1
|
Zeng W, Zhang Y, Zhong W, Chen L, Gao Y, Li C, Zhao Y, Shen C, Zhao R, Shi B, Wang Y. Deciphering immune cell heterogeneity in vascular diseases: Insights from single-cell sequencing. Int Immunopharmacol 2025; 157:114719. [PMID: 40306113 DOI: 10.1016/j.intimp.2025.114719] [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: 02/11/2025] [Revised: 04/12/2025] [Accepted: 04/21/2025] [Indexed: 05/02/2025]
Abstract
The complexity and diversity of vascular diseases highlight the urgent need to study their pathogenesis, particularly the key role of immune cell-mediated inflammatory responses in their development. While previous reviews have outlined the involvement of immune cells in vascular pathology, a comprehensive understanding of their dynamic changes, functional states, and intercellular interactions remains incomplete. Recent advances in single-cell sequencing (SCS) have provided unprecedented insights into immune cell heterogeneity, enabling the identification of novel subpopulations and their roles in disease progression.This review extends prior work by systematically summarizing the latest applications of SCS in vascular diseases, highlighting newly discovered immune cell subsets, their interactions, and their impact on vascular pathology. By addressing current gaps in the literature-such as the functional plasticity of immune cells and their temporal dynamics-this review offers new perspectives on immune-mediated mechanisms in vascular diseases and proposes novel therapeutic strategies for their prevention and treatment.
Collapse
Affiliation(s)
- Weirong Zeng
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China
| | - Yu Zhang
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China
| | - Wanyue Zhong
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China
| | - Lei Chen
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China
| | - Yixuan Gao
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China
| | - Chaofu Li
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China
| | - Yongchao Zhao
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China
| | - Changyin Shen
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China
| | - Ranzun Zhao
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China.
| | - Bei Shi
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China.
| | - Yan Wang
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China.
| |
Collapse
|
2
|
Jia L, Wang Y, Jin C, Ma Y, Wang Y, Song L, Shen J, Xie Y, Xiang M. Macrophage heme oxygenase-1 modulates peroxynitrite-mediated vascular injury and exacerbates abdominal aortic aneurysm development. Am J Physiol Cell Physiol 2025; 328:C1808-C1821. [PMID: 40261277 DOI: 10.1152/ajpcell.00525.2023] [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: 10/11/2023] [Revised: 11/01/2023] [Accepted: 04/11/2025] [Indexed: 04/24/2025]
Abstract
Inflammatory reactions mediated by macrophages are profoundly related to the depletion of smooth muscle cells (SMCs) in abdominal aortic aneurysm (AAA) development. The findings from our previous investigation indicate that heme oxygenase-1 (HO-1) in macrophages exacerbates proinflammatory responses and oxidative damage. Therefore, the aim of this work was to gain insight into the function of HO-1 derived from macrophages and elucidate the underlying molecular mechanisms involved in AAA development. In this study, we discovered a dramatic increase in HO-1 expression in the infiltrated macrophages in experimental calcium phosphate-induced AAA tissues. Myeloid conditional HO-1-deficient mice displayed slower luminal area enlargement, as well as diminished inducible nitric oxide synthase (iNOS)-positive M1 macrophage activation, peroxynitrite generation, and SMCs apoptosis in aneurysmal tissues compared with littermate controls. Furthermore, we showed that inhibiting HO-1 eliminated the protein expression of iNOS induced by lipopolysaccharide/interferon-γ in bone marrow-derived macrophages, whereas the mRNA expression remained unaffected. Suppressing iNOS in macrophages alleviated SMCs apoptosis by decreasing nitric oxide generation in a coculture system in vitro. In summary, our study illustrates that macrophage-derived HO-1 strengthens AAA development through boosting the production of iNOS-dependent peroxynitrite and the deterioration of SMCs. These findings reveal potential therapeutic targets for resolving aneurysmal diseases.NEW & NOTEWORTHY This article illustrates the role of macrophage-derived heme oxygenase-1 (HO-1) in the development of abdominal aortic aneurysm (AAA). HO-1 deletion in macrophages hindered AAA development by reducing inducible nitric oxide synthase (iNOS)-dependent peroxynitrite production and smooth muscle cells (SMCs) apoptosis in vivo. Mechanistically, inhibition of HO-1 reduced the stimulated iNOS protein production in macrophages by lipopolysaccharide/interferon-γ. Moreover, suppressing iNOS in macrophages prevented SMCs apoptosis by decreasing nitric oxide generation in vitro.
Collapse
MESH Headings
- Animals
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/enzymology
- Aortic Aneurysm, Abdominal/chemically induced
- Aortic Aneurysm, Abdominal/genetics
- Aortic Aneurysm, Abdominal/metabolism
- Macrophages/enzymology
- Macrophages/pathology
- Peroxynitrous Acid/metabolism
- Nitric Oxide Synthase Type II/metabolism
- Nitric Oxide Synthase Type II/genetics
- Apoptosis
- Mice
- Myocytes, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/metabolism
- Heme Oxygenase-1/metabolism
- Heme Oxygenase-1/genetics
- Mice, Inbred C57BL
- Mice, Knockout
- Male
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/enzymology
- Disease Models, Animal
- Aorta, Abdominal/pathology
- Aorta, Abdominal/enzymology
- Nitric Oxide/metabolism
- Membrane Proteins
Collapse
Affiliation(s)
- Liangliang Jia
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, People's Republic of China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, People's Republic of China
| | - Yufei Wang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, People's Republic of China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, People's Republic of China
| | - Chunna Jin
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, People's Republic of China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, People's Republic of China
| | - Yuankun Ma
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, People's Republic of China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, People's Republic of China
| | - Yidong Wang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, People's Republic of China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, People's Republic of China
| | - Liuguang Song
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, People's Republic of China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, People's Republic of China
| | - Jian Shen
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, People's Republic of China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, People's Republic of China
| | - Yao Xie
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, People's Republic of China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, People's Republic of China
| | - Meixiang Xiang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, People's Republic of China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, People's Republic of China
| |
Collapse
|
3
|
Zhou Y, Chen Y, Cui Y, Gan N, Xiang Q, Li M, Zeng W, Zheng XL, Dai X, Peng J, Tang Z. Inhibition of VSMC Ferroptosis Mitigates Pathological Vascular Remodeling: A Novel Therapeutic Strategy for Abdominal Aortic Aneurysm. J Cardiovasc Transl Res 2025:10.1007/s12265-025-10621-2. [PMID: 40259193 DOI: 10.1007/s12265-025-10621-2] [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: 01/18/2025] [Accepted: 04/10/2025] [Indexed: 04/23/2025]
Abstract
Ferroptosis plays a key role in abdominal aortic aneurysm (AAA) development. This study explores whether and how ferroptosis regulates AAA progression. Ferroptosis was confirmed in human AAA tissue. In vitro experiments with primary mouse vascular smooth muscle cells (VSMCs) and abdominal aortic rings revealed that angiotensin II (Ang II) triggered ferroptosis in VSMCs. Ferrostatin-1 (Fer-1), a potent ferroptosis inhibitor, effectively suppressed this effect. Additionally, the ferroptosis inducer erastin and Ang II can both promoted pathological remodeling of abdominal aortic rings, but Fer-1 significantly suppressed these effects. In AAA mouse model, Fer-1 treatment reduced AAA formation. Mechanistically, RNA-sequencing analysis revealed that Fer-1 regulates VSMC contractile function, suppresses inflammation, and mitigates extracellular matrix remodeling. These findings highlight the critical role of VSMC ferroptosis in AAA pathogenesis and demonstrate that ferroptosis inhibition effectively reduces pathological vascular remodeling, making it a promising therapeutic strategy for preventing AAA.
Collapse
Affiliation(s)
- Yating Zhou
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Yanyu Chen
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Yuting Cui
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Ni Gan
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Qiong Xiang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Man Li
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Wen Zeng
- Shaoyang Branch of Key Laboratory for Arteriosclerology of Hunan Province, The Central Hospital of Shaoyang, Shaoyang, 421001, China
| | - Xi-Long Zheng
- Departments of Biochemistry & Molecular Biology and Physiology & Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Xiaoyan Dai
- Clinical Research Center, the Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421002, China.
- Hunan Provincial Key Laboratory of Basic and Clinical Pharmacological Research of Gastrointestinal Cancer, Hengyang, 421002, China.
| | - Juan Peng
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, China.
- Shaoyang Branch of Key Laboratory for Arteriosclerology of Hunan Province, The Central Hospital of Shaoyang, Shaoyang, 421001, China.
| | - Zhihan Tang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, China.
- Shaoyang Branch of Key Laboratory for Arteriosclerology of Hunan Province, The Central Hospital of Shaoyang, Shaoyang, 421001, China.
| |
Collapse
|
4
|
Wang HW, Huang YC, Fang YW, Jang TN, Chen M, Tsai MH. Investigating long-term risk of aortic aneurysm and dissection from fluoroquinolones and the key contributing factors using machine learning methods. Sci Rep 2025; 15:13130. [PMID: 40240493 PMCID: PMC12003681 DOI: 10.1038/s41598-025-97787-6] [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: 10/04/2024] [Accepted: 04/07/2025] [Indexed: 04/18/2025] Open
Abstract
The connection between fluoroquinolones and severe heart conditions, such as aortic aneurysm (AA) and aortic dissection (AD), has been acknowledged, but the full extent of long-term risks remains uncertain. Addressing this knowledge deficit, a retrospective cohort study was conducted in Taiwan, utilizing data from the National Health Insurance Research Database spanning from 2004 to 2010, with follow-up lasting until 2019. The study included 232,552 people who took fluoroquinolones and the same number of people who didn't, matched for age, sex, and index year. The Cox regression model was enlisted to calculate the hazard ratio (HR) for AA/AD onset. Additionally, five machine learning algorithms assisted in pinpointing critical determinants for AA/AD among those with fluoroquinolones. Intriguingly, within the longest follow-up duration of 16 years, exposed patients presented with a markedly higher incidence of AA/AD unexposed patients (80 vs. 30 per 100,000 person-years). After adjusting for multiple factors, exposure to fluoroquinolones was linked to a higher risk of AA/AD (HR 1.62, 95%CI 1.45-1.78). Machine learning identified ten factors that significantly affected AA/AD risk in those exposed. The findings illustrate a 62% elevation in the long-term risk of adverse outcomes associated with AA/AD following the administration of fluoroquinolones and concurrently delineate the salient factors contributing to AA/AD, underscoring the imperative for healthcare practitioners to meticulously evaluate the implications of prescribing these antibiotics in light of the associated risks and determinants.
Collapse
Affiliation(s)
- Hsiao-Wei Wang
- Division of Infectious Disease, Department of Internal Medicine, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Yen-Chun Huang
- Department of Artificial Intelligence, Tamkang University, No.151, Yingzhuan Rd., Tamsui Dist, New Taipei City, Taiwan
| | - Yu-Wei Fang
- Division of Nephrology, Department of Internal Medicine, Shin-Kong Wu Ho-Su Memorial Hospital, No. 95, Wen-Chang Rd, Shih-Lin Dist, Taipei, 11101, Taiwan
- Department of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Tsrang-Neng Jang
- Division of Infectious Disease, Department of Internal Medicine, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
- Department of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Mingchih Chen
- Graduate Institute of Business Administration, College of Management, Fu Jen Catholic University, New Taipei City, Taiwan
- Artificial Intelligence Development Center, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Ming-Hsien Tsai
- Division of Nephrology, Department of Internal Medicine, Shin-Kong Wu Ho-Su Memorial Hospital, No. 95, Wen-Chang Rd, Shih-Lin Dist, Taipei, 11101, Taiwan.
- Department of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan.
| |
Collapse
|
5
|
Kucher AN, Koroleva IA, Nazarenko MS. Exploring Disparities in Atherosclerosis Comorbidity with Aortic Aneurysm. Biomedicines 2025; 13:593. [PMID: 40149570 PMCID: PMC11940622 DOI: 10.3390/biomedicines13030593] [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: 11/25/2024] [Revised: 01/13/2025] [Accepted: 01/22/2025] [Indexed: 03/29/2025] Open
Abstract
Aortic aneurysm (AA) and atherosclerosis (AS) of various vascular beds are asymptomatic for a long time and are relatively common pathological conditions that lead to life-threatening and disabling complications. In this review, we discuss the current understanding of the high variation in direct and inverse comorbidity of AA and AS as presented in scientific publications. Estimates of AA and AS comorbidity depend on several factors, such as the location of AA (ascending or descending thoracic aorta or abdominal aorta), familial or sporadic cases of AA, syndromic forms of AA, and/or aortic valve pathology (bicuspid aortic valve [BAV]). To identify the causes of the comorbidity of AA and AS, it is important to consider and characterise many factors in detail. These factors include clinical characteristics of the patients included in a study (age, sex) and risk factors (mainly the presence of monogenic forms and BAV, hypertension, hypercholesterolaemia, diabetes mellitus, and cigarette smoking). Additionally, it is essential to consider characteristics of the disease course and the nature of multimorbidity and to take into account pathologies not only of the cardiovascular system but also of other organ systems, with special attention to metabolic and endocrine disorders.
Collapse
Affiliation(s)
| | | | - Maria S. Nazarenko
- Research Institute of Medical Genetics, Tomsk National Research Medical Center, Russian Academy of Sciences, 634050 Tomsk, Russia
| |
Collapse
|
6
|
Yan R, Zhang X, Wang H, Wang T, Ren G, Sun Q, Liang F, Zhu Y, Huang W, Yu HD. Autonomous, Moisture-Driven Flexible Electrogenerative Dressing for Enhanced Wound Healing. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2418074. [PMID: 39962841 DOI: 10.1002/adma.202418074] [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: 11/20/2024] [Revised: 01/19/2025] [Indexed: 03/27/2025]
Abstract
Electrotherapy has shown considerable potential in treating chronic wounds, but conventional approaches relying on bulky external power supplies and mechanical force are limited in their clinical utility. This study introduces an autonomous, moisture-driven flexible electrogenerative dressing (AMFED) that overcomes these limitations. The AMFED integrates a moist-electric generator (MEG), an antibacterial hydrogel dressing, and concentric molybdenum (Mo) electrodes to provide a self-sustaining electrical supply and potent antibacterial activity against Staphylococcus aureus and Escherichia coli. The MEG harnesses chemical energy from moisture to produce a stable direct current of 0.61 V without external input, delivering this therapeutic electrical stimulation to the wound site through the Mo electrodes. The AMFED facilitates macrophage polarization toward reparative M2 phenotype and regulates inflammatory cytokines. Moreover, in vivo studies suggest that the AMFED group significantly enhances chronic wound healing, with an approximate 41% acceleration compared to the control group. Using a diabetic mouse wound model, the AMFED demonstrates its effectiveness in promoting nerve regulation, epithelial migration, and vasculogenesis. These findings present a novel and efficient platform for accelerating chronic wound healing.
Collapse
Affiliation(s)
- Ren Yan
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
| | - Xueliang Zhang
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Hai Wang
- Department of Vascular Surgery, Renji Hospital, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
| | - Tikang Wang
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
| | - Guozhang Ren
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
| | - Qizeng Sun
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
| | - Fei Liang
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
| | - Yangzhi Zhu
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90064, USA
| | - Wei Huang
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
| | - Hai-Dong Yu
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
| |
Collapse
|
7
|
Yuan X, Shen G, Xiao H, Wang Z, Ma Y, Qin X. Netrin-1 and RGMa: Novel Regulators of Atherosclerosis-Related Diseases. Cardiovasc Drugs Ther 2025; 39:211-219. [PMID: 37439909 DOI: 10.1007/s10557-023-07478-5] [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] [Accepted: 05/26/2023] [Indexed: 07/14/2023]
Abstract
BACKGROUNDS Neuronal guidance proteins (NGPs) have been demonstrated to guide the elongation of neuronal axonal growth cones in the developing central nervous system. Non-neuronal functions of NGPs have also been described, especially in relation to atherosclerosis. FINDINGS Netrin-1 and repulsive guidance molecule a (RGMa) are NGPs that have been shown to regulate endothelial cell adhesion and angiogenesis, macrophage migration and apoptosis, smooth muscle cells (SMCs) phenotypic dedifferentiation and mobility, chemokine activities, and inflammatory responses during atherosclerosis initiation and progression. PURPOSES However, mechanistic studies have generated controversy about the specific role of Netrin-1 in atherosclerosis due to the diversity of its structure, receptors and cell sources, and the actions of RGMa in atherosclerosis have not been reported in previous reviews. Therefore, the current work reviews the evidence for roles of Netrin-1 and RGMa in the initiation and progression of atherosclerosis and discusses potential therapeutic targets in the future.
