
| |
Collapse
|
15
|
Zhang X, Heo GS, Li A, Lahad D, Detering L, Tao J, Gao X, Zhang X, Luehmann H, Sultan D, Lou L, Venkatesan R, Li R, Zheng J, Amrute J, Lin CY, Kopecky BJ, Gropler RJ, Bredemeyer A, Lavine K, Liu Y. Development of a CD163-Targeted PET Radiotracer That Images Resident Macrophages in Atherosclerosis. J Nucl Med 2024; 65:775-780. [PMID: 38548349 PMCID: PMC11064833 DOI: 10.2967/jnumed.123.266910] [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: 10/23/2023] [Revised: 02/26/2024] [Indexed: 05/03/2024] Open
Abstract
Tissue-resident macrophages are complementary to proinflammatory macrophages to promote the progression of atherosclerosis. The noninvasive detection of their presence and dynamic variation will be important to the understanding of their role in the pathogenesis of atherosclerosis. The goal of this study was to develop a targeted PET radiotracer for imaging CD163-positive (CD163+) macrophages in multiple mouse atherosclerosis models and assess the potential of CD163 as a biomarker for atherosclerosis in humans. Methods: CD163-binding peptide was identified using phage display and conjugated with a NODAGA chelator for 64Cu radiolabeling ([64Cu]Cu-ICT-01). CD163-overexpressing U87 cells were used to measure the binding affinity of [64Cu]Cu-ICT-01. Biodistribution studies were performed on wild-type C57BL/6 mice at multiple time points after tail vein injection. The sensitivity and specificity of [64Cu]Cu-ICT-01 in imaging CD163+ macrophages upregulated on the surface of atherosclerotic plaques were assessed in multiple mouse atherosclerosis models. Immunostaining, flow cytometry, and single-cell RNA sequencing were performed to characterize the expression of CD163 on tissue-resident macrophages. Human carotid atherosclerotic plaques were used to measure the expression of CD163+ resident macrophages and test the binding specificity of [64Cu]Cu-ICT-01. Results: [64Cu]Cu-ICT-01 showed high binding affinity to U87 cells. The biodistribution study showed rapid blood and renal clearance with low retention in all major organs at 1, 2, and 4 h after injection. In an ApoE-/- mouse model, [64Cu]Cu-ICT-01 demonstrated sensitive and specific detection of CD163+ macrophages and capability for tracking the progression of atherosclerotic lesions; these findings were further confirmed in Ldlr-/- and PCSK9 mouse models. Immunostaining showed elevated expression of CD163+ macrophages across the plaques. Flow cytometry and single-cell RNA sequencing confirmed the specific expression of CD163 on tissue-resident macrophages. Human tissue characterization demonstrated high expression of CD163+ macrophages on atherosclerotic lesions, and ex vivo autoradiography revealed specific binding of [64Cu]Cu-ICT-01 to human CD163. Conclusion: This work reported the development of a PET radiotracer binding CD163+ macrophages. The elevated expression of CD163+ resident macrophages on human plaques indicated the potential of CD163 as a biomarker for vulnerable plaques. The sensitivity and specificity of [64Cu]Cu-ICT-01 in imaging CD163+ macrophages warrant further investigation in translational settings.
