1
|
Bao Y, Chen J, Han X, He Y, Yang T, Shi X, Chen J, Gu L, Wang S, Xie L, Wang H, Wang L. Calbindin 2 as a Novel Biomarker and Therapeutic Target for Abdominal Aortic Aneurysm: Integrative Analysis of Human Proteomes and Genetics. J Am Heart Assoc 2025; 14:e039195. [PMID: 40314374 DOI: 10.1161/jaha.124.039195] [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: 10/01/2024] [Accepted: 04/08/2025] [Indexed: 05/03/2025]
Abstract
BACKGROUND Abdominal aortic aneurysm (AAA) is a clinical life-threatening issue. No pharmacological treatments are currently approved for the prevention and treatment of AAA. Therefore, identifying novel biomarkers and therapeutic targets is crucial for improving AAA management and outcomes. METHODS To identify plasma proteins with potential causal effects on AAA, we integrated genetic evidence from proteome-wide Mendelian randomization, genetic correlation, and colocalization analysis. The role of identified proteins in AAA was further explored through the phenome-wide association study and mediation analysis. Multiomics data analysis, including bulk RNA sequencing, single-cell/single-nucleus RNA sequencing, and spatial transcriptomics, was employed to characterize the expression patterns of these proteins. Experimental validation was performed using an AAA model in apolipoprotein E-deficient mice infused with angiotensin II. Druggability analysis was conducted to identify drug candidates, which were tested in preclinical mouse models. RESULTS CALB2 (calbindin 2) was identified as having a causal effect on AAA and may influence the progression of AAA through the regulation of lipid metabolism. Multiomics analysis revealed that CALB2 is predominantly expressed in the mesothelial cells of adipose tissues. Inhibition of CALB2 in an AAA mouse model alleviated AAA progression. Druggability analysis identified lenalidomide and genistein as potential therapeutic candidates, and experiments confirmed their efficacy in preventing AAA development. CONCLUSIONS This study identifies CALB2 as being associated with an increased risk of AAA and suggests that i might be a novel biomarker and therapeutic molecule for AAA management. Lenalidomide and genistein hold promising potential as treatments for patients with AAA.
Collapse
Affiliation(s)
- Yulin Bao
- Department of Cardiology The First Affiliated Hospital with Nanjing Medical University Nanjing Jiangsu China
| | - Jiayi Chen
- Department of Cardiology The First Affiliated Hospital with Nanjing Medical University Nanjing Jiangsu China
| | - Xudong Han
- Department of Cardiology The First Affiliated Hospital with Nanjing Medical University Nanjing Jiangsu China
| | - Ye He
- Department of Cardiology The First Affiliated Hospital with Nanjing Medical University Nanjing Jiangsu China
| | - Tongtong Yang
- Department of Cardiology The First Affiliated Hospital with Nanjing Medical University Nanjing Jiangsu China
| | - Xinying Shi
- Department of Cardiology The First Affiliated Hospital with Nanjing Medical University Nanjing Jiangsu China
| | - Jiawen Chen
- Department of Cardiology The First Affiliated Hospital with Nanjing Medical University Nanjing Jiangsu China
| | - Lingfeng Gu
- Department of Cardiology The First Affiliated Hospital with Nanjing Medical University Nanjing Jiangsu China
| | - Sibo Wang
- Department of Cardiology The First Affiliated Hospital with Nanjing Medical University Nanjing Jiangsu China
| | - Liping Xie
- 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
| | - Hao Wang
- Department of Cardiology The First Affiliated Hospital with Nanjing Medical University Nanjing Jiangsu China
| | - Liansheng Wang
- Department of Cardiology The First Affiliated Hospital with Nanjing Medical University Nanjing Jiangsu China
| |
Collapse
|
2
|
Heller K, Doukas P, Uhl C, Gombert A. Sex-Specific Characteristics of Perivascular Fat in Aortic Aneurysms. J Clin Med 2025; 14:3071. [PMID: 40364103 PMCID: PMC12072766 DOI: 10.3390/jcm14093071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 04/17/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025] Open
Abstract
Aortic aneurysms (AAs), the dilation or widening of the aorta, lead to dissection or rupture with high morbidity and mortality if untreated. AA displays gender disparities in its prevalence, progression and outcomes, with women having worse outcomes and faster aneurysm growth. However, current guidelines do not address gender dimorphism, emphasizing the urgent need for personalized treatment strategies and further research. Perivascular adipose tissue (PVAT), a unique type of fat surrounding blood vessels, plays a critical role in maintaining vasomotor tone and vascular homeostasis, with dysfunction associated with chronic inflammation and vessel-wall remodeling. Indeed, PVAT dysfunction promotes the development of aortic aneurysms, with hormonal and biomechanical factors exacerbating the pathological vascular microenvironment. The sexually dimorphic characteristics of PVAT include morphological, immunological, and hormonally mediated differences. Thus, targeting PVAT-mediated mechanisms may be a promising option for the (gender-specific) therapeutic management of cardiovascular pathologies. This review examines the emerging importance of PVAT in vascular health, its potential therapeutic implications for AA, and identifies gaps in the current state of research.