Collapse
Affiliation(s)
- Xiaofan Yuan
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 1 Yixueyuan Road, Chongqing, Yuzhong District, China
| | - Guanru Shen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 1 Yixueyuan Road, Chongqing, Yuzhong District, China
| | - Hongmei Xiao
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 1 Yixueyuan Road, Chongqing, Yuzhong District, China
| | - Zijie Wang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 1 Yixueyuan Road, Chongqing, Yuzhong District, China
| | - Yue Ma
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 1 Yixueyuan Road, Chongqing, Yuzhong District, China
| | - Xinyue Qin
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 1 Yixueyuan Road, Chongqing, Yuzhong District, China.
| |
Collapse
|
8
|
Jing J, Sun Q, Zhang R, Yang C, Yang J, Ma C, Li T. ATG4B as a novel biomarker for abdominal aortic aneurysm: integrated evaluation through experimental and bioinformatics analyses. Mol Cell Biochem 2025; 480:385-397. [PMID: 38478221 DOI: 10.1007/s11010-024-04984-4] [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: 01/17/2024] [Accepted: 03/01/2024] [Indexed: 01/03/2025]
Abstract
Autophagy related gene 4B (ATG4B) plays a central role in autophagy machinery, but its clinical relevance to AAA remains unknown. In this study, 205 AAA patients and 205 age- and sex-matched controls were included to detect the serum ATG4B levels. Meanwhile, abdominal aortic specimens from 24 AAA patients and 6 human organ donors were collected to evaluate the mRNA and in situ protein expression of ATG4B. We observed significantly higher ATG4B mRNA and protein expression levels in AAA group compared to those in control group, with a positive correlation between mRNA levels and serum/in situ protein levels (serum, r = 0.518, P = 0.010; in situ, r = 0.453, P = 0.026). Serum ATG4B exhibited the diagnostic potential for AAA (AUC = 0.702, sensitivity = 75.6%) and intraluminal thrombus recognition (AUC = 0.602, sensitivity = 67.9%). Logistic regression revealed a significant association between elevated serum ATG4B and an increased risk of AAA and intraluminal thrombus formation. Deceased patients displayed higher baseline serum ATG4B levels, which could predict postoperative mortality (HR = 1.028, 95%CI = 1.007-1.049, P = 0.009, AUC = 0.612, sensitivity = 84.6%). The bioinformatics analysis suggested that ATG4B may modulate cellular autophagy and influence pathways associated with inflammation, lipid metabolism, or apoptosis, thereby contributing to the occurrence and development of AAA. The drug-gene interaction network identified 13 potential therapeutic drugs targeting ATG4B. In conclusion, ATG4B may serve as a promising biomarker for the diagnosis and prognostic assessment of AAA patients and play a key role in the pathogenesis of AAA.
Collapse
Affiliation(s)
- Jingjing Jing
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Qiuyan Sun
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Rongqi Zhang
- The High School Affiliated to Renmin University of China, Beijing, 100080, China
| | - Chao Yang
- Trauma Center and Department of Burns, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Jun Yang
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, No.155 Nanjing Bei Street, Heping District, Shenyang, Liaoning Province, 110001, P.R. China
- Clinical Medical Research Center of Imaging in Liaoning Province, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Chunyan Ma
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, No.155 Nanjing Bei Street, Heping District, Shenyang, Liaoning Province, 110001, P.R. China
- Clinical Medical Research Center of Imaging in Liaoning Province, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Tan Li
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, No.155 Nanjing Bei Street, Heping District, Shenyang, Liaoning Province, 110001, P.R. China.
- Clinical Medical Research Center of Imaging in Liaoning Province, The First Hospital of China Medical University, Shenyang, 110001, China.
| |
Collapse
|
9
|
Palstrøm NB, Nielsen KB, Campbell AJ, Soerensen M, Rasmussen LM, Lindholt JS, Beck HC. Affinity-Enriched Plasma Proteomics for Biomarker Discovery in Abdominal Aortic Aneurysms. Proteomes 2024; 12:37. [PMID: 39728917 DOI: 10.3390/proteomes12040037] [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: 10/29/2024] [Revised: 12/02/2024] [Accepted: 12/06/2024] [Indexed: 12/28/2024] Open
Abstract
Abdominal aortic aneurysm (AAA) is a life-threatening condition characterized by the weakening and dilation of the abdominal aorta. Few diagnostic biomarkers have been proposed for this condition. We performed mass spectrometry-based proteomics analysis of affinity-enriched plasma from 45 patients with AAA and 45 matched controls to identify changes to the plasma proteome and potential diagnostic biomarkers. Gene ontology analysis revealed a significant upregulation of the proteins involved in inflammation, coagulation, and extracellular matrix in AAA patients, while proteins related to angiogenesis were among those downregulated. Using recursive feature elimination, we identified a subset of 10 significantly regulated proteins that were highly predictive of AAA. A random forest classifier trained on these proteins achieved an area under the curve (AUC) of 0.93 [95% CI: 0.91-0.95] using cross-validation. Further validation in a larger cohort is necessary to confirm these results.
Collapse
Affiliation(s)
- Nicolai Bjødstrup Palstrøm
- Center for Clinical Proteomics, Odense University Hospital, 5000 Odense, Denmark
- Department of Clinical Biochemistry, Odense University Hospital, 5000 Odense, Denmark
| | - Kristian Boje Nielsen
- Center for Clinical Proteomics, Odense University Hospital, 5000 Odense, Denmark
- Department of Clinical Biochemistry, Odense University Hospital, 5000 Odense, Denmark
| | - Amanda Jessica Campbell
- Center for Clinical Proteomics, Odense University Hospital, 5000 Odense, Denmark
- Department of Clinical Biochemistry, Odense University Hospital, 5000 Odense, Denmark
| | - Mette Soerensen
- Research Unit for Epidemiology, Biostatistics and Biodemography, Department of Public Health, University of Southern Denmark, 5230 Odense, Denmark
| | | | - Jes Sanddal Lindholt
- Department of Cardiothoracic and Vascular Surgery, Odense University Hospital, 5000 Odense, Denmark
| | - Hans Christian Beck
- Center for Clinical Proteomics, Odense University Hospital, 5000 Odense, Denmark
- Department of Clinical Biochemistry, Odense University Hospital, 5000 Odense, Denmark
| |
Collapse
|
10
|
Li S, Fu W, Wang L. Role of macrophages in aortic dissection pathogenesis: insights from preclinical studies to translational prospective. SCIENCE CHINA. LIFE SCIENCES 2024; 67:2354-2367. [PMID: 39358669 DOI: 10.1007/s11427-024-2693-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 07/25/2024] [Indexed: 10/04/2024]
Abstract
Aortic dissection is a critical vascular disease that is characterized by a high mortality rate and inflammation significantly influences its onset and progression. Recent studies highlight the integral role of macrophages, key players in the immune system, in the pathological landscape of aortic dissection. These cells are involved in crucial processes, such as the remodeling of the extracellular matrix, immunocyte infiltration, and phenotypic switching of smooth muscle cells, which are essential for the structural integrity and functional dynamics of the aortic wall. Despite these insights, the specific contributions of macrophages to the development and progression of aortic dissection remains unclear. This review explores the pathogenesis of aortic dissection with a focus on macrophages and describes their origins, phenotypic variations, and potential roles based on the most recent research findings. Furthermore, we discuss key molecules related to macrophages during aortic dissection, their interactions with other cellular components within the aorta, and the implications of these interactions for future therapeutic strategies. This comprehensive analysis aimed to improve our understanding of macrophages in aortic dissection and promote the development of targeted interventions.
Collapse
Affiliation(s)
- Shiyi Li
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Vascular Surgery Institute of Fudan University, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Weiguo Fu
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Vascular Surgery Institute of Fudan University, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
- Key Laboratory of Panvascular Disease Precision Medicine, Zhongshan Hospital Xiamen, Fudan University, Xiamen, 361015, China
| | - Lixin Wang
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Vascular Surgery Institute of Fudan University, Shanghai, 200032, China.
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China.
- Key Laboratory of Panvascular Disease Precision Medicine, Zhongshan Hospital Xiamen, Fudan University, Xiamen, 361015, China.
- Fudan Zhangjiang Institute, Shanghai, 201203, China.
| |
Collapse
|
11
|
Badary DM, Elsaied H, Abdel-Fadeil MR, Ali MK, Abou-Taleb H, Iraqy HM. Possible Role of Netrin-1/Deleted in Colorectal Cancer/Vascular Endothelial Growth Factor Signaling Pathway in the Pathogenesis of Placenta Accreta Spectrum: A Case-control Study. Int J Gynecol Pathol 2024; 43:565-572. [PMID: 38289148 DOI: 10.1097/pgp.0000000000001017] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
SUMMARY Netrin-1, an epithelial-secreted protein, plays a key role in placental formation through the promotion of cytotrophoblast proliferation and placental vascular development. These effects are mediated through several receptors, including the deleted in colorectal cancer (DCC) receptor. Placenta accreta spectrum (PAS) is an exaggerated trophoblastic invasion into the uterine myometrium. The exact etiology is unknown, but it is believed that increased trophoblastic invasion, defect decidualization, and/or abnormal angiogenesis might play a role. Our study aimed to investigate the suggested role of macrophage-induced netrin-1/DCC/vascular endothelial growth factor (VEGF) signaling in PAS pathogenesis. A total of 29 women with PAS (as cases) and 29 women with normal pregnancies (as controls) were enrolled in the study. At delivery, placental tissues of both groups were collected and processed for the evaluation of placental netrin-1 level by enzyme-linked immunoassay technique and immunohistochemical analysis of tissue DCC receptor. Placental tissue netrin-1 level of PAS cases showed a statistically significantly higher value than those in the normal group. Significant overexpression of DCC receptors, VEGF, and enhanced macrophage recruitment was noted in PAS cases in comparison to the normal placenta. Macrophage-induced netrin-1/DCC/VEGF signaling might be involved in PAS pathogenesis through the enhancement of trophoblastic angiogenesis.
Collapse
|
12
|
Zhang L, Li D, Bao S. A bibliometric and visualization analysis of global trends and frontiers on macrophages in abdominal aortic aneurysms research. Medicine (Baltimore) 2024; 103:e40274. [PMID: 39470505 PMCID: PMC11521088 DOI: 10.1097/md.0000000000040274] [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: 08/27/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Macrophages are key regulators of the inflammatory and innate immune responses. Researchers have shown that aberrant expression of macrophages contributes to the development of abdominal aortic aneurysms (AAA). However, a comprehensive bibliometric analysis exploring the research status and knowledge mapping of this area is lacking. This study aimed to explore the research status, knowledge mapping and hotspots of macrophages in AAA research from a bibliometric perspective. METHODS In this study, we retrieved articles published between 2000 and 2022 on macrophages associated with AAA research from the Web of Science Core Collection (WoSCC) database. The retrieved literature data were further analyzed using Citespace and VOSviewer software. RESULTS A total of 918 qualified publications related to AAA-associated macrophages were retrieved. The number of publications in this field has been increasing annually. China and the United States were the 2 main drivers in this field, contributing to more than 64% of the publications. In addition, the US had the most publications, top institutions, and expert researchers, dominating in research on macrophages in AAA. The Harvard University was the most productive institution, with 60 publications. The journal with the most publications was Arteriosclerosis, Thrombosis, and Vascular Biology (86). Daugherty Alan was the most prolific author (28 publications) and he was also the most cited co- author. Furthermore, the exploration of established animal models, macrophage-related inflammatory-microenvironment, macrophage-related immune mechanism, clinical translation and molecular imaging research remained future research directions in this field. CONCLUSIONS Our findings offered new insights for scholars in this field. They will help researchers explore new directions for their work.
Collapse
Affiliation(s)
- Liang Zhang
- Department of Breast Surgery, Hubei Provincial Clinical Research Center for Breast Cancer, Wuhan Clinical Research Center for Breast Cancer, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Dongyu Li
- Department of VIP In-Patient Ward, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Shiyang Bao
- Department of VIP In-Patient Ward, The First Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
13
|
Wang J, Ye W, Zou J, Yang P, Jin M, Zheng Z, Zhou C, Qiu W, Lu J, Li C, Guo S, Xu Y, Huang Z, Liu P, Liu Z. Targeting the smooth muscle cell Keap1-Nrf2-GSDMD-pyroptosis axis by cryptotanshinone prevents abdominal aortic aneurysm formation. Theranostics 2024; 14:6516-6542. [PMID: 39479449 PMCID: PMC11519792 DOI: 10.7150/thno.98400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 09/25/2024] [Indexed: 11/02/2024] Open
Abstract
Rationale: Abdominal aortic aneurysm (AAA) is an inflammatory, fatal aortic disease that currently lacks any effective drugs. Cryptotanshinone (CTS) is a prominent and inexpensive bioactive substance derived from Salvia miltiorrhiza Bunge, a well-known medicinal herb for treating cardiovascular diseases through its potent anti-inflammatory properties. Nevertheless, the therapeutic effect of CTS on AAA formation remains unknown. Methods: To investigate the therapeutic effect of CTS in AAA, variety of experimental approaches were employed, majorly including AAA mouse model establishment, real-time polymerase chain reaction (PCR), RNA sequencing, western blot, co-immunoprecipitation, scanning/transmission electron microscopy (SEM/TEM), enzyme-linked immunosorbent assay (ELISA), seahorse analysis, immunohistochemistry, and confocal imaging. Results: In this study, we demonstrated that CTS suppressed the formation of AAA in apolipoprotein E knock-out (ApoE-/-) mice infused with Ang II. A combination of network pharmacology and whole transcriptome sequencing analysis indicated that activation of the Keap1-Nrf2 pathway and regulation of programmed cell death in vascular smooth muscle cells (VSMCs) are closely linked to the anti-AAA effect of CTS. Mechanistically, CTS promoted the transcription of Nrf2 target genes, particularly Hmox-1, which prevented the activation of NLRP3 and GSDMD-initiated pyroptosis in VSMCs, thereby mitigating VSMC inflammation and maintaining the VSMC contractile phenotype. Subsequently, by utilizing molecular docking, together with the cellular thermal shift assay (CETSA) and isothermal titration calorimetry (ITC), a particular binding site was established between CTS and Keap1 at Arg415. To confirm the binding site, site-directed mutagenesis was performed, which intriguingly showed that the Arg415 mutation eliminated the binding between CTS and the Keap1-Nrf2 protein and abrogated the antioxidant and anti-pyroptosis effects of CTS. Furthermore, VSMC-specific Nrf2 knockdown in mice dramatically reversed the protective action of CTS in AAA and the inhibitory effect of CTS on VSMC pyroptosis. Conclusion: Naturally derived CTS exhibits promising efficacy as a treatment drug for AAA through its targeting of the Keap1-Nrf2-GSDMD-pyroptosis axis in VSMCs.
Collapse
MESH Headings
- Animals
- Male
- Mice
- Angiotensin II/metabolism
- Angiotensin II/pharmacology
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/drug therapy
- Aortic Aneurysm, Abdominal/pathology
- Disease Models, Animal
- Kelch-Like ECH-Associated Protein 1/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/drug effects
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- NF-E2-Related Factor 2/metabolism
- Phenanthrenes/pharmacology
- Pyroptosis/drug effects
- Signal Transduction/drug effects
Collapse
Affiliation(s)
- Jiaojiao Wang
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Key Laboratory of Computer-Aided Drug Design of Dongguan City, Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 511443, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 511443, China
| | - Weile Ye
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 511443, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 511443, China
| | - Jiami Zou
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 511443, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 511443, China
| | - Pinglian Yang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 511443, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 511443, China
| | - Mei Jin
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 511443, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 511443, China
| | - Zhihua Zheng
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 511443, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 511443, China
| | - Chunhong Zhou
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 511443, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 511443, China
| | - Wanlu Qiu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 511443, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 511443, China
| | - Jing Lu
- National and Local United Engineering Lab of Druggability and New Drugs Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangdong Province Engineering Laboratory for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Chengzhi Li
- Department of Interventional Radiology and Vascular Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Shuai Guo
- School of Basic Medical Sciences, State Key Lab of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yiming Xu
- School of Basic Medical Sciences, State Key Lab of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China
| | - Zunnan Huang
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Key Laboratory of Computer-Aided Drug Design of Dongguan City, Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China
| | - Peiqing Liu
- National and Local United Engineering Lab of Druggability and New Drugs Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangdong Province Engineering Laboratory for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Zhiping Liu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 511443, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 511443, China
| |
Collapse
|
14
|
Xu J, Liu Z, Yang Q, Ma Q, Zhou Y, Cai Y, Zhao D, Zhao G, Lu T, Ouyang K, Hong M, Kim HW, Shi H, Zhang J, Fulton D, Miller C, Malhotra R, Weintraub NL, Huo Y. Adenosine kinase inhibition protects mice from abdominal aortic aneurysm via epigenetic modulation of VSMC inflammation. Cardiovasc Res 2024; 120:1202-1217. [PMID: 38722818 PMCID: PMC11368124 DOI: 10.1093/cvr/cvae093] [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: 11/11/2023] [Revised: 12/23/2023] [Accepted: 01/26/2024] [Indexed: 09/03/2024] Open
Abstract
AIMS Abdominal aortic aneurysm (AAA) is a common, serious vascular disease with no effective pharmacological treatment. The nucleoside adenosine plays an important role in modulating vascular homeostasis, which prompted us to determine whether adenosine kinase (ADK), an adenosine metabolizing enzyme, modulates AAA formation via control of the intracellular adenosine level, and to investigate the underlying mechanisms. METHODS AND RESULTS We used a combination of genetic and pharmacological approaches in murine models of AAA induced by calcium chloride (CaCl2) application or angiotensin II (Ang II) infusion to study the role of ADK in the development of AAA. In vitro functional assays were performed by knocking down ADK with adenovirus-short hairpin RNA in human vascular smooth muscle cells (VSMCs), and the molecular mechanisms underlying ADK function were investigated using RNA-sequencing, isotope tracing, and chromatin immunoprecipitation quantitative polymerase chain reaction (ChIP-qPCR). The heterozygous deficiency of ADK protected mice from CaCl2- and Ang II-induced AAA formation. Moreover, specific knockout of ADK in VSMCs prevented Ang II-induced AAA formation, as evidenced by reduced aortic extracellular elastin fragmentation, neovascularization, and aortic inflammation. Mechanistically, ADK knockdown in VSMCs markedly suppressed the expression of inflammatory genes associated with AAA formation, and these effects were independent of adenosine receptors. The metabolic flux and ChIP-qPCR results showed that ADK knockdown in VSMCs decreased S-adenosylmethionine (SAM)-dependent transmethylation, thereby reducing H3K4me3 binding to the promoter regions of the genes that are associated with inflammation, angiogenesis, and extracellular elastin fragmentation. Furthermore, the ADK inhibitor ABT702 protected mice from CaCl2-induced aortic inflammation, extracellular elastin fragmentation, and AAA formation. CONCLUSION Our findings reveal a novel role for ADK inhibition in attenuating AAA via epigenetic modulation of key inflammatory genes linked to AAA pathogenesis.