Collapse
Affiliation(s)
- Xiuli Zhang
- Mallinckrodt Institute of Radiology, University of Missouri, Columbia, Missouri
| | - Gyu Seong Heo
- Mallinckrodt Institute of Radiology, University of Missouri, Columbia, Missouri
| | - Alexandria Li
- Mallinckrodt Institute of Radiology, University of Missouri, Columbia, Missouri
| | - Divangana Lahad
- Mallinckrodt Institute of Radiology, University of Missouri, Columbia, Missouri
| | - Lisa Detering
- Mallinckrodt Institute of Radiology, University of Missouri, Columbia, Missouri
| | - Joan Tao
- Department of Medicine, University of Missouri, Columbia, Missouri
| | - Xuefeng Gao
- Mallinckrodt Institute of Radiology, University of Missouri, Columbia, Missouri
| | - Xiaohui Zhang
- Mallinckrodt Institute of Radiology, University of Missouri, Columbia, Missouri
| | - Hannah Luehmann
- Mallinckrodt Institute of Radiology, University of Missouri, Columbia, Missouri
| | - Deborah Sultan
- Mallinckrodt Institute of Radiology, University of Missouri, Columbia, Missouri
| | - Lanlan Lou
- Mallinckrodt Institute of Radiology, University of Missouri, Columbia, Missouri
| | - Rajiu Venkatesan
- Mallinckrodt Institute of Radiology, University of Missouri, Columbia, Missouri
| | - Ran Li
- Mallinckrodt Institute of Radiology, University of Missouri, Columbia, Missouri
| | - Jie Zheng
- Mallinckrodt Institute of Radiology, University of Missouri, Columbia, Missouri
| | - Junedh Amrute
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri; and
| | - Chieh-Yu Lin
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri
| | - Benjamin J Kopecky
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri; and
| | - Robert J Gropler
- Mallinckrodt Institute of Radiology, University of Missouri, Columbia, Missouri
| | - Andrea Bredemeyer
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri; and
| | - Kory Lavine
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri; and
| | - Yongjian Liu
- Mallinckrodt Institute of Radiology, University of Missouri, Columbia, Missouri;
| |
Collapse
|
16
|
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
|
17
|
Jiang Y, Sun Z, Ge Z, Tao Z, Liu M, Zhong W, Dong N, Xu L, Wang H, Xu Y, Shen X. Differential expression of Semaphorin-7A /CD163-positive macrophages in large artery and cardiogenic stroke. BMC Neurol 2024; 24:70. [PMID: 38373967 PMCID: PMC10875813 DOI: 10.1186/s12883-024-03559-6] [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: 10/22/2023] [Accepted: 02/01/2024] [Indexed: 02/21/2024] Open
Abstract
BACKGROUND Identification of the causes of stroke of undetermined etiology, specifically cardioembolism (CE) and non-CE causes, can inform treatment planning and prognosis prediction. The objective of this study was to analyze the disparities in thrombus composition, particularly Semaphorin-7A (Sema7A) and CD163, between patients diagnosed with large-artery atherosclerosis (LAA) and those with CE, and to investigate their potential association with prognosis. METHODS Thrombi were collected from patients who underwent mechanical thrombectomy at two hospitals. The patients were categorized into two groups: LAA and CE. We compared the levels of Sema7A and CD163 between these groups and analyzed their relationships with stroke severity, hemorrhagic transformation and prognosis. RESULTS The study involved a total of 67 patients. Sema7A expression was found to be significantly higher in the CE group compared to LAA (p < 0.001). Conversely, no statistically significant differences were observed for CD163 between the groups. The presence of Sema7A/CD163 did not show any associations with stroke severity or hemorrhagic transformation (all p > 0.05). However, both Sema7A (OR, 2.017; 95% CI, 1.301-3.518; p = 0.005) and CD163 (OR, 2.283; 95% CI, 1.252-5.724; p = 0.03) were associated with the poor prognosis for stroke, after adjusting for stroke severity. CONCLUSION This study highlights that CE thrombi exhibited higher levels of Sema7A expression compared to LAA thrombi. Moreover, we found a positive correlation between Sema7A/CD163 levels and the poor prognosis of patients with acute ischemic stroke.
Collapse
Affiliation(s)
- Yi Jiang
- Department of Geriatrics, Bengbu Medical College Clinical College of Lianyungang Second People's Hospital, Lianyungang, 222000, China
| | - Zhichao Sun
- Department of Pathology, Lianyungang Second People's Hospital, Lianyungang, 222000, China
| | - Zhonglin Ge
- Department of Neurology, Lianyungang Second People's Hospital, Lianyungang, 222000, China.
| | - Zhonghai Tao
- Department of Neurology, Lianyungang Second People's Hospital, Lianyungang, 222000, China
| | - Mengqian Liu
- Department of Geriatrics, Lianyungang Hospital Affiliated to Jiangsu University, Lianyungang, 222000, China
| | - Wen Zhong
- Department of Geriatrics, Lianyungang Hospital Affiliated to Jiangsu University, Lianyungang, 222000, China
| | - Nan Dong
- Department of Neurology, Shaoxing Central Hospital, Shaoxing, China
| | - Lei Xu
- Department of Pathology, Lianyungang Second People's Hospital, Lianyungang, 222000, China
| | - Hui Wang
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yiwen Xu
- Department of Infectious Disease, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Xiaozhu Shen
- Department of Geriatrics, Bengbu Medical College Clinical College of Lianyungang Second People's Hospital, Lianyungang, 222000, China.