Collapse
Affiliation(s)
- Katja Heller
- Department of Vascular Surgery, RWTH Aachen University Hospital, 52074 Aachen, Germany; (P.D.); (C.U.); (A.G.)
| | | | | | | |
Collapse
|
3
|
Wang Y, Zhang X, Li X, Cheng M, Cui X. The vascular microenvironment and its stem cells regulate vascular homeostasis. Front Cell Dev Biol 2025; 13:1544129. [PMID: 40114970 PMCID: PMC11922910 DOI: 10.3389/fcell.2025.1544129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 02/17/2025] [Indexed: 03/22/2025] Open
Abstract
The vascular microenvironment comprises of anatomical structures, extracellular matrix components, and various cell populations, which play a crucial role in regulating vascular homeostasis and influencing vascular structure and function. Under physiological conditions, intrinsic regulation of the vascular microenvironment is required to sustain vascular homeostasis. In contrast, under pathological conditions, alterations to this microenvironment lead to vascular injury and pathological remodeling. According to the anatomy, the vascular microenvironment can be subdivided into three sections from the inside out. The vascular endothelial microenvironment, centered on vascular endothelial cells (VECs), includes the extracellular matrix and various vascular physicochemical factors. The VECs interact with vascular physicochemical factors to regulate the function of various parenchymal cells, including hepatocytes, neurons and tumor cells. The vascular wall microenvironment, comprising the vasa vasorum and their unique stem/progenitor cell niches, plays a pivotal role in vascular inflammation and pathological remodeling. Additionally, the perivascular microenvironment, which includes perivascular adipose tissue, consists of adipocytes and stem cells, which contribute to the pathological processes of atherosclerosis. It is anticipated that targeted regulation of the vascular microenvironment will emerge as a novel approach for the treatment of various diseases. Accordingly, this review will examine the structure of the vascular microenvironment, the regulation of vascular function by vascular cells and stem/progenitor cells, and the role of the vascular microenvironment in regulating cardiovascular diseases.