Collapse
MESH Headings
- Animals
- Humans
- Male
- Mice
- Adenosine/metabolism
- Adenosine/analogs & derivatives
- Adenosine Kinase/antagonists & inhibitors
- Angiotensin II/metabolism
- Aorta, Abdominal/pathology
- Aorta, Abdominal/metabolism
- Aorta, Abdominal/enzymology
- Aortic Aneurysm, Abdominal/prevention & control
- Aortic Aneurysm, Abdominal/chemically induced
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/enzymology
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/genetics
- Aortitis/prevention & control
- Aortitis/enzymology
- Aortitis/pathology
- Aortitis/metabolism
- Aortitis/chemically induced
- Aortitis/genetics
- Calcium Chloride
- Cells, Cultured
- Disease Models, Animal
- DNA Methylation
- Epigenesis, Genetic
- Inflammation Mediators/metabolism
- Mice, Inbred C57BL
- Morpholines
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/drug effects
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Protein Kinase Inhibitors/pharmacology
- Pyrimidines
- Signal Transduction
Collapse
Affiliation(s)
- Jiean Xu
- Department of Physiology, Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, 232 Waihuan East Road, University Town, Guangzhou, 510006, China
- Vascular Biology Center, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, Augusta, GA 30912, USA
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Zhiping Liu
- Vascular Biology Center, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, Augusta, GA 30912, USA
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 510632, China
| | - Qiuhua Yang
- Vascular Biology Center, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, Augusta, GA 30912, USA
| | - Qian Ma
- Vascular Biology Center, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, Augusta, GA 30912, USA
| | - Yaqi Zhou
- Vascular Biology Center, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, Augusta, GA 30912, USA
| | - Yongfeng Cai
- Vascular Biology Center, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, Augusta, GA 30912, USA
| | - Dingwei Zhao
- Vascular Biology Center, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, Augusta, GA 30912, USA
| | - Guizhen Zhao
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Tammy Lu
- Vascular Biology Center, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, Augusta, GA 30912, USA
- Emory University, Atlanta, GA 30322, USA
| | - Kunfu Ouyang
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Mei Hong
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Ha Won Kim
- Vascular Biology Center, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, Augusta, GA 30912, USA
| | - Huidong Shi
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Jifeng Zhang
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - David Fulton
- Vascular Biology Center, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, Augusta, GA 30912, USA
| | - Clint Miller
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22903, USA
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA 22903, USA
| | - Rajeev Malhotra
- Division of Cardiology, Department of Medicine, Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Neal L Weintraub
- Vascular Biology Center, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, Augusta, GA 30912, USA
| | - Yuqing Huo
- Vascular Biology Center, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, Augusta, GA 30912, USA
| |
Collapse
|
15
|
Shu F, Huang H, Xiao S, Xia Z, Zheng Y. Netrin-1 co-cross-linked hydrogel accelerates diabetic wound healing in situ by modulating macrophage heterogeneity and promoting angiogenesis. Bioact Mater 2024; 39:302-316. [PMID: 38827174 PMCID: PMC11143790 DOI: 10.1016/j.bioactmat.2024.04.019] [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: 01/20/2024] [Revised: 04/16/2024] [Accepted: 04/18/2024] [Indexed: 06/04/2024] Open
Abstract
Diabetic wounds, characterized by prolonged inflammation and impaired vascularization, are a serious complication of diabetes. This study aimed to design a gelatin methacrylate (GelMA) hydrogel for the sustained release of netrin-1 and evaluate its potential as a scaffold to promote diabetic wound healing. The results showed that netrin-1 was highly expressed during the inflammation and proliferation phases of normal wounds, whereas it synchronously exhibited aberrantly low expression in diabetic wounds. Neutralization of netrin-1 inhibited normal wound healing, and the topical application of netrin-1 accelerated diabetic wound healing. Mechanistic studies demonstrated that netrin-1 regulated macrophage heterogeneity via the A2bR/STAT/PPARγ signaling pathway and promoted the function of endothelial cells, thus accelerating diabetic wound healing. These data suggest that netrin-1 is a potential therapeutic target for diabetic wounds.
Collapse
Affiliation(s)
- Futing Shu
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Hongchao Huang
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Shichu Xiao
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Zhaofan Xia
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
- Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, Shanghai, 200433, People's Republic of China
| | - Yongjun Zheng
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| |
Collapse
|
16
|
Yuan Z, Shu L, Fu J, Yang P, Wang Y, Sun J, Zheng M, Liu Z, Yang J, Song J, Song S, Cai Z. Single-Cell RNA Sequencing Deconstructs the Distribution of Immune Cells Within Abdominal Aortic Aneurysms in Mice. Arterioscler Thromb Vasc Biol 2024; 44:1986-2003. [PMID: 39051127 DOI: 10.1161/atvbaha.124.321129] [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: 04/20/2024] [Accepted: 07/15/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND Inflammation is a key component in the development of abdominal aortic aneurysm (AAA), yet insights into the roles of immune cells and their interactions in this process are limited. METHODS Using single-cell RNA transcriptomic analysis, we deconstructed the CD45+ cell population in elastase-induced murine AAA at the single-cell level. We isolated each group of immune cells from murine AAA tissue at different time points and divided them into several subtypes, listed the remarkable differentially expressed genes, explored the developmental trajectories of immune cells, and demonstrated the interactions among them. RESULTS Our findings reveal significant differences in several immune cell subsets, including macrophages, dendritic cells, and T cells, within the AAA microenvironment compared with the normal aorta. Especially, conventional dendritic cell type 1 exclusively existed in the AAA tissue rather than the normal aortas. Via CellChat analysis, we identified several intercellular communication pathways like visfatin, which targets monocyte differentiation and neutrophil extracellular trap-mediated interaction between neutrophils and dendritic cells, which might contribute to AAA development. Some of these pathways were validated in human AAA. CONCLUSIONS Despite the absence of external pathogenic stimuli, AAA tissues develop a complex inflammatory microenvironment involving numerous immune cells. In-depth studies of the inflammatory network shall provide new strategies for patients with AAA.
Collapse
MESH Headings
- Aortic Aneurysm, Abdominal/immunology
- Aortic Aneurysm, Abdominal/genetics
- Aortic Aneurysm, Abdominal/chemically induced
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/metabolism
- Animals
- Single-Cell Analysis
- Disease Models, Animal
- Mice, Inbred C57BL
- Aorta, Abdominal/pathology
- Aorta, Abdominal/metabolism
- Aorta, Abdominal/immunology
- Mice
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Humans
- Macrophages/metabolism
- Macrophages/immunology
- Male
- Transcriptome
- RNA-Seq
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Gene Expression Profiling/methods
- Pancreatic Elastase
- Cell Communication
Collapse
Affiliation(s)
- Zhen Yuan
- Departments of Cardiology (Z.Y., L.S., Y.W., Z.C.), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, China (Z.Y., L.S., Y.W., Z.C.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Z.Y., L.S., Y.W., Z.C.)
| | - Li Shu
- Departments of Cardiology (Z.Y., L.S., Y.W., Z.C.), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, China (Z.Y., L.S., Y.W., Z.C.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Z.Y., L.S., Y.W., Z.C.)
| | - Jiantao Fu
- Institute of Hepatology and Metabolic Diseases, Hangzhou Normal University, China (J.F., P.Y., J.Y.)
| | - Peipei Yang
- Institute of Hepatology and Metabolic Diseases, Hangzhou Normal University, China (J.F., P.Y., J.Y.)
| | - Yidong Wang
- Departments of Cardiology (Z.Y., L.S., Y.W., Z.C.), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, China (Z.Y., L.S., Y.W., Z.C.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Z.Y., L.S., Y.W., Z.C.)
| | - Jie Sun
- Pathology (J. Sun, M.Z.), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Mengsha Zheng
- Pathology (J. Sun, M.Z.), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhenjie Liu
- Vascular Surgery (Z.L.), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jin Yang
- Institute of Hepatology and Metabolic Diseases, Hangzhou Normal University, China (J.F., P.Y., J.Y.)
| | - Jiangping Song
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, China (J. Song, S.S.)
| | - Shen Song
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, China (J. Song, S.S.)
| | - Zhejun Cai
- Departments of Cardiology (Z.Y., L.S., Y.W., Z.C.), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, China (Z.Y., L.S., Y.W., Z.C.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Z.Y., L.S., Y.W., Z.C.)
| |
Collapse
|
17
|
Hussen NHA, Qadir SH, Rahman HS, Hamalaw YY, Kareem PSS, Hamza BA. Long-term toxicity of fluoroquinolones: a comprehensive review. Drug Chem Toxicol 2024; 47:795-806. [PMID: 37501614 DOI: 10.1080/01480545.2023.2240036] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 07/11/2023] [Accepted: 07/14/2023] [Indexed: 07/29/2023]
Abstract
Fluoroquinolones (FQs) are highly potent bactericidal antibiotics with broad-spectrum activity against Gram-negative/positive bacteria. The Food and Drug Administration (FDA) anticipated the presence of a long-lasting incapacity of Fluoroquinolone Associated Toxicity (FQAT), which is not officially documented yet. This review aimed to précis the existing information on FQA long-term toxicity, such as cardiotoxicity, aortic aneurysm, tendon rupture, nephrotoxicity, hepatotoxicity, peripheral neuropathy, vagus nervous dysfunction, reactive oxygen species (ROS), phototoxicity, glucose hemostasis, and central nervous system (CNS) toxicity. We are focused on the CNS toxicity of FQs, either due to the direct action of the FQs on CNS receptors or by other drug co-administration, including nonsteroidal anti-inflammatory disease (NSAIDs) and theophylline. Due to the nature of the R7 side chain, FQs containing unsubstituted 7-piperazine and 7-pyrrolidine have the most significant effect. The gamma-aminobutyric acid-A (GABAA) receptor and CNS effects are inhibited through at least three possible mechanisms. Firstly, by the pharmacological action of the quinolone directly. Secondly, FQ-NSAIDs interact pharmacodynamically in which the interaction between the FQ and a receptor is significantly altered by the presence of another drug that interacts with the same receptor. An example may be the interaction between NSAIDs and some FQs. Thirdly, a pharmacokinetic drug-drug interaction leads to a higher concentration of quinolone or the other drug. An example may be the interaction between theophylline and benzodiazepines with some FQs.
Collapse
Affiliation(s)
- Narmin Hama Amin Hussen
- Department of Pharmacognosy and Pharmaceutical Chemistry, College of Pharmacy, University of Sulaimani, Sulaimaniyah, Iraq
| | - Shnyar Hamid Qadir
- Department of Biochemistry and Clinical Chemistry, College of Pharmacy, University of Sulaimani, Sulaimaniyah, Iraq
| | - Heshu Sulaiman Rahman
- Department of Physiology, College of Medicine, University of Sulaimani, Sulaimaniyah, Iraq
- Department of Medical Laboratory Sciences, Komar University of Science and Technology, Sulaimaniyah, Iraq
| | - Yusra Yassin Hamalaw
- Department of Pharmacognosy and Pharmaceutical Chemistry, College of Pharmacy, University of Sulaimani, Sulaimaniyah, Iraq
| | - Parsan Siyamand Shekh Kareem
- Department of Pharmacognosy and Pharmaceutical Chemistry, College of Pharmacy, University of Sulaimani, Sulaimaniyah, Iraq
| | - Botan Aziz Hamza
- Department of Pharmacognosy and Pharmaceutical Chemistry, College of Pharmacy, University of Sulaimani, Sulaimaniyah, Iraq
| |
Collapse
|
18
|
Quelquejay H, Al-Rifai R, Silvestro M, Vandestienne M, Ferreira I, Mirault T, Henrion D, Zhong X, Santos-Zas I, Goudot G, Alayrac P, Robidel E, Autret G, Balvay D, Taleb S, Tedgui A, Boulanger CM, Zernecke A, Saliba AE, Hadchouel J, Ramkhelawon B, Cochain C, Bergaya S, Jeunemaitre X, Ait-Oufella H. L-Wnk1 Deletion in Smooth Muscle Cells Causes Aortitis and Inflammatory Shift. Circ Res 2024; 135:488-502. [PMID: 38979610 DOI: 10.1161/circresaha.124.324366] [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: 02/02/2024] [Accepted: 06/27/2024] [Indexed: 07/10/2024]
Abstract
BACKGROUND The long isoform of the Wnk1 (with-no-lysine [K] kinase 1) is a ubiquitous serine/threonine kinase, but its role in vascular smooth muscle cells (VSMCs) pathophysiology remains unknown. METHODS AngII (angiotensin II) was infused in Apoe-/- to induce experimental aortic aneurysm. Mice carrying an Sm22-Cre allele were cross-bred with mice carrying a floxed Wnk1 allele to specifically investigate the functional role of Wnk1 in VSMCs. RESULTS Single-cell RNA-sequencing of the aneurysmal abdominal aorta from AngII-infused Apoe-/- mice revealed that VSMCs that did not express Wnk1 showed lower expression of contractile phenotype markers and increased inflammatory activity. Interestingly, WNK1 gene expression in VSMCs was decreased in human abdominal aortic aneurysm. Wnk1-deficient VSMCs lost their contractile function and exhibited a proinflammatory phenotype, characterized by the production of matrix metalloproteases, as well as cytokines and chemokines, which contributed to local accumulation of inflammatory macrophages, Ly6Chi monocytes, and γδ T cells. Sm22Cre+Wnk1lox/lox mice spontaneously developed aortitis in the infrarenal abdominal aorta, which extended to the thoracic area over time without any negative effect on long-term survival. AngII infusion in Sm22Cre+Wnk1lox/lox mice aggravated the aortic disease, with the formation of lethal abdominal aortic aneurysms. Pharmacological blockade of γδ T-cell recruitment using neutralizing anti-CXCL9 (anti-CXC motif chemokine ligand 9) antibody treatment, or of monocyte/macrophage using Ki20227, a selective inhibitor of CSF1 receptor, attenuated aortitis. Wnk1 deletion in VSMCs led to aortic wall remodeling with destruction of elastin layers, increased collagen content, and enhanced local TGF-β (transforming growth factor-beta) 1 expression. Finally, in vivo TGF-β blockade using neutralizing anti-TGF-β antibody promoted saccular aneurysm formation and aorta rupture in Sm22 Cre+ Wnk1lox/lox mice but not in control animals. CONCLUSION Wnk1 is a key regulator of VSMC function. Wnk1 deletion promotes VSMC phenotype switch toward a pathogenic proinflammatory phenotype, orchestrating deleterious vascular remodeling and spontaneous severe aortitis in mice.
Collapse
MESH Headings
- Animals
- Aortitis/genetics
- Aortitis/metabolism
- Aortitis/pathology
- Mice
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Angiotensin II
- Aortic Aneurysm, Abdominal/genetics
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/pathology
- Humans
- WNK Lysine-Deficient Protein Kinase 1/genetics
- WNK Lysine-Deficient Protein Kinase 1/metabolism
- Mice, Inbred C57BL
- Male
- Cells, Cultured
- Mice, Knockout, ApoE
- Disease Models, Animal
- Inflammation/metabolism
- Inflammation/genetics
- Inflammation/pathology
- Aorta, Abdominal/metabolism
- Aorta, Abdominal/pathology
Collapse
Affiliation(s)
- Helene Quelquejay
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
| | - Rida Al-Rifai
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
| | - Michele Silvestro
- Division of Vascular and Endovascular Surgery, Department of Surgery and Department of Cell Biology, New York University Langone Medical Center (M.S., B.R.)
| | - Marie Vandestienne
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
| | - Irmine Ferreira
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
| | - Tristan Mirault
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
| | - Daniel Henrion
- MITOVASC Department, Team 2 (CarMe), ICAT SFR (Interactions Cellulaires et Applications Thérapeutiques Structure Fédérale de Recherche), University of Angers, Inserm U1083, France (D.H.)
| | - Xiaodan Zhong
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
| | - Icia Santos-Zas
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
- Laboratorio de Endocrinología Celular, Área de Endocrinología Molecular y Celular Instituto de Investigación Sanitaria de Santiago, Complejo Hospitalario Universitario de Santiago, Santiago de Compostela, Spain (I.S.-Z.)
| | - Guillaume Goudot
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
| | - Paul Alayrac
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
| | - Estelle Robidel
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
| | - Gwennhael Autret
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
| | - Daniel Balvay
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
| | - Soraya Taleb
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
| | - Alain Tedgui
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
| | - Chantal M Boulanger
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
| | - Alma Zernecke
- Institute of Experimental Biomedicine, University Hospital Würzburg, Germany (A.Z., C.C.)
| | - Antoine-Emmanuel Saliba
- Helmholtz Institute for RNA-Based Infection Research, Helmholtz-Center for Infection Research, Würzburg, Germany (A.-E.S.)
| | - Juliette Hadchouel
- Inserm UMRS 1155, Tenon Hospital (J.H.), Sorbonne Université, Paris, France
| | - Bhama Ramkhelawon
- Division of Vascular and Endovascular Surgery, Department of Surgery and Department of Cell Biology, New York University Langone Medical Center (M.S., B.R.)
| | - Clement Cochain
- Institute of Experimental Biomedicine, University Hospital Würzburg, Germany (A.Z., C.C.)
| | - Sonia Bergaya
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
| | - Xavier Jeunemaitre
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
| | - Hafid Ait-Oufella
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
- Medical Intensive Care Unit, Hôpital Saint-Antoine, AP-HP (Assistance Publique- Hôpitaux de Paris) (H.A.-O.), Sorbonne Université, Paris, France
| |
Collapse
|
19
|
Zhou S, Zhang X, Ni W, He Y, Li M, Wang C, Bai Y, Zhang H, Yao M. An Immune-Regulating Polysaccharide Hybrid Hydrogel with Mild Photothermal Effect and Anti-Inflammatory for Accelerating Infected Wound Healing. Adv Healthc Mater 2024; 13:e2400003. [PMID: 38711313 DOI: 10.1002/adhm.202400003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 04/24/2024] [Indexed: 05/08/2024]
Abstract
Bacterial infections and excessive inflammation present substantial challenges for clinical wound healing. Hydrogels with mild photothermal (PTT) effects have emerged as promising agents owing to their dual actions: positive effects on cells and negative effects on bacteria. Here, an injectable self-healing hydrogel of oxidized konjac glucomannan/arginine-modified chitosan (OKGM/CS-Arg, OC) integrated with protocatechualdehyde-@Fe (PF) nanoparticles capable of effectively absorbing near-infrared radiation is synthesized successfully. The OC/PF hydrogels exhibit excellent mechanical properties, biocompatibility, and antioxidant activity. Moreover, in synergy with PTT, OC/PF demonstrates potent antibacterial effects while concurrently stimulating cell migration and new blood vessel formation. In methicillin-resistant Staphylococcus aureus-infected full-thickness mouse wounds, the OC/PF hydrogel displays remarkable antibacterial and anti-inflammatory activities, and accelerates wound healing by regulating the wound immune microenvironment and promoting M2 macrophage polarization. Consequently, the OC/PF hydrogel represents a novel therapeutic approach for treating multidrug-resistant bacterial infections and offers a technologically advanced solution for managing infectious wounds in clinical settings.