| |
Collapse
|
18
|
Grzesiak L, Amaya-Garrido A, Feuillet G, Malet N, Swiader A, Sarthou MK, Wahart A, Ramel D, Gayral S, Schanstra JP, Klein J, Laffargue M. Leucine-Rich Alpha-2 Glycoprotein 1 Accumulates in Complicated Atherosclerosis and Promotes Calcification. Int J Mol Sci 2023; 24:16537. [PMID: 38003727 PMCID: PMC10671851 DOI: 10.3390/ijms242216537] [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: 10/09/2023] [Revised: 11/09/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Atherosclerosis is the primary cause of cardiovascular disease. The development of plaque complications, such as calcification and neo-angiogenesis, strongly impacts plaque stability and is a good predictor of mortality in patients with atherosclerosis. Despite well-known risk factors of plaque complications, such as diabetes mellitus and chronic kidney disease, the mechanisms involved are not fully understood. We and others have identified that the concentration of circulating leucine-rich α-2 glycoprotein 1 (LRG1) was increased in diabetic and chronic kidney disease patients. Using apolipoprotein E knockout mice (ApoE-/-) (fed with Western diet) that developed advanced atherosclerosis and using human carotid endarterectomy, we showed that LRG1 accumulated into an atherosclerotic plaque, preferentially in calcified areas. We then investigated the possible origin of LRG1 and its functions on vascular cells and found that LRG1 expression was specifically enhanced in endothelial cells via inflammatory mediators and not in vascular smooth muscle cells (VSMC). Moreover, we identified that LRG1 was able to induce calcification and SMAD1/5-signaling pathways in VSMC. In conclusion, our results identified for the first time that LRG1 is a direct contributor to vascular calcification and suggest a role of this molecule in the development of plaque complications in patients with atherosclerosis.
Collapse
Affiliation(s)
- Lucile Grzesiak
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1297, Institute of Cardiovascular and Metabolic Disease, 31432 Toulouse, France
- Department of Biology, Université Toulouse III Paul-Sabatier, 31062 Toulouse, France
| | - Ana Amaya-Garrido
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1297, Institute of Cardiovascular and Metabolic Disease, 31432 Toulouse, France
- Department of Biology, Université Toulouse III Paul-Sabatier, 31062 Toulouse, France
| | - Guylène Feuillet
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1297, Institute of Cardiovascular and Metabolic Disease, 31432 Toulouse, France
- Department of Biology, Université Toulouse III Paul-Sabatier, 31062 Toulouse, France
| | - Nicole Malet
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1297, Institute of Cardiovascular and Metabolic Disease, 31432 Toulouse, France
- Department of Biology, Université Toulouse III Paul-Sabatier, 31062 Toulouse, France
| | - Audrey Swiader
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1297, Institute of Cardiovascular and Metabolic Disease, 31432 Toulouse, France
- Department of Biology, Université Toulouse III Paul-Sabatier, 31062 Toulouse, France
| | - Marie-Kerguelen Sarthou
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1297, Institute of Cardiovascular and Metabolic Disease, 31432 Toulouse, France
- Department of Biology, Université Toulouse III Paul-Sabatier, 31062 Toulouse, France
| | - Amandine Wahart
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1297, Institute of Cardiovascular and Metabolic Disease, 31432 Toulouse, France
- Department of Biology, Université Toulouse III Paul-Sabatier, 31062 Toulouse, France
| | - Damien Ramel
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1297, Institute of Cardiovascular and Metabolic Disease, 31432 Toulouse, France
- Department of Biology, Université Toulouse III Paul-Sabatier, 31062 Toulouse, France
| | - Stéphanie Gayral
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1297, Institute of Cardiovascular and Metabolic Disease, 31432 Toulouse, France
- Department of Biology, Université Toulouse III Paul-Sabatier, 31062 Toulouse, France
| | - Joost Peter Schanstra
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1297, Institute of Cardiovascular and Metabolic Disease, 31432 Toulouse, France
- Department of Biology, Université Toulouse III Paul-Sabatier, 31062 Toulouse, France
| | - Julie Klein
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1297, Institute of Cardiovascular and Metabolic Disease, 31432 Toulouse, France
- Department of Biology, Université Toulouse III Paul-Sabatier, 31062 Toulouse, France
| | - Muriel Laffargue
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1297, Institute of Cardiovascular and Metabolic Disease, 31432 Toulouse, France
- Department of Biology, Université Toulouse III Paul-Sabatier, 31062 Toulouse, France
| |
Collapse
|
19
|
Drueke TB, Massy ZA. Do macrophages inhibit or promote atheroma plaque calcification and stability? Kidney Int 2023; 104:412-415. [PMID: 37331514 DOI: 10.1016/j.kint.2023.06.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 06/06/2023] [Indexed: 06/20/2023]
Affiliation(s)
- Tilman B Drueke
- Inserm Unit 1018, Team 5, CESP, Hôpital Paul Brousse, Paris-Sud University (UPS), Villejuif, France; Versailles Saint-Quentin-en-Yvelines University (Paris-Ile-de-France-Ouest University, UVSQ), Villejuif, France.