Collapse
Affiliation(s)
- Yanhui Wang
- Medical Physiology Laboratory, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| | - Xiaoyun Zhang
- Medical Physiology Laboratory, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| | - Xin Li
- Medical Physiology Laboratory, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| | - Min Cheng
- Medical Physiology Laboratory, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| | - Xiaodong Cui
- Medical Physiology Laboratory, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| |
Collapse
|
4
|
Zhu Q, Luo D, Li Y, Yu L, Zhang Z, Ouyang F, Li L, Lu M, Hu C, Dong Y, Ma C, Liang Y, Zhao TJ, Chen FJ, Li P, Yang TS. CIDEC/FSP27 exacerbates obesity-related abdominal aortic aneurysm by promoting perivascular adipose tissue inflammation. LIFE METABOLISM 2025; 4:loae035. [PMID: 39872985 PMCID: PMC11770823 DOI: 10.1093/lifemeta/loae035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 08/23/2024] [Accepted: 09/17/2024] [Indexed: 01/30/2025]
Abstract
Abdominal aortic aneurysm (AAA) is strongly correlated with obesity, partially due to the abnormal expansion of abdominal perivascular adipose tissue (PVAT). Cell death-inducing DNA fragmentation factor-like effector C (CIDEC), also known as fat-specific protein 27 (FSP27) in rodents, is specifically expressed in adipose tissue where it mediates lipid droplet fusion and adipose tissue expansion. Whether and how CIDEC/FSP27 plays a role in AAA pathology remains elusive. Here, we show that FSP27 exacerbates obesity and angiotensin Ⅱ (Ang Ⅱ)-induced AAA progression. FSP27 deficiency in mice inhibited high-fat diet-induced PVAT expansion and inflammation. Both global and adipose tissue-specific FSP27 ablation significantly decreased obesity-related AAA incidence. Deficiency of FSP27 in adipocytes abrogated matrix metalloproteinase-12 (MMP12) expression in aortic tissues. Infiltrated macrophages, which partially colocalize with MMP12, were significantly decreased in the FSP27-deficient aorta. Mechanistically, knockdown of Fsp27 in 3T3-L1 adipocytes inhibited C-C motif chemokine ligand 2 (CCL2) expression and secretion through a c-Jun N-terminal kinase (JNK)-dependent pathway, thereby leading to reduced induction of macrophage migration, while Cidec overexpression rescued this effect. Overall, our study demonstrates that CIDEC/FSP27 in adipose tissue contributes to obesity-related AAA formation, at least in part, by enhancing PVAT inflammation and macrophage infiltration, thus shedding light on its significance as a key regulator in the context of obesity-related AAA.
Collapse
Affiliation(s)
- Qing Zhu
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
| | - Da Luo
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
- Shanghai Qi Zhi Institute, Shanghai 200030, China
| | - Yining Li
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
| | - Liyang Yu
- State Key Laboratory of Membrane Biology and Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100086, China
| | - Zixuan Zhang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
| | - Feng Ouyang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
| | - Liangkui Li
- State Key Laboratory of Membrane Biology and Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100086, China
| | - Manxi Lu
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
| | - Changyong Hu
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
| | - Yinuo Dong
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
| | - Chengxin Ma
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
| | - Yan Liang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
| | - Tong-Jin Zhao
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
| | - Feng-Jung Chen
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
- Shanghai Qi Zhi Institute, Shanghai 200030, China
| | - Peng Li
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
- Shanghai Qi Zhi Institute, Shanghai 200030, China
- State Key Laboratory of Membrane Biology and Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100086, China
- School of Life Sciences, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Tian-Shu Yang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
- Shanghai Qi Zhi Institute, Shanghai 200030, China
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
5
|