Collapse
Affiliation(s)
- ShengZhe Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
| | - Xueliang Zhang
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Wei Ni
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan, 430000, P. R. China
| | - Yu He
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
| | - Ming Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
| | - Caixia Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
| | - Yubing Bai
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
| | - Hao Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
| | - Min Yao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
- Institute of Traumatic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 201999, P. R. China
| |
Collapse
|
20
|
Zou J, Zheng Z, Ye W, Jin M, Yang P, Little PJ, Wang J, Liu Z. Targeting the smooth muscle cell KEAP1-Nrf2-STING axis with pterostilbene attenuates abdominal aortic aneurysm. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155696. [PMID: 38763007 DOI: 10.1016/j.phymed.2024.155696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 04/27/2024] [Accepted: 04/29/2024] [Indexed: 05/21/2024]
Abstract
BACKGROUND Abdominal aortic aneurysm (AAA) is a life-threatening aortic disease, and to date, there are currently no effective pharmacological treatments to address this condition. Activation of cytosolic DNA sensing adaptor stimulator of interferon genes (STING) signaling is a crucial mechanism in AAA formation. PURPOSE This study investigated pterostilbene (Pt), a naturally occurring polyphenol and resveratrol analogue, as a STING inhibitor for preventing AAA. METHODS We evaluated the effect of Pt on AAA formation in angiotensin II (AngII)-infused apolipoprotein E-deficient (ApoE-/-) mice. We used histological analysis, MMP activity measurement, western blot, and immunohistochemistry to detect AAA formation and development. We applied RNA sequencing, molecular docking, cellular thermal shift assay (CETSA) and functional studies to dissect the molecular mechanism of Pt-regulating KEAP1-Nrf2-STING signaling. We conditionally knocked down Nrf2 in vascular smooth muscle cells (VSMCs) in vivo to investigate its role in Pt-mediated protective effects on AAA. RESULTS Pt effectively blocked the formation of AAA in AngII-infused ApoE-/- mice. Whole transcriptome sequencing analysis revealed that nuclear factor erythroid 2-related factor 2 (Nrf2) and STING pathway in VSMCs were linked to the anti-AAA effects of pterostilbene. Mechanistically, Pt upregulated Nrf2 target genes (e.g., HO-1 and NQO1) through activation of the KEAP1/Nrf2 signaling, which restricted the immunostimulatory axis of mtDNA-STING-TBK1-NF-κB, thereby alleviating VSMC inflammation and preserving the VSMC contractile phenotype. Subsequently, molecular docking and CETSA revealed a binding mode between Pt and KEAP1/Nrf2. Intriguingly, the inhibitory effect of Pt on STING signaling and the protective role of Pt in AAA were largely abrogated by VSMC-specific Nrf2 knockdown in mice. CONCLUSION Collectively, naturally derived Pt shows promising efficacy for the treatment of AAA by targeting the KEAP1-Nrf2-STING axis in VSMCs.
Collapse
Affiliation(s)
- Jiami Zou
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Zhihua Zheng
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Weile Ye
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Mei Jin
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Pinglian Yang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Peter J Little
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, QLD 4102, Australia; Department of Pharmacy, Guangzhou Xinhua University, Guangzhou 510520, China
| | - Jiaojiao Wang
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, Dongguan, Guangdong, 523808, China.
| | - Zhiping Liu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
21
|
Song T, Zhao S, Luo S, Chen C, Liu X, Wu X, Sun Z, Cao J, Wang Z, Wang Y, Yu B, Zhang Z, Du X, Li X, Han Z, Chen H, Chen F, Wang L, Wang H, Sun K, Han Y, Xie L, Ji Y. SLC44A2 regulates vascular smooth muscle cell phenotypic switching and aortic aneurysm. J Clin Invest 2024; 134:e173690. [PMID: 38916960 PMCID: PMC11324303 DOI: 10.1172/jci173690] [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: 07/07/2023] [Accepted: 06/13/2024] [Indexed: 06/27/2024] Open
Abstract
Aortic aneurysm is a life-threatening disease with limited interventions that is closely related to vascular smooth muscle cell (VSMC) phenotypic switching. SLC44A2, a member of the solute carrier series 44 (SLC44) family, remains undercharacterized in the context of cardiovascular diseases. Venn diagram analysis based on microarray and single-cell RNA sequencing identified SLC44A2 as a major regulator of VSMC phenotypic switching in aortic aneurysm. Screening for Slc44a2 among aortic cell lineages demonstrated its predominant location in VSMCs. Elevated levels of SLC44A2 were evident in the aorta of both patients with abdominal aortic aneurysm and angiotensin II-infused (Ang II-infused) Apoe-/- mice. In vitro, SLC44A2 silencing promoted VSMCs toward a synthetic phenotype, while SLC44A2 overexpression attenuated VSMC phenotypic switching. VSMC-specific SLC44A2-knockout mice were more susceptible to aortic aneurysm under Ang II infusion, while SLC44A2 overexpression showed protective effects. Mechanistically, SLC44A2's interaction with NRP1 and ITGB3 activates TGF-β/SMAD signaling, thereby promoting contractile gene expression. Elevated SLC44A2 in aortic aneurysm is associated with upregulated runt-related transcription factor 1 (RUNX1). Furthermore, low-dose lenalidomide (LEN; 20 mg/kg/day) suppressed aortic aneurysm progression by enhancing SLC44A2 expression. These findings reveal that the SLC44A2-NRP1-ITGB3 complex is a major regulator of VSMC phenotypic switching and provide a potential therapeutic approach (LEN) for aortic aneurysm treatment.
Collapse
MESH Headings
- Animals
- Humans
- Male
- Mice
- Angiotensin II/pharmacology
- Aortic Aneurysm/genetics
- Aortic Aneurysm/metabolism
- Aortic Aneurysm/pathology
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/genetics
- Membrane Transport Proteins/genetics
- Membrane Transport Proteins/metabolism
- Mice, Knockout
- Mice, Knockout, ApoE
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Phenotype
- Signal Transduction
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/metabolism
Collapse
Affiliation(s)
- Tianyu Song
- Gusu School, Nanjing Medical University, Suzhou, China
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shuang Zhao
- Gusu School, Nanjing Medical University, Suzhou, China
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shanshan Luo
- Gusu School, Nanjing Medical University, Suzhou, China
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chuansheng Chen
- Gusu School, Nanjing Medical University, Suzhou, China
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xingeng Liu
- Gusu School, Nanjing Medical University, Suzhou, China
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaoqi Wu
- Gusu School, Nanjing Medical University, Suzhou, China
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhongxu Sun
- Gusu School, Nanjing Medical University, Suzhou, China
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jiawei Cao
- Gusu School, Nanjing Medical University, Suzhou, China
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ziyu Wang
- Gusu School, Nanjing Medical University, Suzhou, China
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yineng Wang
- Gusu School, Nanjing Medical University, Suzhou, China
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Bo Yu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), and
| | - Zhiren Zhang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), and
- Department of Cardiology, Central Laboratory, The First Affiliated Hospital of Harbin Medical University, NHC Key Laboratory of Cell Transplantation, Harbin Medical University, China
| | - Xiaolong Du
- Department of Vascular Surgery, The Affiliated Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Xiaoqiang Li
- Department of Vascular Surgery, The Affiliated Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Zhijian Han
- Department of Urology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hongshan Chen
- Gusu School, Nanjing Medical University, Suzhou, China
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Feng Chen
- Department of Forensic Medicine, and
| | - Liansheng Wang
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Hong Wang
- Center for Metabolic Disease Research, Department of Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, USA
| | - Kangyun Sun
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Yi Han
- Critical Care Department, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Liping Xie
- Gusu School, Nanjing Medical University, Suzhou, China
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yong Ji
- Gusu School, Nanjing Medical University, Suzhou, China
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), and
| |
Collapse
|
22
|
Yu H, Jiao X, Lv Q, Li L, Du Y, Hu C, Du Z, Li F, Wang Y, Gao X, Han L, Sun X, Chen D, Qin Y. ATF4 Contributes to Abdominal Aortic Aneurysm Formation via Modulating M1 Macrophage Polarization and Inflammation. Aging Dis 2024; 16:1691-1708. [PMID: 38913045 PMCID: PMC12101539 DOI: 10.14336/ad.2024.0116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 06/10/2024] [Indexed: 06/25/2024] Open
Abstract
Abdominal aortic aneurysm (AAA) is a potentially life-threatening vascular disease primarily in the male elderly population, but there is a lack of approved medical therapies to prevent the progression and rupture of AAA. Activating Transcription Factor 4 (ATF4) has been established to be involved in cardiovascular diseases, such as heart failure and calcific aortic valve disease. However, the role of ATF4 in the pathogenesis of AAA remains unclear. We found that ATF4 expression was significantly increased in patients with AAA and mouse models of AAA and was mainly confined to macrophages in arteries. ATF4 knockdown significantly attenuated aneurysm formation in experimental mouse model of AAA, while ATF4 overexpression promoted the development of AAA. RNA sequencing suggested that ATF4 was strongly related to the biological function of acute inflammatory response. Macrophages-specific ATF4 knockout significantly reduced the incidence and development of AAA, and decreased M1 polarization of macrophages in mice. Sphingomyelin phosphodiesterase 3 (SMPD3), a regulator of inflammatory responses in monocytes/macrophages, has been identified as a target gene of ATF4 through RNA sequencing, ChIP sequencing, and standard ChIP analyses. ATF4 induces M1 polarization of macrophages through the activation of SMPD3, thereby promoting inflammatory responses. Together, these results suggest that ATF4 mediated macrophage M1 polarization by regulating the expression of target genes SMPD3, leading to an increased inflammatory response, which further promotes the formation and development of AAA. These findings suggest ATF4 may be a new therapeutic target for AAA.
Collapse
Affiliation(s)
- Huahui Yu
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Anzhen Hospital, Capital Medical University, Ministry of Education, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, China.
| | - Xiaolu Jiao
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Anzhen Hospital, Capital Medical University, Ministry of Education, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, China.
| | - Qianwen Lv
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Anzhen Hospital, Capital Medical University, Ministry of Education, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, China.
| | - Linyi Li
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Anzhen Hospital, Capital Medical University, Ministry of Education, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, China.
| | - Yunhui Du
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Anzhen Hospital, Capital Medical University, Ministry of Education, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, China.
| | - Chaowei Hu
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Anzhen Hospital, Capital Medical University, Ministry of Education, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, China.
| | - Zhiyong Du
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Anzhen Hospital, Capital Medical University, Ministry of Education, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, China.
| | - Fan Li
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Anzhen Hospital, Capital Medical University, Ministry of Education, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, China.
| | - Yu Wang
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Anzhen Hospital, Capital Medical University, Ministry of Education, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, China.
| | - Xiaoqian Gao
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Anzhen Hospital, Capital Medical University, Ministry of Education, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, China.
| | - Lijie Han
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Anzhen Hospital, Capital Medical University, Ministry of Education, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, China.
| | - Xuechun Sun
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Anzhen Hospital, Capital Medical University, Ministry of Education, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, China.
| | - Dong Chen
- Department of Pathology, Beijing AnZhen Hospital, Capital Medical University, Beijing, China
| | - Yanwen Qin
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Anzhen Hospital, Capital Medical University, Ministry of Education, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, China.
| |
Collapse
|
23
|
Chen R, Zhang H, Tang B, Luo Y, Yang Y, Zhong X, Chen S, Xu X, Huang S, Liu C. Macrophages in cardiovascular diseases: molecular mechanisms and therapeutic targets. Signal Transduct Target Ther 2024; 9:130. [PMID: 38816371 PMCID: PMC11139930 DOI: 10.1038/s41392-024-01840-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 04/02/2024] [Accepted: 04/21/2024] [Indexed: 06/01/2024] Open
Abstract
The immune response holds a pivotal role in cardiovascular disease development. As multifunctional cells of the innate immune system, macrophages play an essential role in initial inflammatory response that occurs following cardiovascular injury, thereby inducing subsequent damage while also facilitating recovery. Meanwhile, the diverse phenotypes and phenotypic alterations of macrophages strongly associate with distinct types and severity of cardiovascular diseases, including coronary heart disease, valvular disease, myocarditis, cardiomyopathy, heart failure, atherosclerosis and aneurysm, which underscores the importance of investigating macrophage regulatory mechanisms within the context of specific diseases. Besides, recent strides in single-cell sequencing technologies have revealed macrophage heterogeneity, cell-cell interactions, and downstream mechanisms of therapeutic targets at a higher resolution, which brings new perspectives into macrophage-mediated mechanisms and potential therapeutic targets in cardiovascular diseases. Remarkably, myocardial fibrosis, a prevalent characteristic in most cardiac diseases, remains a formidable clinical challenge, necessitating a profound investigation into the impact of macrophages on myocardial fibrosis within the context of cardiac diseases. In this review, we systematically summarize the diverse phenotypic and functional plasticity of macrophages in regulatory mechanisms of cardiovascular diseases and unprecedented insights introduced by single-cell sequencing technologies, with a focus on different causes and characteristics of diseases, especially the relationship between inflammation and fibrosis in cardiac diseases (myocardial infarction, pressure overload, myocarditis, dilated cardiomyopathy, diabetic cardiomyopathy and cardiac aging) and the relationship between inflammation and vascular injury in vascular diseases (atherosclerosis and aneurysm). Finally, we also highlight the preclinical/clinical macrophage targeting strategies and translational implications.
Collapse
Affiliation(s)
- Runkai Chen
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China
| | - Hongrui Zhang
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China
| | - Botao Tang
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China
| | - Yukun Luo
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China
| | - Yufei Yang
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China
| | - Xin Zhong
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China
| | - Sifei Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Xinjie Xu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.
| | - Shengkang Huang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.
| | - Canzhao Liu
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China.
| |
Collapse
|
24
|
Bruls S, Musumeci L, Courtois A, Hustinx R, Sakalihasan S, Namur G, Defraigne JO, Sakalihasan N. Can Biomarkers and PET Imaging Predict Abdominal Aortic Aneurysm Growth Rate? J Clin Med 2024; 13:2448. [PMID: 38673721 PMCID: PMC11051427 DOI: 10.3390/jcm13082448] [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: 02/27/2024] [Revised: 04/12/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Background: Abdominal aortic aneurysm (AAA) is a life-threatening condition due to the risk of aneurysm growth and rupture. Biomarkers linked to AAA pathogenesis are attractive candidates for AAA diagnosis and prognosis. The aim of this study was to assess circulating biomarkers levels relationship with PET imaging positivity and their predictive value in AAA growth rate. Methods: A total of 164 patients with AAA had whole body [18F]FDG PET/CT examination and blood drawn for biomarkers analysis at inclusion. Of these, 121 patients had at least one follow-up imaging assessment for AAA progression. Median (quartiles) imaging follow-up period was 32.8 months (15.2-69.6 months). Results: At baseline, PET was visually positive in 28 (17%) patients. Among PET+ patients, female proportion was higher compared to PET-patients (respectively, n = 6, 21.4% vs. n = 11, 8.1%, p = 0.046). Biomarkers of inflammation (CRP, CCL18), of proteolytic activity (MMP9), of extracellular matrix, and calcification regulation (OPN, OPG) were all significantly increased in PET+ patients (p < 0.05). During follow-up, rapid AAA growth (increase in size ≥ 1 cm per year) was observed in 36 (29.8%) patients and several biomarkers (CRP, MMP9, OPN, and OPG) were increased in those patients compared to patients without rapid growth (p < 0.05). Conclusions: Although PET positivity at baseline was not associated with rapid growth, CRP levels showed a significant association.
Collapse
Affiliation(s)
- Samuel Bruls
- Department of Cardiovascular and Thoracic Surgery, University Hospital of Liège, 4000 Liège, Belgium; (S.B.); (L.M.); (J.-O.D.)
| | - Lucia Musumeci
- Department of Cardiovascular and Thoracic Surgery, University Hospital of Liège, 4000 Liège, Belgium; (S.B.); (L.M.); (J.-O.D.)