| | - Ziad A Massy
- Inserm Unit 1018, Team 5, CESP, Hôpital Paul Brousse, Paris-Sud University (UPS), Villejuif, France; Versailles Saint-Quentin-en-Yvelines University (Paris-Ile-de-France-Ouest University, UVSQ), Villejuif, France; Department of Nephrology, Ambroise Paré University Hospital, APHP, Boulogne-Billancourt/Paris, France
| |
Collapse
|
20
|
Sakamoto A, Suwa K, Kawakami R, Finn AV, Maekawa Y, Virmani R, Finn AV. Significance of Intra-plaque Hemorrhage for the Development of High-Risk Vulnerable Plaque: Current Understanding from Basic to Clinical Points of View. Int J Mol Sci 2023; 24:13298. [PMID: 37686106 PMCID: PMC10487895 DOI: 10.3390/ijms241713298] [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: 07/17/2023] [Revised: 08/18/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
Acute coronary syndromes due to atherosclerotic coronary artery disease are a leading cause of morbidity and mortality worldwide. Intra-plaque hemorrhage (IPH), caused by disruption of intra-plaque leaky microvessels, is one of the major contributors of plaque progression, causing a sudden increase in plaque volume and eventually plaque destabilization. IPH and its healing processes are highly complex biological events that involve interactions between multiple types of cells in the plaque, including erythrocyte, macrophages, vascular endothelial cells and vascular smooth muscle cells. Recent investigations have unveiled detailed molecular mechanisms by which IPH leads the development of high-risk "vulnerable" plaque. Current advances in clinical diagnostic imaging modalities, such as magnetic resonance image and intra-coronary optical coherence tomography, increasingly allow us to identify IPH in vivo. To date, retrospective and prospective clinical trials have revealed the significance of IPH as detected by various imaging modalities as a reliable prognostic indicator of high-risk plaque. In this review article, we discuss recent advances in our understanding for the significance of IPH on the development of high-risk plaque from basic to clinical points of view.
Collapse
Affiliation(s)
- Atsushi Sakamoto
- CVPath Institute, Inc., Gaithersburg, MD 20878, USA; (A.S.); (R.K.); (A.V.F.); (R.V.)
- Division of Cardiology, Internal Medicine III, Hamamatsu University School of Medicine, Hamamatsu 431-3125, Japan; (K.S.); (Y.M.)
| | - Kenichiro Suwa
- Division of Cardiology, Internal Medicine III, Hamamatsu University School of Medicine, Hamamatsu 431-3125, Japan; (K.S.); (Y.M.)
| | - Rika Kawakami
- CVPath Institute, Inc., Gaithersburg, MD 20878, USA; (A.S.); (R.K.); (A.V.F.); (R.V.)
| | - Alexandra V. Finn
- CVPath Institute, Inc., Gaithersburg, MD 20878, USA; (A.S.); (R.K.); (A.V.F.); (R.V.)
| | - Yuichiro Maekawa
- Division of Cardiology, Internal Medicine III, Hamamatsu University School of Medicine, Hamamatsu 431-3125, Japan; (K.S.); (Y.M.)
| | - Renu Virmani
- CVPath Institute, Inc., Gaithersburg, MD 20878, USA; (A.S.); (R.K.); (A.V.F.); (R.V.)
| | - Aloke V. Finn
- CVPath Institute, Inc., Gaithersburg, MD 20878, USA; (A.S.); (R.K.); (A.V.F.); (R.V.)
| |
Collapse
|
21
|
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
|