Zhang ZB, Cheng YW, Xu L, Li JQ, Pan X, Zhu M, Chen XH, Sun AJ, Lin JR, Gao PJ. Activation of β3-adrenergic receptor by mirabegron prevents aortic dissection/aneurysm by promoting lymphangiogenesis in perivascular adipose tissue. Cardiovasc Res 2024; 120:2307-2319. [PMID: 39288197 DOI: 10.1093/cvr/cvae213] [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: 01/19/2024] [Revised: 05/06/2024] [Accepted: 07/23/2024] [Indexed: 09/19/2024] Open
Abstract
AIMS β3-Adrenergic receptor (β3-AR) is essential for cardiovascular homoeostasis through regulating adipose tissue function. Perivascular adipose tissue (PVAT) has been implicated in the pathogenesis of aortic dissection and aneurysm (AD/AA). Here, we aim to investigate β3-AR activation-mediated PVAT function in AD/AA. METHODS AND RESULTS Aortas from patients with thoracic aortic dissection (TAD) were collected to detect β3-AR expression in PVAT. ApoE-/- and β-aminopropionitrile monofumarate (BAPN)-treated C57BL/6 mice were induced with Angiotensin II (AngII) to simulate AD/AA and subsequently received either placebo or mirabegron, a β3-AR agonist. The results demonstrated an up-regulation of β3-AR in PVAT of TAD patients and AD/AA mice. Moreover, activation of β3-AR by mirabegron significantly prevented AngII-induced AD/AA formation in mice. RNA-sequencing analysis of adipocytes from PVAT revealed a notable increase of the lymphangiogenic factor, vascular endothelial growth factor C (VEGF-C), in mirabegron-treated mice. Consistently, enhanced lymphangiogenesis was found in PVAT with mirabegron treatment. Mechanistically, the number of CD4+/CD8+ T cells and CD11c+ cells was reduced in PVAT but increased in adjacent draining lymph nodes of mirabegron-treated mice, indicating the improved draining and clearance of inflammatory cells in PVAT by lymphangiogenesis. Importantly, adipocyte-specific VEGF-C knockdown by the adeno-associated virus system restrained lymphangiogenesis and exacerbated inflammatory cell infiltration in PVAT, which ultimately abolished the protection of mirabegron on AD/AA. In addition, the conditional medium derived from mirabegron-treated adipocytes activated the proliferation and tube formation of LECs, which was abrogated by the silencing of VEGF-C in adipocytes. CONCLUSION Our findings illustrated the therapeutic potential of β3-AR activation by mirabegron on AD/AA, which promoted lymphangiogenesis by increasing adipocyte-derived VEGF-C and, therefore, ameliorated PVAT inflammation.
Collapse
MESH Headings
- Animals
- Aortic Dissection/metabolism
- Aortic Dissection/pathology
- Aortic Dissection/prevention & control
- Aortic Dissection/chemically induced
- Aortic Dissection/physiopathology
- Aortic Dissection/genetics
- Lymphangiogenesis/drug effects
- Adrenergic beta-3 Receptor Agonists/pharmacology
- Mice, Inbred C57BL
- Receptors, Adrenergic, beta-3/metabolism
- Receptors, Adrenergic, beta-3/genetics
- Acetanilides/pharmacology
- Vascular Endothelial Growth Factor C/metabolism
- Vascular Endothelial Growth Factor C/genetics
- Humans
- Male
- Disease Models, Animal
- Adipose Tissue/metabolism
- Adipose Tissue/drug effects
- Adipose Tissue/pathology
- Aortic Aneurysm, Thoracic/metabolism
- Aortic Aneurysm, Thoracic/pathology
- Aortic Aneurysm, Thoracic/prevention & control
- Aortic Aneurysm, Thoracic/physiopathology
- Aortic Aneurysm, Thoracic/genetics
- Aortic Aneurysm, Thoracic/chemically induced
- Aortic Aneurysm, Thoracic/drug therapy
- Mice, Knockout, ApoE
- Thiazoles/pharmacology
- Signal Transduction
- Aorta, Thoracic/metabolism
- Aorta, Thoracic/pathology
- Aorta, Thoracic/physiopathology
- Aorta, Thoracic/drug effects
- Angiotensin II
- Adipocytes/metabolism
- Adipocytes/pathology
- Adipocytes/drug effects
- Cells, Cultured
- Mice
- Lymphatic Vessels/metabolism
- Lymphatic Vessels/drug effects
- Lymphatic Vessels/pathology
- Lymphatic Vessels/physiopathology
- Female
- Middle Aged
Collapse
Affiliation(s)
- Ze-Bei Zhang
- 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, 197 Ruijin, 2 Road, Shanghai 200025, China
| | - Yu-Wen Cheng
- 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, 197 Ruijin, 2 Road, Shanghai 200025, China
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lian Xu
- 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, 197 Ruijin, 2 Road, Shanghai 200025, China