- Surgical Research Center (GIGA—Cardiovascular Science Unit), University Hospital of Liège, 4000 Liège, Belgium;
| | - Audrey Courtois
- Surgical Research Center (GIGA—Cardiovascular Science Unit), University Hospital of Liège, 4000 Liège, Belgium;
| | - Roland Hustinx
- Department of Nuclear Medicine, University Hospital of Liège, 4000 Liège, Belgium;
| | | | - Gauthier Namur
- Department of Nuclear Medicine, CHC Mont-Légia, 4000 Liège, Belgium;
| | - Jean-Olivier Defraigne
- Department of Cardiovascular and Thoracic Surgery, University Hospital of Liège, 4000 Liège, Belgium; (S.B.); (L.M.); (J.-O.D.)
| | - Natzi Sakalihasan
- Department of Cardiovascular and Thoracic Surgery, University Hospital of Liège, 4000 Liège, Belgium; (S.B.); (L.M.); (J.-O.D.)
- Surgical Research Center (GIGA—Cardiovascular Science Unit), University Hospital of Liège, 4000 Liège, Belgium;
| |
Collapse
|
25
|
Barberán J, de la Cuerda A, Tejeda González MI, López Aparicio A, Monfort Vinuesa C, Ramos Sánchez A, Barberán LC. Safety of fluoroquinolones. REVISTA ESPANOLA DE QUIMIOTERAPIA : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE QUIMIOTERAPIA 2024; 37:127-133. [PMID: 38140798 PMCID: PMC10945095 DOI: 10.37201/req/143.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 12/19/2023] [Indexed: 06/01/2023]
Abstract
Fluoroquinolones (FQs) are one of the most commonly prescribed classes of antibiotics. Although they were initially well tolerated in randomized clinical trials, subsequent epidemiological studies have reported an increased risk of threatening, severe, long-lasting, disabling and irreversible adverse effects (AEs), related to neurotoxicity and collagen degradation, such as tendonitis, Achilles tendon rupture, aortic aneurysm, and retinal detachment. This article reviews the main potentially threatening AEs, the alarms issued by regulatory agencies and therapeutic alternatives.
Collapse
Affiliation(s)
- J Barberán
- José Barberán, Hospital Universitario HM Montepríncipe, Facultad HM Hospitales de Ciencias de la Salud, Universidad Camilo José Cela, Madrid, Spain.
| | | | | | | | | | | | | |
Collapse
|
26
|
Ross JB, Myers LM, Noh JJ, Collins MM, Carmody AB, Messer RJ, Dhuey E, Hasenkrug KJ, Weissman IL. Depleting myeloid-biased haematopoietic stem cells rejuvenates aged immunity. Nature 2024; 628:162-170. [PMID: 38538791 PMCID: PMC11870232 DOI: 10.1038/s41586-024-07238-x] [Citation(s) in RCA: 71] [Impact Index Per Article: 71.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 02/26/2024] [Indexed: 04/01/2024]
Abstract
Ageing of the immune system is characterized by decreased lymphopoiesis and adaptive immunity, and increased inflammation and myeloid pathologies1,2. Age-related changes in populations of self-renewing haematopoietic stem cells (HSCs) are thought to underlie these phenomena3. During youth, HSCs with balanced output of lymphoid and myeloid cells (bal-HSCs) predominate over HSCs with myeloid-biased output (my-HSCs), thereby promoting the lymphopoiesis required for initiating adaptive immune responses, while limiting the production of myeloid cells, which can be pro-inflammatory4. Ageing is associated with increased proportions of my-HSCs, resulting in decreased lymphopoiesis and increased myelopoiesis3,5,6. Transfer of bal-HSCs results in abundant lymphoid and myeloid cells, a stable phenotype that is retained after secondary transfer; my-HSCs also retain their patterns of production after secondary transfer5. The origin and potential interconversion of these two subsets is still unclear. If they are separate subsets postnatally, it might be possible to reverse the ageing phenotype by eliminating my-HSCs in aged mice. Here we demonstrate that antibody-mediated depletion of my-HSCs in aged mice restores characteristic features of a more youthful immune system, including increasing common lymphocyte progenitors, naive T cells and B cells, while decreasing age-related markers of immune decline. Depletion of my-HSCs in aged mice improves primary and secondary adaptive immune responses to viral infection. These findings may have relevance to the understanding and intervention of diseases exacerbated or caused by dominance of the haematopoietic system by my-HSCs.
Collapse
Affiliation(s)
- Jason B Ross
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Ludwig Center for Cancer Stem Cell Research and Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Lara M Myers
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Joseph J Noh
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Ludwig Center for Cancer Stem Cell Research and Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Madison M Collins
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
- Department of Biological and Physical Sciences, Montana State University Billings, Billings, MT, USA
| | - Aaron B Carmody
- Research Technologies Branch, Rocky Mountain Laboratories, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Ronald J Messer
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Erica Dhuey
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Ludwig Center for Cancer Stem Cell Research and Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Kim J Hasenkrug
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA.
| | - Irving L Weissman
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA.
- Ludwig Center for Cancer Stem Cell Research and Medicine, Stanford University School of Medicine, Stanford, CA, USA.
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
27
|
Yang H, Zhou T, Liu B. Macrophage-mediated downregulation of lncRNA Carmn in mouse abdominal aortic aneurysm. Vascul Pharmacol 2024; 154:107264. [PMID: 38097098 PMCID: PMC10939852 DOI: 10.1016/j.vph.2023.107264] [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] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/03/2023] [Accepted: 12/07/2023] [Indexed: 12/18/2023]
Abstract
The long noncoding RNA (lncRNA) CARMN (cardiac mesoderm enhancer associated noncoding RNA) is a highly conserved lncRNA that expresses primarily by smooth muscle cells (SMCs). Recent literature demonstrates that CARMN plays a critical role in the differentiation and maintaining of the contractile state of vascular SMCs. Because aortic SMCs show diminished contractile proteins in abdominal aortic aneurysms (AAAs), we hypothesize that the expression of CARMN is downregulated in the aortic wall affected by aneurysm. In this study, we analyzed publicly available single-cell or bulk RNA sequencing data comparing healthy and aneurysmal mouse aortic tissues. In both healthy and diseased aortas, Carmn expression was enriched in SMCs characterized by the high expression of SMC-specific contractile proteins including Myh11 and Acta2. Carmn expression levels varied among the sub-clusters of SMCs and consequently along the aortic tree. Comparing to the corresponding sham aorta, aortas from 3 distinct AAA models contained less Carmn. To validate the Carmn downregulation, we induced AAA using the Angiotensin II and CaCl2 models. In situ hybridization showed that Carmn mRNA located in the nuclei of SMCs and became downregulated within a few days following the aneurysm induction. Mechanistically, we tested whether Carmn expression is regulated by infiltrating macrophages --- the predominant inflammatory cells found in aneurysmal tissues --- by treating healthy mouse aortic SMCs with media conditioned by macrophages primed with pro-inflammatory or anti-inflammatory cytokines. PCR analysis showed that inflammatory macrophages reduced the expression of Carmn and contractile genes including Myh11 and Acta2. Taken together, our results from bioinformatic and experimental analyses demonstrate that Carmn is downregulated in different AAA models, likely by inflammatory macrophages. The negative regulation of Carmn in AAA tissues may explain at least in part the loss of SMC contractile state during the pathogenesis of this progressive degenerative disease.
Collapse
Affiliation(s)
- Huan Yang
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Ting Zhou
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Bo Liu
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States.
| |
Collapse
|
28
|
Offline to online: my musical odyssey to publication. Nat Commun 2024; 15:214. [PMID: 38172104 PMCID: PMC10764772 DOI: 10.1038/s41467-023-44474-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024] Open
|
29
|
Cho MJ, Lee MR, Park JG. Aortic aneurysms: current pathogenesis and therapeutic targets. Exp Mol Med 2023; 55:2519-2530. [PMID: 38036736 PMCID: PMC10766996 DOI: 10.1038/s12276-023-01130-w] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/29/2023] [Accepted: 08/30/2023] [Indexed: 12/02/2023] Open
Abstract
Aortic aneurysm is a chronic disease characterized by localized expansion of the aorta, including the ascending aorta, arch, descending aorta, and abdominal aorta. Although aortic aneurysms are generally asymptomatic, they can threaten human health by sudden death due to aortic rupture. Aortic aneurysms are estimated to lead to 150,000 ~ 200,000 deaths per year worldwide. Currently, there are no effective drugs to prevent the growth or rupture of aortic aneurysms; surgical repair or endovascular repair is the only option for treating this condition. The pathogenic mechanisms and therapeutic targets for aortic aneurysms have been examined over the past decade; however, there are unknown pathogenic mechanisms involved in cellular heterogeneity and plasticity, the complexity of the transforming growth factor-β signaling pathway, inflammation, cell death, intramural neovascularization, and intercellular communication. This review summarizes the latest research findings and current pathogenic mechanisms of aortic aneurysms, which may enhance our understanding of aortic aneurysms.
Collapse
Affiliation(s)
- Min Ji Cho
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Mi-Ran Lee
- Department of Biomedical Laboratory Science, Jungwon University, 85 Munmu-ro, Goesan-eup, Goesan-gun, Chungbuk, 28024, Republic of Korea
| | - Jong-Gil Park
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea.
- Department of Bioscience, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea.
| |
Collapse
|
30
|
Li Y, Li R, Li Y, Li G, Zhao Y, Mou H, Chen Y, Xiao L, Gong K. Transcription Factor TCF3 Promotes Macrophage-Mediated Inflammation and MMP Secretion in Abdominal Aortic Aneurysm by Regulating miR-143-5p /CCL20. J Cardiovasc Pharmacol 2023; 82:458-469. [PMID: 37721971 PMCID: PMC10691663 DOI: 10.1097/fjc.0000000000001484] [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: 06/06/2023] [Accepted: 08/28/2023] [Indexed: 09/20/2023]
Abstract
ABSTRACT Damage to the abdominal aortic wall and the local inflammatory response are key factors resulting in abdominal aortic aneurysm (AAA) formation. During this process, macrophage polarization plays a key role. However, in AAA, the regulatory mechanism of macrophages is still unclear, and further research is needed. In this study, we found that the transcription factor TCF3 was expressed at low levels in AAA. We overexpressed TCF3 and found that TCF3 could inhibit MMP and inflammatory factor expression and promote M2 macrophage polarization, thereby inhibiting the progression of AAA. Knocking down TCF3 could promote M1 polarization and MMP and inflammatory factor expression. In addition, we found that TCF3 increased miR-143-5p expression through transcriptional activation of miR-143-5p , which further inhibited expression of the downstream chemokine CCL20 and promoted M2 macrophage polarization. Our research indicates that TCF3-mediated macrophage polarization plays a key regulatory role in AAA, complementing the role and mechanism of macrophages in the occurrence and development of AAA and providing a scientific basis for AAA treatment.
Collapse
Affiliation(s)
- Yuejin Li
- Department of General Surgery, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Rougang Li
- Department of General Surgery, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Yu Li
- Department of General Surgery, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Guosan Li
- Department of General Surgery, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Yiman Zhao
- Department of General Surgery, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Houyu Mou
- Department of General Surgery, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Yi Chen
- Department of General Surgery, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Le Xiao
- Department of General Surgery, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Kunmei Gong
- Department of General Surgery, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| |
Collapse
|
31
|
Scipione CA, Hyduk SJ, Polenz CK, Cybulsky MI. Unveiling the Hidden Landscape of Arterial Diseases at Single-Cell Resolution. Can J Cardiol 2023; 39:1781-1794. [PMID: 37716639 DOI: 10.1016/j.cjca.2023.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/25/2023] [Accepted: 09/11/2023] [Indexed: 09/18/2023] Open
Abstract
High-resolution single-cell technologies have shed light on the pathogenesis of cardiovascular diseases by enabling the discovery of novel cellular and transcriptomic signatures associated with various conditions, and uncovering new contributions of inflammatory processes, immunity, metabolic stress, and risk factors. We review the information obtained from studies using single-cell technologies in tissues with atherosclerosis and aortic aneurysms. Insights are provided on the biology of endothelial, smooth muscle, and immune cells in the arterial intima and media. In addition to cellular diversity, numerous examples of plasticity and phenotype switching are highlighted and presented in the context of normal cell functions.
Collapse
Affiliation(s)
- Corey A Scipione
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Departments of Laboratory Medicine and Pathobiology and Immunology, University of Toronto, Toronto, Ontario, Canada.
| | - Sharon J Hyduk
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Chanele K Polenz
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Departments of Laboratory Medicine and Pathobiology and Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Myron I Cybulsky
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Departments of Laboratory Medicine and Pathobiology and Immunology, University of Toronto, Toronto, Ontario, Canada; Peter Munk Cardiac Centre, University Health Network, Toronto, Ontario, Canada.
| |
Collapse
|
32
|
Atkinson G, Bianco R, Di Gregoli K, Johnson JL. The contribution of matrix metalloproteinases and their inhibitors to the development, progression, and rupture of abdominal aortic aneurysms. Front Cardiovasc Med 2023; 10:1248561. [PMID: 37799778 PMCID: PMC10549934 DOI: 10.3389/fcvm.2023.1248561] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 09/07/2023] [Indexed: 10/07/2023] Open
Abstract
Abdominal aortic aneurysms (AAAs) account for up to 8% of deaths in men aged 65 years and over and 2.2% of women. Patients with AAAs often have atherosclerosis, and intimal atherosclerosis is generally present in AAAs. Accordingly, AAAs are considered a form of atherosclerosis and are frequently referred to as atherosclerotic aneurysms. Pathological observations advocate inflammatory cell infiltration alongside adverse extracellular matrix degradation as key contributing factors to the formation of human atherosclerotic AAAs. Therefore, macrophage production of proteolytic enzymes is deemed responsible for the damaging loss of ECM proteins, especially elastin and fibrillar collagens, which characterise AAA progression and rupture. Matrix metalloproteinases (MMPs) and their regulation by tissue inhibitors metalloproteinases (TIMPs) can orchestrate not only ECM remodelling, but also moderate the proliferation, migration, and apoptosis of resident aortic cells, alongside the recruitment and subsequent behaviour of inflammatory cells. Accordingly, MMPs are thought to play a central regulatory role in the development, progression, and eventual rupture of abdominal aortic aneurysms (AAAs). Together, clinical and animal studies have shed light on the complex and often diverse effects MMPs and TIMPs impart during the development of AAAs. This dichotomy is underlined from evidence utilising broad-spectrum MMP inhibition in animal models and clinical trials which have failed to provide consistent protection from AAA progression, although more encouraging results have been observed through deployment of selective inhibitors. This review provides a summary of the supporting evidence connecting the contribution of individual MMPs to AAA development, progression, and eventual rupture. Topics discussed include structural, functional, and cell-specific diversity of MMP members; evidence from animal models of AAA and comparisons with findings in humans; the dual role of MMPs and the requirement to selectively target individual MMPs; and the advances in identifying aberrant MMP activity. As evidenced, our developing understanding of the multifaceted roles individual MMPs perform during the progression and rupture of AAAs, should motivate clinical trials assessing the therapeutic potential of selective MMP inhibitors, which could restrict AAA-related morbidity and mortality worldwide.
Collapse
Affiliation(s)
| | | | | | - Jason L. Johnson
- Laboratory of Cardiovascular Pathology, Department of Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
33
|
Yodsanit N, Shirasu T, Huang Y, Yin L, Islam ZH, Gregg AC, Riccio AM, Tang R, Kent EW, Wang Y, Xie R, Zhao Y, Ye M, Zhu J, Huang Y, Hoyt N, Zhang M, Hossack JA, Salmon M, Kent KC, Guo LW, Gong S, Wang B. Targeted PERK inhibition with biomimetic nanoclusters confers preventative and interventional benefits to elastase-induced abdominal aortic aneurysms. Bioact Mater 2023; 26:52-63. [PMID: 36875050 PMCID: PMC9975632 DOI: 10.1016/j.bioactmat.2023.02.009] [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: 04/19/2022] [Revised: 02/08/2023] [Accepted: 02/08/2023] [Indexed: 02/25/2023] Open
Abstract
Abdominal aortic aneurysm (AAA) is a progressive aortic dilatation, causing ∼80% mortality upon rupture. Currently, there is no approved drug therapy for AAA. Surgical repairs are invasive and risky and thus not recommended to patients with small AAAs which, however, account for ∼90% of the newly diagnosed cases. It is therefore a compelling unmet clinical need to discover effective non-invasive strategies to prevent or slow down AAA progression. We contend that the first AAA drug therapy will only arise through discoveries of both effective drug targets and innovative delivery methods. There is substantial evidence that degenerative smooth muscle cells (SMCs) orchestrate AAA pathogenesis and progression. In this study, we made an exciting finding that PERK, the endoplasmic reticulum (ER) stress Protein Kinase R-like ER Kinase, is a potent driver of SMC degeneration and hence a potential therapeutic target. Indeed, local knockdown of PERK in elastase-challenged aorta significantly attenuated AAA lesions in vivo. In parallel, we also conceived a biomimetic nanocluster (NC) design uniquely tailored to AAA-targeting drug delivery. This NC demonstrated excellent AAA homing via a platelet-derived biomembrane coating; and when loaded with a selective PERK inhibitor (PERKi, GSK2656157), the NC therapy conferred remarkable benefits in both preventing aneurysm development and halting the progression of pre-existing aneurysmal lesions in two distinct rodent models of AAA. In summary, our current study not only establishes a new intervention target for mitigating SMC degeneration and aneurysmal pathogenesis, but also provides a powerful tool to facilitate the development of effective drug therapy of AAA.
Collapse
Affiliation(s)
- Nisakorn Yodsanit
- Department of Biomedical Engineering, Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Takuro Shirasu
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Yitao Huang
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
- The Biomedical Sciences Graduate Program (BIMS), School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Li Yin
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Zain Husain Islam
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | | | - Alessandra Marie Riccio
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Runze Tang
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Eric William Kent
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Yuyuan Wang
- Department of Biomedical Engineering, Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Ruosen Xie
- Department of Biomedical Engineering, Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Yi Zhao
- Department of Biomedical Engineering, Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Mingzhou Ye
- Department of Biomedical Engineering, Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Jingcheng Zhu
- Department of Biomedical Engineering, Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Yi Huang
- Department of Biomedical Engineering, School of Engineering, University of Virginia, Charlottesville, VA, 22908, USA
| | - Nicholas Hoyt
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
- School of Medicine and Health Sciences, George Washington University, Washington, DC, 20052, USA
| | - Mengxue Zhang
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - John A. Hossack
- Department of Biomedical Engineering, School of Engineering, University of Virginia, Charlottesville, VA, 22908, USA
| | - Morgan Salmon
- Department of Cardiac Surgery, Michigan Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - K. Craig Kent
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Lian-Wang Guo
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Shaoqin Gong
- Department of Biomedical Engineering, Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Bowen Wang
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| |
Collapse
|
34
|
Huang J, Jiang Y, Ji R, Jia Y, Wang S, Zhou Z, Wang S, Wang J, Yang Q, Bai H, Zhu X, Jiang B, Ben J, Zhang H, Li X, Chen Q. Macrophage scavenger receptor A1 antagonizes abdominal aortic aneurysm via upregulating IRG1. Biochem Pharmacol 2023; 213:115631. [PMID: 37257722 DOI: 10.1016/j.bcp.2023.115631] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 05/17/2023] [Accepted: 05/24/2023] [Indexed: 06/02/2023]
Abstract
AIMS Abdominal aortic aneurysm (AAA) is a common, usually asymptomatic disease with high mortality and limited therapeutic options. Extensive extracellular matrix (ECM) fragmentation and transmural inflammation act as major pathological processes of AAA. However, the underlying regulatory mechanisms remain incompletely understood. Herein, we aimed to investigate the role of scavenger receptor A1 (SR-A1), a key pattern recognition receptor modulating macrophage activity, in pathogenesis of AAA. METHODS AND RESULTS The AAA model was generated by administration of angiotensin II (Ang II) into apolipoprotein E knockout mice or peri-arterial application of calcium phosphate in C57BJ/6L mice. We found that SR-A1 was markedly down-regulated in the macrophages isolated from murine AAA aortas. Global or myeloid-specific ablation of SR-A1 aggravated vascular inflammation, loss of vascular smooth muscle cells and degradation of the extracellular matrix. These effects of SR-A1 deficiency on AAA development were mediated by suppressed immunoresponsive gene 1 (IRG1) and increased inflammatory response in macrophages. Mechanically, binding of SR-A1 with Lyn led to STAT3 phosphorylation and translocation into the nucleus, in which STAT3 promoted IRG1 transcription through directly binding to its promoter. Restoration of macrophage SR-A1 in SR-A1-deficient mice by bone marrow transplantation or administration of 4-octyl itaconate, the derivate of IRG1 product itaconate, could relieve murine AAA. CONCLUSION Our study reveals a protective effect of macrophage SR-A1-STAT3-IRG1 axis against aortic aneurysm formation via inhibiting inflammation.
Collapse
Affiliation(s)
- Jianan Huang
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease and Molecular Intervention, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Yunlong Jiang
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease and Molecular Intervention, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Ruiyuan Ji
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease and Molecular Intervention, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Yutian Jia
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease and Molecular Intervention, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Saiya Wang
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease and Molecular Intervention, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Zhongqiu Zhou
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease and Molecular Intervention, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Siying Wang
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease and Molecular Intervention, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Jie Wang
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease and Molecular Intervention, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Qing Yang
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease and Molecular Intervention, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Hui Bai
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease and Molecular Intervention, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China; The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
| | - Xudong Zhu
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease and Molecular Intervention, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China; The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
| | - Bin Jiang
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease and Molecular Intervention, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China; The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
| | - Jingjing Ben
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease and Molecular Intervention, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China; The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
| | - Hanwen Zhang
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease and Molecular Intervention, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China; The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
| | - Xiaoyu Li
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease and Molecular Intervention, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China.
| | - Qi Chen
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease and Molecular Intervention, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China; The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
35
|
Sun H, Liu F, Lin Z, Jiang Z, Wen X, Xu J, Zhang Z, Ma R. Silencing of NOTCH3 Signaling in Meniscus Smooth Muscle Cells Inhibits Fibrosis and Exacerbates Degeneration in a HEYL-Dependent Manner. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207020. [PMID: 37026620 PMCID: PMC10238196 DOI: 10.1002/advs.202207020] [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: 11/29/2022] [Revised: 02/08/2023] [Indexed: 06/04/2023]
Abstract
The mechanisms of meniscus fibrosis and novel ways to enhance fibrosis is unclear. This work reveals human meniscus fibrosis initiated at E24 weeks. Smooth muscle cell cluster is identified in embryonic meniscus, and the combined analysis with previous data suggests smooth muscle cell in embryonic meniscus as precursors of progenitor cells in the mature meniscus. NOTCH3 is constantly expressed in smooth muscle cells throughout embryogenesis to adulthood. Inhibition of NOTCH3 signaling in vivo inhibits meniscus fibrosis and exacerbates degeneration. Continuous histological sections show that HEYL, NOTCH3 downstream target gene, is expressed consistently with NOTCH3. HEYL knockdown in meniscus cells attenuated the COL1A1 upregulation by CTGF and TGF-β stimulation. Thus, this study discovers the existence of smooth muscle cells and fibers in the meniscus. Inhibition of NOTCH3 signaling in meniscus smooth muscle cells in a HEYL-dependent manner prevented meniscus fibrosis and exacerbated degeneration. Therefore, NOTCH3/HEYL signaling might be a potential therapeutic target for meniscus fibrosis.
Collapse
Affiliation(s)
- Hao Sun
- Department of Orthopaedic surgerySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510120China
| | - Fangzhou Liu
- Department of Orthopaedic surgerySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510120China
| | - Zhencan Lin
- Department of Orthopaedic surgerySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510120China
| | - Zongrui Jiang
- Department of Joint SurgeryFirst Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Xingzhao Wen
- Department of Joint SurgeryFirst Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Jie Xu
- Department of Orthopaedic surgerySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510120China
| | - Zhiqi Zhang
- Department of Joint SurgeryFirst Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Ruofan Ma
- Department of Orthopaedic surgerySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510120China
| |
Collapse
|
36
|
Bontekoe J, Liu B. Single-cell RNA sequencing provides novel insights to pathologic pathways in abdominal aortic aneurysm. Front Cardiovasc Med 2023; 10:1172080. [PMID: 37288252 PMCID: PMC10241995 DOI: 10.3389/fcvm.2023.1172080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 05/09/2023] [Indexed: 06/09/2023] Open
Abstract
There is gaining popularity in the use of single-cell technology and analysis in studying the pathogenesis of abdominal aortic aneurysm (AAA). As there are no current pharmacologic therapies for impeding aneurysm growth or preventing AAA rupture, identifying key pathways involved in AAA formation is critical for the development of future therapies. Single-cell RNA sequencing (scRNA-seq) technology provides an unbiased and global view of transcriptomic characteristics within each of the major cell types in aneurysmal tissues. In this brief review, we examine the current literature utilizing scRNA-seq for the analysis of AAA and discuss trends and future utility of this technology.
Collapse
Affiliation(s)
- Jack Bontekoe
- Division of Vascular Surgery, Department of Surgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, United States
| | - Bo Liu
- Division of Vascular Surgery, Department of Surgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, United States
- Department of Cellular and Regenerative Biology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, United States
| |
Collapse
|
37
|
Hong SY, Jiang HC, Xu WC, Zeng HS, Wang SG, Qin BL. Bioinformatics analysis reveals the potential role of matrix metalloproteinases in immunity and urolithiasis. Front Immunol 2023; 14:1158379. [PMID: 37006258 PMCID: PMC10050583 DOI: 10.3389/fimmu.2023.1158379] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 03/06/2023] [Indexed: 03/17/2023] Open
Abstract
BackgroundThe pathogenesis of urolithiasis remains unclear, making the development of medications for treatment and prevention stagnant. Randall’s plaques (RPs) begin as interstitial calcium phosphate crystal deposits, grow outward and breach the renal papillary surface, acting as attachment for CaOx stones. Since matrix metalloproteinases (MMPs) can degrade all components of extracellular matrix (ECM), they might participate in the breach of RPs. Besides, MMPs can modulate the immune response and inflammation, which were confirmed to be involved in urolithiasis. We aimed to investigate the role of MMPs in the development of RPs and stone formation.MethodsThe public dataset GSE73680 was mined to identify differentially expressed MMPs (DEMMPs) between normal tissues and RPs. WGCNA and three machine learning algorithms were performed to screen the hub DEMMPs. In vitro experiments were conducted for validation. Afterwards, RPs samples were classified into clusters based on the hub DEMMPs expression. Differentially expressed genes (DEGs) between clusters were identified and functional enrichment analysis and GSEA were applied to explore the biological role of DEGs. Moreover, the immune infiltration levels between clusters were evaluated by CIBERSORT and ssGSEA.ResultsFive DEMMPs, including MMP1, MMP3, MMP9, MMP10, and MMP12, were identified between normal tissues and RPs, and all of them were elevated in RPs. Based on WGCNA and three machine learning algorithms, all of five DEMMPs were regarded as hub DEMMPs. In vitro validation found the expression of hub DEMMPs also increased in renal tubular epithelial cells under lithogenic environment. RPs samples were divided into two clusters and cluster A exhibited higher expression of hub DEMMPs compared to cluster B. Functional enrichment analysis and GSEA found DEGs were enriched in immune-related functions and pathways. Moreover, increased infiltration of M1 macrophages and enhanced levels of inflammation were observed in cluster A by immune infiltration analysis.ConclusionWe assumed that MMPs might participate in RPs and stone formation through ECM degradation and macrophages-mediated immune response and inflammation. Our findings offer a novel perspective on the role of MMPs in immunity and urolithiasis for the first time, and provide potential biomarkers to develop targets for treatment and prevention.
Collapse
Affiliation(s)
- Sen-Yuan Hong
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong-Cheng Jiang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wen-Chao Xu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - He-Song Zeng
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shao-Gang Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Shao-Gang Wang, ; Bao-Long Qin,
| | - Bao-Long Qin
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Shao-Gang Wang, ; Bao-Long Qin,
| |
Collapse
|
38
|
Du LJ, Sun JY, Zhang WC, Liu Y, Liu Y, Lin WZ, Liu T, Zhu H, Wang YL, Shao S, Zhou LJ, Chen BY, Lu H, Li RG, Jia F, Duan SZ. NCOR1 maintains the homeostasis of vascular smooth muscle cells and protects against aortic aneurysm. Cell Death Differ 2023; 30:618-631. [PMID: 36151473 PMCID: PMC9984378 DOI: 10.1038/s41418-022-01065-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 09/11/2022] [Accepted: 09/12/2022] [Indexed: 11/08/2022] Open
Abstract
Phenotypic modulation of vascular smooth muscle cells (VSMCs) plays critical roles in the pathogenesis of aortic aneurysm (AA). The function of nuclear receptor corepressor1 (NCOR1) in regulation of VSMC phenotype and AA is unclear. Herein, using smooth muscle NCOR1 knockout mice, we demonstrated that smooth muscle NCOR1 deficiency decreased both mRNA and protein levels of contractile genes, impaired stress fibers formation and RhoA pathway activation, reduced synthesis of elastin and collagens, and induced the expression and activity of MMPs, manifesting a switch from contractile to degradative phenotype of VSMCs. NCOR1 modulated VSMC phenotype through 3 different mechanisms. First, NCOR1 deficiency increased acetylated FOXO3a to inhibit the expression of Myocd, which downregulated contractile genes. Second, deletion of NCOR1 derepressed NFAT5 to induce the expression of Rgs1, thus impeding RhoA activation. Third, NCOR1 deficiency increased the expression of Mmp12 and Mmp13 by derepressing ATF3. Finally, a mouse model combined apoE knockout mice with angiotensin II was used to study the role of smooth muscle NCOR1 in the development of AA. The results showed that smooth muscle NCOR1 deficiency increased the incidence of aortic aneurysms and exacerbated medial degeneration in angiotensin II-induced AA mouse model. Collectively, our data illustrated that NCOR1 interacts with FOXO3a, NFAT5, and ATF3 to maintain contractile phenotype of VSMCs and suppress AA development. Manipulation of smooth muscle NCOR1 may be a potential approach for AA treatment.
Collapse
Affiliation(s)
- Lin-Juan Du
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China
| | - Jian-Yong Sun
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China
| | - Wu-Chang Zhang
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China
| | - Yuan Liu
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China
| | - Yan Liu
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China
| | - Wen-Zhen Lin
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China
| | - Ting Liu
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China
| | - Hong Zhu
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China
| | - Yong-Li Wang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Shuai Shao
- Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Lu-Jun Zhou
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China
| | - Bo-Yan Chen
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China
| | - Hongjian Lu
- Department of Rehabilitation, Nantong First People's Hospital, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu, 226001, China
| | - Ruo-Gu Li
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| | - Feng Jia
- Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
- Department of Neurosurgery, Nantong First People's Hospital, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu, 226001, China.
| | - Sheng-Zhong Duan
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China.
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China.
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
39
|
Steinberger KJ, Eubank TD. The Underexplored Landscape of Hypoxia-Inducible Factor 2 Alpha and Potential Roles in Tumor Macrophages: A Review. OXYGEN (BASEL, SWITZERLAND) 2023; 3:45-76. [PMID: 37124241 PMCID: PMC10137047 DOI: 10.3390/oxygen3010005] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Low tissue oxygenation, termed hypoxia, is a characteristic of solid tumors with negative consequences. Tumor-associated macrophages (TAMs) accumulate in hypoxic tumor regions and correlate with worse outcomes in cancer patients across several tumor types. Thus, the molecular mechanism in which macrophages respond to low oxygen tension has been increasingly investigated in the last decade. Hypoxia stabilizes a group of hypoxia-inducible transcription factors (HIFs) reported to drive transcriptional programs involved in cell survival, metabolism, and angiogenesis. Though both tumor macrophage HIF-1α and HIF-2α correlate with unfavorable tumor microenvironments, most research focuses on HIF-1α as the master regulator of hypoxia signaling, because HIF-1α expression was originally identified in several cancer types and correlates with worse outcome in cancer patients. The relative contribution of each HIFα subunit to cell phenotypes is poorly understood especially in TAMs. Once thought to have overlapping roles, recent investigation of macrophage HIF-2α has demonstrated a diverse function from HIF-1α. Little work has been published on the differential role of hypoxia-dependent macrophage HIF-2α when compared to HIF-1α in the context of tumor biology. This review highlights cellular HIF-2α functions and emphasizes the gap in research investigating oxygen-dependent functions of tumor macrophage HIF-2α.
Collapse
Affiliation(s)
- Kayla J. Steinberger
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV 26505, USA
- In Vivo Multifunctional Magnetic Resonance Center, West Virginia University, Morgantown, WV 26505, USA
- West Virginia University Cancer Institute, Morgantown, WV 26505, USA
| | - Timothy D. Eubank
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV 26505, USA
- In Vivo Multifunctional Magnetic Resonance Center, West Virginia University, Morgantown, WV 26505, USA
- West Virginia University Cancer Institute, Morgantown, WV 26505, USA
| |
Collapse
|
40
|
Xu X, Hua X, Mo H, Hu S, Song J. Single-cell RNA sequencing to identify cellular heterogeneity and targets in cardiovascular diseases: from bench to bedside. Basic Res Cardiol 2023; 118:7. [PMID: 36750503 DOI: 10.1007/s00395-022-00972-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 12/09/2022] [Accepted: 12/09/2022] [Indexed: 02/09/2023]
Abstract
The mechanisms of cardiovascular diseases (CVDs) remain incompletely elucidated. Single-cell RNA sequencing (scRNA-seq) has enabled the profiling of single-cell transcriptomes at unprecedented resolution and throughput, which is critical for deciphering cardiovascular cellular heterogeneity and underlying disease mechanisms, thereby facilitating the development of therapeutic strategies. In this review, we summarize cellular heterogeneity in cardiovascular homeostasis and diseases as well as the discovery of potential disease targets based on scRNA-seq, and yield new insights into the promise of scRNA-seq technology in precision medicine and clinical application.
Collapse
Affiliation(s)
- Xinjie Xu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Xiumeng Hua
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Han Mo
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, 518057, China
| | - Shengshou Hu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.
| | - Jiangping Song
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.
| |
Collapse
|
41
|
Zhang Z, Zhuang J, Sun D, Ding Q, Zheng H, Li H, Zhang X, Du Y, Ma T, Meng Q. Netrin-1 Monoclonal Antibody-Functionalized Nanoparticle Loaded with Metformin Prevents the Progression of Abdominal Aortic Aneurysms. Int J Nanomedicine 2023; 18:627-639. [PMID: 36777816 PMCID: PMC9912973 DOI: 10.2147/ijn.s400993] [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: 12/10/2022] [Accepted: 02/01/2023] [Indexed: 02/09/2023] Open
Abstract
Background Abdominal aortic aneurysms (AAAs) are a global health and economic burden. Therapeutic strategies to inhibit the progression of AAAs are currently lacking. Recently, the therapeutic effect of metformin on aneurysms has attracted considerable interest. However, the unfavorable pharmacokinetic properties of metformin limit its feasibility for AAA treatment. Methods and Results We constructed a metformin-loaded netrin-1-responsive AAA-targeted nanoparticle (Tgt-NP-Met) for AAA management. Evaluation of the therapeutic effect of Tgt-NP-Met was performed by in vitro and in vivo experiments. Our results showed that the binding of netrin-1 monoclonal antibodies enhanced the AAA-targeting capability of nanoparticles (NPs). Moreover, Tgt-NP-Met administration prevented AAA development and reduced the aneurysm diameter in apolipoprotein E (ApoE)-deficient (ApoE-/-) mice that received continuous infusion of angiotensin II. Furthermore, metformin prevented AAA progression by inhibiting the transformation of vascular smooth muscle cells (VSMCs) from a contractile phenotype to a synthetic phenotype, which is mediated by macrophage infiltration and activation. Conclusion Our findings identify metformin as a functional suppressor for macrophage-mediated phenotypic transformation of VSMCs and Tgt-NP-Met as an efficient therapeutic strategy for AAA management.
Collapse
Affiliation(s)
- Zhiwei Zhang
- Department of Vascular Surgery, General Surgery Clinical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Jiawei Zhuang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, People’s Republic of China
| | - Daohan Sun
- Department of Vascular Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, People’s Republic of China
| | - Qingwei Ding
- Department of Vascular Surgery, General Surgery Clinical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Hui Zheng
- Department of Vascular Surgery, General Surgery Clinical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Haixiang Li
- Department of Vascular Surgery, General Surgery Clinical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Xiaoyu Zhang
- Department of Vascular Surgery, General Surgery Clinical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Yaming Du
- Department of Vascular Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, People’s Republic of China
| | - Teng Ma
- Department of Vascular Surgery, General Surgery Clinical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Qingyou Meng
- Department of Vascular Surgery, General Surgery Clinical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China,Correspondence: Qingyou Meng; Teng Ma, Email ;
| |
Collapse
|
42
|
Steffen BT, Pankow JS, Norby FL, Lutsey PL, Demmer RT, Guan W, Pankratz N, Li A, Liu G, Matsushita K, Tin A, Tang W. Proteomics Analysis of Genetic Liability of Abdominal Aortic Aneurysm Identifies Plasma Neogenin and Kit Ligand: The ARIC Study. Arterioscler Thromb Vasc Biol 2023; 43:367-378. [PMID: 36579647 PMCID: PMC9995137 DOI: 10.1161/atvbaha.122.317984] [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: 05/30/2022] [Accepted: 12/13/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUND Genome-wide association studies have reported 23 gene loci related to abdominal aortic aneurysm (AAA)-a potentially lethal condition characterized by a weakened dilated vessel wall. This study aimed to identify proteomic signatures and pathways related to these risk loci to better characterize AAA genetic susceptibility. METHODS Plasma concentrations of 4870 proteins were determined using a DNA aptamer-based array. Linear regression analysis estimated the associations between the 23 risk alleles and plasma protein levels with adjustments for potential confounders in a race-stratified analysis of 1671 Black and 7241 White participants. Significant proteins were then evaluated for their prediction of clinical AAA (454 AAA events in 11 064 individuals), and those significantly associated with AAA were further interrogated using Mendelian randomization analysis. RESULTS Risk variants proximal to PSRC1-CELSR2-SORT1, PCIF1-ZNF335-MMP9, RP11-136O12.2/TRIB1, ZNF259/APOA5, IL6R, PCSK9, LPA, and APOE were associated with 118 plasma proteins in Whites and 59 were replicated in Black participants. Novel associations with clinical AAA incidence were observed for kit ligand (HR, 0.59 [95% CI, 0.42-0.82] for top versus first quintiles) and neogenin (HR, 0.64 [95% CI, 0.46-0.88]) over a median 21.2-year follow-up; neogenin was also associated with ultrasound-detected asymptomatic AAA (N=4295; 57 asymptomatic AAA cases). Mendelian randomization inverse variance weighted estimates suggested that AAA risk is promoted by lower levels of kit ligand (OR per SD=0.67; P=1.4×10-5) and neogenin (OR per SD=0.50; P=0.03). CONCLUSIONS Low levels of neogenin and kit ligand may be novel risk factors for AAA development in potentially causal pathways. These findings provide insights and potential targets to reduce AAA susceptibility.
Collapse
Affiliation(s)
- Brian T. Steffen
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN 55454
- Division of Health Data Science, Department of Surgery, University of Minnesota, Minneapolis, MN 55455
| | - James S. Pankow
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN 55454
| | - Faye L. Norby
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Health System, Los Angeles, CA 90048
| | - Pamela L. Lutsey
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN 55454
| | - Ryan T. Demmer
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN 55454
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY 10032
| | - Weihua Guan
- Division of Biostatistics, University of Minnesota School of Public Health, Minneapolis, MN, 55455
| | - Nathan Pankratz
- Department of Laboratory Medicine & Pathology, University of Minnesota, Minneapolis, MN 55455
| | - Aixin Li
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN 55454
| | - Guning Liu
- Division of Epidemiology, Human Genetics and Environmental Sciences, The University of Texas Health Science Center, School of Public Health, Houston, TX 77030
| | - Kunihiro Matsushita
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205
- Welch Center for Prevention, Epidemiology and Clinical Research, Baltimore, MD 21205
| | - Adrienne Tin
- Division of Nephrology, Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216
| | - Weihong Tang
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN 55454
| |
Collapse
|
43
|
Wu S, Liu S, Wang B, Li M, Cheng C, Zhang H, Chen N, Guo X. Single-cell transcriptome in silico analysis reveals conserved regulatory programs in macrophages/monocytes of abdominal aortic aneurysm from multiple mouse models and human. Front Cardiovasc Med 2023; 9:1062106. [PMID: 36698942 PMCID: PMC9868255 DOI: 10.3389/fcvm.2022.1062106] [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: 10/05/2022] [Accepted: 12/16/2022] [Indexed: 01/10/2023] Open
Abstract
Abdominal aortic aneurysm (AAA) is a life-threatening disease and there is currently a lack of effective treatment to prevent it rupturing. ScRNA-seq studies of AAA are still lacking. In the study, we analyzed the published AAA scRNA-seq datasets from the mouse elastase-induced model, CaCl2 treatment model, Ang II-induced model and human by using bioinformatic approaches and in silico analysis. A total of 26 cell clusters were obtained and 11 cell types were identified from multiple mouse AAA models. Also, the proportion of Mφ/Mo increased in the AAA group and Mφ/Mo was divided into seven subtypes. There were significant differences in transcriptional regulation patterns of Mφ/Mo in different AAA models. The enrichment pathways of upregulated or downregulated genes from Mφ/Mo in the three mouse datasets were different. The actived regulons of Mφ/Mo had strong specificity and the repressed regulons showed high consistency. The co-upregulated genes as well as actived regulons and co-downregulated genes as well as repressed regulons were closely correlated and formed regulatory networks. Mφ/Mo from human AAA dataset was divided into five subtypes. The proportion of three macrophage subpopulations increased but the proportion of two monocyte subpopulations decreased. In the AAA group, the upregulated or downregulated genes of Mφ/Mo were enriched in different pathways. After further analyzing the genes in Mφ/Mo of both mouse and human scRNA-seq datasets, two genes were upregulated in the four datasets, IL-1B and THBS1. In conclusion, in silico analysis of scRNA-seq revealed that Mφ/Mo and their regulatory related genes as well as interaction networks played an important role in the pathogenesis of AAA.
Collapse
Affiliation(s)
- Shiyong Wu
- Department of Vascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shibiao Liu
- Department of Vascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Baoheng Wang
- Department of Vascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Meng Li
- Department of Vascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Chao Cheng
- Center for Genome Analysis, Wuhan Ruixing Biotechnology Co., Ltd., Wuhan, China
| | - Hairong Zhang
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China,*Correspondence: Hairong Zhang,
| | - Ningheng Chen
- Department of Vascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China,Ningheng Chen,
| | - Xueli Guo
- Department of Vascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China,Xueli Guo,
| |
Collapse
|
44
|
Lowis C, Ramara Winaya A, Kumari P, Rivera CF, Vlahos J, Hermantara R, Pratama MY, Ramkhelawon B. Mechanosignals in abdominal aortic aneurysms. Front Cardiovasc Med 2023; 9:1021934. [PMID: 36698932 PMCID: PMC9868277 DOI: 10.3389/fcvm.2022.1021934] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 11/29/2022] [Indexed: 01/11/2023] Open
Abstract
Cumulative evidence has shown that mechanical and frictional forces exert distinct effects in the multi-cellular aortic layers and play a significant role in the development of abdominal aortic aneurysms (AAA). These mechanical cues collectively trigger signaling cascades relying on mechanosensory cellular hubs that regulate vascular remodeling programs leading to the exaggerated degradation of the extracellular matrix (ECM), culminating in lethal aortic rupture. In this review, we provide an update and summarize the current understanding of the mechanotransduction networks in different cell types during AAA development. We focus on different mechanosensors and stressors that accumulate in the AAA sac and the mechanotransduction cascades that contribute to inflammation, oxidative stress, remodeling, and ECM degradation. We provide perspectives on manipulating this mechano-machinery as a new direction for future research in AAA.
Collapse
Affiliation(s)
- Christiana Lowis
- Division of Vascular and Endovascular Surgery, Department of Surgery, New York University Langone Medical Center, New York, NY, United States
- Department of Biomedicine, Indonesia International Institute for Life-Sciences, Jakarta, Indonesia
| | - Aurellia Ramara Winaya
- Division of Vascular and Endovascular Surgery, Department of Surgery, New York University Langone Medical Center, New York, NY, United States
- Department of Biomedicine, Indonesia International Institute for Life-Sciences, Jakarta, Indonesia
| | - Puja Kumari
- Division of Vascular and Endovascular Surgery, Department of Surgery, New York University Langone Medical Center, New York, NY, United States
- Department of Cell Biology, New York University Langone Medical Center, New York, NY, United States
| | - Cristobal F. Rivera
- Division of Vascular and Endovascular Surgery, Department of Surgery, New York University Langone Medical Center, New York, NY, United States
- Department of Cell Biology, New York University Langone Medical Center, New York, NY, United States
| | - John Vlahos
- Division of Vascular and Endovascular Surgery, Department of Surgery, New York University Langone Medical Center, New York, NY, United States
- Department of Cell Biology, New York University Langone Medical Center, New York, NY, United States
| | - Rio Hermantara
- Department of Biomedicine, Indonesia International Institute for Life-Sciences, Jakarta, Indonesia
| | - Muhammad Yogi Pratama
- Division of Vascular and Endovascular Surgery, Department of Surgery, New York University Langone Medical Center, New York, NY, United States
- Department of Cell Biology, New York University Langone Medical Center, New York, NY, United States
| | - Bhama Ramkhelawon
- Division of Vascular and Endovascular Surgery, Department of Surgery, New York University Langone Medical Center, New York, NY, United States
- Department of Cell Biology, New York University Langone Medical Center, New York, NY, United States
| |
Collapse
|
45
|
Han Y, Yan L, Xia L, Li S, Zhang Q, jin C. Global trends and Frontier topics about vascular smooth muscle cells phenotype switch: A bibliometric analysis from 1999 to 2021. Front Pharmacol 2022; 13:1004525. [PMID: 36452231 PMCID: PMC9702355 DOI: 10.3389/fphar.2022.1004525] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 11/02/2022] [Indexed: 03/31/2024] Open
Abstract
Objective: Vascular smooth muscle cell phenotype switch (VSMCPS) plays a significant role in vascular remodeling. This study aimed to conduct a bibliometric analysis and visualize the knowledge map of research on VSMCPS. Methods: We retrieved publications focusing on VSMCPS from the Web of Science Core Collection database (SCI-EXPANDED) from 1999 to 2021. Using bibliometric tools, VOSviewer and CiteSpace, we identified the most productive researchers, journals, institutions, and countries. At the same time, the trends, hot topics, and knowledge networks were analyzed and visualized. Results: A total of 2213 publications were included in this analysis. The number of annual publications in the VSMCPS field exhibited an upward trend and could be roughly divided into three phases. Until 2006, the most prolific authors were from the United States. As of 2008, the number of articles published in China increased dramatically to reach 126 papers in 2020. As of 2014, China was the most productive country in this field. The United States ranked first in the number of highly-influential authors, institutions, and literature from 1999 to 2022. Owens GK, Hata, Akiko, and Wen, jin-kun were the most prolific authors. Arteriosclerosis Thrombosis and Vascular Biology, Circulation Research, and Cardiovascular Research were the top-ranked journals in this field. "Vascular remodeling," "atherosclerosis," "neointima," "hypertension", and "inflammation" were the main researched topics. New diseases, new mechanisms, and new phenotype (e.g., micro RNA, macrophage-like-cell, hypoxia, autophagy, long noncoding RNA, oxidative stress, endoplasmic reticulum stress, senescence, aging, abdominal aortic aneurysm, and aortic dissection) represent the trending topics in recent years. Conclusion: This study systematically analyzed and visualized the knowledge map of VSMCPS over the past 2 decades. Our findings provide a comprehensive overview for scholars who want to understand current trends and new research frontiers in this area.
Collapse
Affiliation(s)
- Ying Han
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Langchao Yan
- Mini-invasive Neurosurgery and Translational Medical Center, Xi’an Central Hospital, Xi’an Jiaotong University, Xi’an, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Lu Xia
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Shifu Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Hypothalamic-Pituitary Research Center, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Qian Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Hypothalamic-Pituitary Research Center, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Chen jin
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Hypothalamic-Pituitary Research Center, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
46
|
Hu M, Qiu H, He T, Zhong M. Effect of miRNA-218-5p on Proliferation, Migration, Apoptosis and Inflammation of Vascular Smooth Muscle Cells in Abdominal Aortic Aneurysm and Extracellular Matrix Protein. IRANIAN JOURNAL OF PUBLIC HEALTH 2022; 51:2494-2503. [PMID: 36561253 PMCID: PMC9745393 DOI: 10.18502/ijph.v51i11.11166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 06/11/2022] [Indexed: 11/21/2022]
Abstract
Background To explore the effects of miRNA-218-5p on inflammation and extracellular matrix proteins of vascular smooth muscle cell line in abdominal aortic aneurysm (AAA). Methods miR-218-5p expression was detected with RT-qPCR. The proliferative activity of vascular smooth muscle cells (VSMCs) was detected with CCK-8, the migration was detected by Transwell, and the apoptosis was detected with flow cytometry. The expression levels of inflammatory factors (IL-1β and IL-18) were detected by ELISA. The expression levels of proteins (MMP-9 and Netrin-1) and ADAMTS5 were detected by Western blot. The targeting relationship between miR-218-5p and ADAMTS5 was verified with dual-luciferase reporter assay. Results Up-regulating miR-218-5p could significantly inhibit the proliferation and migration of VSMCs and induced the apoptosis (P<0.05). Down-regulating miR-218-5p could significantly promote the proliferation and migration of VSMCs and inhibit the apoptosis (P<0.05). Up-regulating miR-218-5p could inhibit the expression levels of THP-1 cytoinflammatory factors (IL-8 and IL-1β), MMP-9 and netrin-1. ADAMTS5 was the target gene of miR-218-5p. When there were both overexpression of ADAMTS5 and upregulation of miR-218-5p, the upregulation of miR-218-5p could alleviate the effects of overexpression of ADAMTS5 on the proliferation, migration and apoptosis of VSMCs. Conclusion miR-218-5p/ADAMTS-5 molecular axis regulates the proliferation, migration, and apoptosis of VSMCs, as well as the expression of THP-1 cell inflammatory molecules and extracellular matrix molecules.
Collapse
Affiliation(s)
- Min Hu
- Department of Vascular Surgery, Wuhan Central Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Hui Qiu
- Department of Oncology, Wuhan NO.1 Hospital, Wuhan 430000, China
| | - Tao He
- Department of Vascular Surgery, Wuhan Central Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China,Corresponding Author:
| | - Minyu Zhong
- Department of Oncology, Wuhan NO.1 Hospital, Wuhan 430000, China,Corresponding Author:
| |
Collapse
|
47
|
Barnault R, Verzeroli C, Fournier C, Michelet M, Redavid AR, Chicherova I, Plissonnier ML, Adrait A, Khomich O, Chapus F, Richaud M, Hervieu M, Reiterer V, Centonze FG, Lucifora J, Bartosch B, Rivoire M, Farhan H, Couté Y, Mirakaj V, Decaens T, Mehlen P, Gibert B, Zoulim F, Parent R. Hepatic inflammation elicits production of proinflammatory netrin-1 through exclusive activation of translation. Hepatology 2022; 76:1345-1359. [PMID: 35253915 DOI: 10.1002/hep.32446] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 02/16/2022] [Accepted: 02/17/2022] [Indexed: 12/18/2022]
Abstract
BACKGROUND AND AIMS Netrin-1 displays protumoral properties, though the pathological contexts and processes involved in its induction remain understudied. The liver is a major model of inflammation-associated cancer development, leading to HCC. APPROACH AND RESULTS A panel of cell biology and biochemistry approaches (reverse transcription quantitative polymerase chain reaction, reporter assays, run-on, polysome fractionation, cross linking immunoprecipitation, filter binding assay, subcellular fractionation, western blotting, immunoprecipitation, stable isotope labeling by amino acids in cell culture) on in vitro-grown primary hepatocytes, human liver cell lines, mouse samples and clinical samples was used. We identify netrin-1 as a hepatic inflammation-inducible factor and decipher its mode of activation through an exhaustive eliminative approach. We show that netrin-1 up-regulation relies on a hitherto unknown mode of induction, namely its exclusive translational activation. This process includes the transfer of NTN1 (netrin-1) mRNA to the endoplasmic reticulum and the direct interaction between the Staufen-1 protein and this transcript as well as netrin-1 mobilization from its cell-bound form. Finally, we explore the impact of a phase 2 clinical trial-tested humanized anti-netrin-1 antibody (NP137) in two distinct, toll-like receptor (TLR) 2/TLR3/TLR6-dependent, hepatic inflammatory mouse settings. We observe a clear anti-inflammatory activity indicating the proinflammatory impact of netrin-1 on several chemokines and Ly6C+ macrophages. CONCLUSIONS These results identify netrin-1 as an inflammation-inducible factor in the liver through an atypical mechanism as well as its contribution to hepatic inflammation.
Collapse
Affiliation(s)
- Romain Barnault
- Pathogenesis of Chronic Hepatitis B and C Laboratory - LabEx DEVweCAN, Inserm U1052, Cancer Research Centre of Lyon, Lyon, France.,University of Lyon, Lyon, France.,University Lyon 1, Institut des Sciences Pharmaceutiques et Biologiques, Lyon, France.,CNRS UMR5286, Lyon, France.,Centre Léon Bérard, Lyon, France
| | - Claire Verzeroli
- Pathogenesis of Chronic Hepatitis B and C Laboratory - LabEx DEVweCAN, Inserm U1052, Cancer Research Centre of Lyon, Lyon, France.,University of Lyon, Lyon, France.,University Lyon 1, Institut des Sciences Pharmaceutiques et Biologiques, Lyon, France.,CNRS UMR5286, Lyon, France.,Centre Léon Bérard, Lyon, France
| | - Carole Fournier
- Institute for Advanced Biosciences, Inserm U1209, University of Grenoble-Alpes, La Tronche, France
| | - Maud Michelet
- Pathogenesis of Chronic Hepatitis B and C Laboratory - LabEx DEVweCAN, Inserm U1052, Cancer Research Centre of Lyon, Lyon, France.,University of Lyon, Lyon, France.,University Lyon 1, Institut des Sciences Pharmaceutiques et Biologiques, Lyon, France.,CNRS UMR5286, Lyon, France.,Centre Léon Bérard, Lyon, France
| | - Anna Rita Redavid
- University of Lyon, Lyon, France.,University Lyon 1, Institut des Sciences Pharmaceutiques et Biologiques, Lyon, France.,CNRS UMR5286, Lyon, France.,Centre Léon Bérard, Lyon, France.,Apoptosis, Cancer and Development Laboratory - LabEx DEVweCAN, Inserm U1052, Cancer Research Centre of Lyon, Lyon, France
| | - Ievgeniia Chicherova
- Pathogenesis of Chronic Hepatitis B and C Laboratory - LabEx DEVweCAN, Inserm U1052, Cancer Research Centre of Lyon, Lyon, France.,University of Lyon, Lyon, France.,University Lyon 1, Institut des Sciences Pharmaceutiques et Biologiques, Lyon, France.,CNRS UMR5286, Lyon, France.,Centre Léon Bérard, Lyon, France
| | - Marie-Laure Plissonnier
- University of Lyon, Lyon, France.,University Lyon 1, Institut des Sciences Pharmaceutiques et Biologiques, Lyon, France.,CNRS UMR5286, Lyon, France.,Centre Léon Bérard, Lyon, France.,Inserm U1052, Cancer Research Centre of Lyon, Lyon, France
| | - Annie Adrait
- University of Grenoble-Alpes, Inserm, CEA, UMR BioSanté U1292, CNRS CEA FR2048, Grenoble, France
| | - Olga Khomich
- Pathogenesis of Chronic Hepatitis B and C Laboratory - LabEx DEVweCAN, Inserm U1052, Cancer Research Centre of Lyon, Lyon, France.,University of Lyon, Lyon, France.,University Lyon 1, Institut des Sciences Pharmaceutiques et Biologiques, Lyon, France.,CNRS UMR5286, Lyon, France.,Centre Léon Bérard, Lyon, France
| | - Fleur Chapus
- Single Cell Dynamics Group, Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, North Carolina, USA
| | - Mathieu Richaud
- University of Lyon, Lyon, France.,University Lyon 1, Institut des Sciences Pharmaceutiques et Biologiques, Lyon, France.,CNRS UMR5286, Lyon, France.,Centre Léon Bérard, Lyon, France.,Apoptosis, Cancer and Development Laboratory - LabEx DEVweCAN, Inserm U1052, Cancer Research Centre of Lyon, Lyon, France
| | - Maëva Hervieu
- University of Lyon, Lyon, France.,University Lyon 1, Institut des Sciences Pharmaceutiques et Biologiques, Lyon, France.,CNRS UMR5286, Lyon, France.,Centre Léon Bérard, Lyon, France.,Apoptosis, Cancer and Development Laboratory - LabEx DEVweCAN, Inserm U1052, Cancer Research Centre of Lyon, Lyon, France
| | - Veronika Reiterer
- Institute of Pathophysiology, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Julie Lucifora
- Pathogenesis of Chronic Hepatitis B and C Laboratory - LabEx DEVweCAN, Inserm U1052, Cancer Research Centre of Lyon, Lyon, France.,University of Lyon, Lyon, France.,University Lyon 1, Institut des Sciences Pharmaceutiques et Biologiques, Lyon, France.,CNRS UMR5286, Lyon, France.,Centre Léon Bérard, Lyon, France
| | - Birke Bartosch
- Pathogenesis of Chronic Hepatitis B and C Laboratory - LabEx DEVweCAN, Inserm U1052, Cancer Research Centre of Lyon, Lyon, France.,University of Lyon, Lyon, France.,University Lyon 1, Institut des Sciences Pharmaceutiques et Biologiques, Lyon, France.,CNRS UMR5286, Lyon, France.,Centre Léon Bérard, Lyon, France
| | - Michel Rivoire
- Léon Bérard Cancer Center, Lyon, France.,Université Lyon 1, Lyon, France
| | - Hesso Farhan
- Institute of Pathophysiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Yohann Couté
- University of Grenoble-Alpes, Inserm, CEA, UMR BioSanté U1292, CNRS CEA FR2048, Grenoble, France
| | - Valbona Mirakaj
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Tuebingen, Eberhard-Karls University, Tuebingen, Germany
| | - Thomas Decaens
- Institute for Advanced Biosciences, Inserm U1209, University of Grenoble-Alpes, La Tronche, France
| | - Patrick Mehlen
- University of Lyon, Lyon, France.,University Lyon 1, Institut des Sciences Pharmaceutiques et Biologiques, Lyon, France.,CNRS UMR5286, Lyon, France.,Centre Léon Bérard, Lyon, France.,Apoptosis, Cancer and Development Laboratory - LabEx DEVweCAN, Inserm U1052, Cancer Research Centre of Lyon, Lyon, France
| | - Benjamin Gibert
- University of Lyon, Lyon, France.,University Lyon 1, Institut des Sciences Pharmaceutiques et Biologiques, Lyon, France.,CNRS UMR5286, Lyon, France.,Centre Léon Bérard, Lyon, France.,Apoptosis, Cancer and Development Laboratory - LabEx DEVweCAN, Inserm U1052, Cancer Research Centre of Lyon, Lyon, France
| | - Fabien Zoulim
- Pathogenesis of Chronic Hepatitis B and C Laboratory - LabEx DEVweCAN, Inserm U1052, Cancer Research Centre of Lyon, Lyon, France.,University of Lyon, Lyon, France.,University Lyon 1, Institut des Sciences Pharmaceutiques et Biologiques, Lyon, France.,CNRS UMR5286, Lyon, France.,Centre Léon Bérard, Lyon, France.,Service of Hepato-Gastroenterology, Hospices Civils de Lyon, Lyon, France
| | - Romain Parent
- Pathogenesis of Chronic Hepatitis B and C Laboratory - LabEx DEVweCAN, Inserm U1052, Cancer Research Centre of Lyon, Lyon, France.,University of Lyon, Lyon, France.,University Lyon 1, Institut des Sciences Pharmaceutiques et Biologiques, Lyon, France.,CNRS UMR5286, Lyon, France.,Centre Léon Bérard, Lyon, France
| |
Collapse
|
48
|
He B, Zhan Y, Cai C, Yu D, Wei Q, Quan L, Huang D, Liu Y, Li Z, Liu L, Pan X. Common molecular mechanism and immune infiltration patterns of thoracic and abdominal aortic aneurysms. Front Immunol 2022; 13:1030976. [PMID: 36341412 PMCID: PMC9633949 DOI: 10.3389/fimmu.2022.1030976] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 10/10/2022] [Indexed: 01/02/2024] Open
Abstract
BACKGROUND Aortic disease (aortic aneurysm (AA), dissection (AD)) is a serious threat to patient lives. Little is currently known about the molecular mechanisms and immune infiltration patterns underlying the development and progression of thoracic and abdominal aortic aneurysms (TAA and AAA), warranting further research. METHODS We downloaded AA (includes TAA and AAA) datasets from the GEO database. The potential biomarkers in TAA and AAA were identified using differential expression analysis and two machine-learning algorithms. The discrimination power of the potential biomarkers and their diagnostic accuracy was assessed in validation datasets using ROC curve analysis. Then, GSEA, KEGG, GO and DO analyses were conducted. Furthermore, two immuno-infiltration analysis algorithms were utilized to analyze the common immune infiltration patterns in TAA and AAA. Finally, a retrospective clinical study was performed on 78 patients with AD, and the serum from 6 patients was used for whole exome sequencing (WES). RESULTS The intersection of TAA and AAA datasets yielded 82 differentially expressed genes (DEGs). Subsequently, the biomarkers (CX3CR1 and HBB) were acquired by screening using two machine-learning algorithms and ROC curve analysis. The functional analysis of DEGs showed significant enrichment in inflammation and regulation of angiogenic pathways. Immune cell infiltration analysis revealed that adaptive and innate immune responses were closely linked to AA progression. However, neither CX3CR1 nor HBB was associated with B cell-mediated humoral immunity. CX3CR1 expression was correlated with macrophages and HBB with eosinophils. Finally, our retrospective clinical study revealed a hyperinflammatory environment in aortic disease. The WES study identified disease biomarkers and gene variants, some of which may be druggable. CONCLUSION The genes CX3CR1 and HBB can be used as common biomarkers in TAA and AAA. Large numbers of innate and adaptive immune cells are infiltrated in AA and are closely linked to the development and progression of AA. Moreover, CX3CR1 and HBB are highly correlated with the infiltration of immune cells and may be potential targets of immunotherapeutic drugs. Gene mutation research is a promising direction for the treatment of aortic disease.
Collapse
Affiliation(s)
- Bin He
- Graduate School of Youjiang Medical University for Nationalities, Baise, China
| | - Ya Zhan
- The Third Hospital of MianYang, Sichuan Mental Health Center, MianYang, China
| | - Chunyu Cai
- Graduate School of Youjiang Medical University for Nationalities, Baise, China
| | - Dianyou Yu
- Graduate School of Youjiang Medical University for Nationalities, Baise, China
| | - Qinjiang Wei
- Department of Cardiology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Liping Quan
- Graduate School of Youjiang Medical University for Nationalities, Baise, China
| | - Da Huang
- Department of Cardiology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Yan Liu
- Department of Cardiology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Zhile Li
- Department of Cardiology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Li Liu
- Department of Cardiology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
- College of Clinical Medicine, Youjiang Medical University for Nationalities, Baise, China
| | - Xingshou Pan
- Department of Cardiology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| |
Collapse
|
49
|
Márquez-Sánchez AC, Koltsova EK. Immune and inflammatory mechanisms of abdominal aortic aneurysm. Front Immunol 2022; 13:989933. [PMID: 36275758 PMCID: PMC9583679 DOI: 10.3389/fimmu.2022.989933] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 08/23/2022] [Indexed: 11/13/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) is a life-threatening cardiovascular disease. Immune-mediated infiltration and a destruction of the aortic wall during AAA development plays significant role in the pathogenesis of this disease. While various immune cells had been found in AAA, the mechanisms of their activation and function are still far from being understood. A better understanding of mechanisms regulating the development of aberrant immune cell activation in AAA is essential for the development of novel preventive and therapeutic approaches. In this review we summarize current knowledge about the role of immune cells in AAA and discuss how pathogenic immune cell activation is regulated in this disease.
Collapse
|
50
|
Silvestro M, Rivera CF, Alebrahim D, Vlahos J, Pratama MY, Lu C, Tang C, Harpel Z, Sleiman Tellaoui R, Zias AL, Maldonado DJ, Byrd D, Attur M, Mignatti P, Ramkhelawon B. The Nonproteolytic Intracellular Domain of Membrane-Type 1 Matrix Metalloproteinase Coordinately Modulates Abdominal Aortic Aneurysm and Atherosclerosis in Mice-Brief Report. Arterioscler Thromb Vasc Biol 2022; 42:1244-1253. [PMID: 36073351 PMCID: PMC9993845 DOI: 10.1161/atvbaha.122.317686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND MT1-MMP (membrane-type 1 matrix metalloproteinase, MMP-14) is a transmembrane-anchored protein with an extracellular proteinase domain and a cytoplasmic tail devoid of proteolytic functions but capable of mediating intracellular signaling that regulates tissue homeostasis. MT1-MMP extracellular proteolytic activity has been shown to regulate pathological remodeling in aortic aneurysm and atherosclerosis. However, the role of the nonproteolytic intracellular domain of MT1-MMP in vascular remodeling in abdominal aortic aneurysms (AAA) is unknown. METHODS We generated a mutant mouse that harbors a point mutation (Y573D) in the MT1-MMP cytoplasmic domain that abrogates the MT1-MMP signaling function without affecting its proteolytic activity. These mice and their control wild-type littermates were subjected to experimental AAA modeled by angiotensin II infusion combined with PCSK9 (proprotein convertase subtilisin/kexin type 9) overexpression and high-cholesterol feeding. RESULTS The mutant mice developed more severe AAA than the control mice, with concomitant generation of intraaneurysmal atherosclerotic lesions and dramatically increased macrophage infiltration and elastin degradation. Aortic lesion-associated and bone marrow-derived macrophages from the mutant mice exhibited an enhanced inflammatory state and expressed elevated levels of proinflammatory Netrin-1, a protein previously demonstrated to promote both atherosclerosis and AAA. CONCLUSIONS Our findings show that the cytoplasmic domain of MT1-MMP safeguards from AAA and atherosclerotic plaque development through a proteolysis-independent signaling mechanism associated with Netrin-1 expression. This unexpected function of MT1-MMP unveils a novel mechanism of synchronous onset of AAA and atherogenesis and highlights its importance in the control of vascular wall homeostasis.
Collapse
Affiliation(s)
- Michele Silvestro
- Division of Vascular and Endovascular Surgery, Department of Surgery (M.S., C.F.R., D.A., J.V., M.Y.P., C.T., Z.H., R.S.T., A.L.Z., D.J.M., D.B., B.R.)
| | - Cristobal F Rivera
- Division of Vascular and Endovascular Surgery, Department of Surgery (M.S., C.F.R., D.A., J.V., M.Y.P., C.T., Z.H., R.S.T., A.L.Z., D.J.M., D.B., B.R.)
| | - Dornazsadat Alebrahim
- Division of Vascular and Endovascular Surgery, Department of Surgery (M.S., C.F.R., D.A., J.V., M.Y.P., C.T., Z.H., R.S.T., A.L.Z., D.J.M., D.B., B.R.)
| | - John Vlahos
- Division of Vascular and Endovascular Surgery, Department of Surgery (M.S., C.F.R., D.A., J.V., M.Y.P., C.T., Z.H., R.S.T., A.L.Z., D.J.M., D.B., B.R.)
| | - Muhammad Yogi Pratama
- Division of Vascular and Endovascular Surgery, Department of Surgery (M.S., C.F.R., D.A., J.V., M.Y.P., C.T., Z.H., R.S.T., A.L.Z., D.J.M., D.B., B.R.)
| | - Cuijie Lu
- Division of Vascular and Endovascular Surgery, Department of Surgery (M.S., C.F.R., D.A., J.V., M.Y.P., C.T., Z.H., R.S.T., A.L.Z., D.J.M., D.B., B.R.).,Division of Rheumatology, Department of Medicine (C.L., M.A., P.M.), New York University Langone Medical Center, New York
| | - Claudia Tang
- Division of Vascular and Endovascular Surgery, Department of Surgery (M.S., C.F.R., D.A., J.V., M.Y.P., C.T., Z.H., R.S.T., A.L.Z., D.J.M., D.B., B.R.)
| | - Zander Harpel
- Division of Vascular and Endovascular Surgery, Department of Surgery (M.S., C.F.R., D.A., J.V., M.Y.P., C.T., Z.H., R.S.T., A.L.Z., D.J.M., D.B., B.R.)
| | - Rayan Sleiman Tellaoui
- Division of Vascular and Endovascular Surgery, Department of Surgery (M.S., C.F.R., D.A., J.V., M.Y.P., C.T., Z.H., R.S.T., A.L.Z., D.J.M., D.B., B.R.)
| | - Ariadne L Zias
- Division of Vascular and Endovascular Surgery, Department of Surgery (M.S., C.F.R., D.A., J.V., M.Y.P., C.T., Z.H., R.S.T., A.L.Z., D.J.M., D.B., B.R.)
| | - Delphina J Maldonado
- Division of Vascular and Endovascular Surgery, Department of Surgery (M.S., C.F.R., D.A., J.V., M.Y.P., C.T., Z.H., R.S.T., A.L.Z., D.J.M., D.B., B.R.)
| | - Devon Byrd
- Division of Vascular and Endovascular Surgery, Department of Surgery (M.S., C.F.R., D.A., J.V., M.Y.P., C.T., Z.H., R.S.T., A.L.Z., D.J.M., D.B., B.R.)
| | - Mukundan Attur
- Division of Rheumatology, Department of Medicine (C.L., M.A., P.M.), New York University Langone Medical Center, New York
| | - Paolo Mignatti
- Division of Rheumatology, Department of Medicine (C.L., M.A., P.M.), New York University Langone Medical Center, New York.,Department of Cell Biology (P.M., B.R.), New York University Langone Medical Center, New York
| | - Bhama Ramkhelawon
- Division of Vascular and Endovascular Surgery, Department of Surgery (M.S., C.F.R., D.A., J.V., M.Y.P., C.T., Z.H., R.S.T., A.L.Z., D.J.M., D.B., B.R.).,Department of Cell Biology (P.M., B.R.), New York University Langone Medical Center, New York
| |
Collapse
|