| | - Jia-Qi Li
- 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, 197 Ruijin, 2 Road, Shanghai 200025, China
| | - Xin Pan
- Department of Gerontology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Min Zhu
- 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, 197 Ruijin, 2 Road, Shanghai 200025, China
| | - Xiao-Hui Chen
- 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, 197 Ruijin, 2 Road, Shanghai 200025, China
| | - Ai-Jun Sun
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | - Jing-Rong Lin
- 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, 197 Ruijin, 2 Road, Shanghai 200025, China
| | - Ping-Jin Gao
- 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, 197 Ruijin, 2 Road, Shanghai 200025, China
| |
Collapse
|
6
|
Queiroz M, Sena CM. Perivascular adipose tissue: a central player in the triad of diabetes, obesity, and cardiovascular health. Cardiovasc Diabetol 2024; 23:455. [PMID: 39732729 PMCID: PMC11682657 DOI: 10.1186/s12933-024-02549-9] [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: 09/14/2024] [Accepted: 12/17/2024] [Indexed: 12/30/2024] Open
Abstract
Perivascular adipose tissue (PVAT) is a dynamic tissue that affects vascular function and cardiovascular health. The connection between PVAT, the immune system, obesity, and vascular disease is complex and plays a pivotal role in the pathogenesis of vascular diseases such as atherosclerosis, hypertension, and vascular inflammation. In cardiometabolic diseases, PVAT becomes a significant source of proflammatory adipokines, leading to increased infiltration of immune cells, in cardiometabolic diseases, PVAT becomes a significant source of proinflammatory adipokines, leading to increased infiltration of immune cells, promoting vascular smooth muscle cell proliferation and migrationpromoting vascular smooth muscle cell proliferation and migration. This exacerbates vascular dysfunction by impairing endothelial cell function and promoting endothelial activation. Dysregulated PVAT also contributes to hemodynamic alterations and hypertension through enhanced sympathetic nervous system activity and impaired vasodilatory capacity of PVAT-derived factors. Therapeutic interventions targeting key components of this interaction, such as modulating PVAT inflammation, restoring adipokine balance, and attenuating immune cell activation, hold promise for mitigating obesity-related vascular complications. Lifestyle interventions, pharmacological agents targeting inflammatory pathways, and surgical approaches aimed at reducing PVAT mass or improving adipose tissue function are potential therapeutic avenues for managing vascular diseases associated with obesity and PVAT dysfunction.
Collapse
Affiliation(s)
- Marcelo Queiroz
- Institute of Physiology, iCBR, Faculty of Medicine, University of Coimbra, Subunit 1, polo 3, Azinhaga de Santa Comba, Celas, 3000-548, Coimbra, Portugal
| | - Cristina M Sena
- Institute of Physiology, iCBR, Faculty of Medicine, University of Coimbra, Subunit 1, polo 3, Azinhaga de Santa Comba, Celas, 3000-548, Coimbra, Portugal.
| |
Collapse
|
7
|
Guo E, Liu D, Zhu Z. Phenotypic and functional disparities in perivascular adipose tissue. Front Physiol 2024; 15:1499340. [PMID: 39588268 PMCID: PMC11586278 DOI: 10.3389/fphys.2024.1499340] [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: 09/20/2024] [Accepted: 10/29/2024] [Indexed: 11/27/2024] Open
Abstract
The adipose tissue surrounding blood vessels is known as perivascular adipose tissue (PVAT), which represents a distinct ectopic fat depot that adheres to the majority of the vasculature. In recent years, owing to its unique location and function, PVAT has been regarded as a new type of adipose tissue distinct from traditional visceral fat. It releases adipokines with vasoconstrictive functions, which regulate vascular function through paracrine and endocrine mechanisms. Interestingly, PVAT can be categorized as white, brown or a mixture of both depending on its anatomical location. Brown adipose tissue (BAT) is located adjacent to the thoracic aorta in rodents, while a mix of brown and white tissue surrounds the abdominal aorta. PVAT exhibits regional phenotypic differences in different parts of the vasculature bed, which may lead to heterogeneity in the secretion profiles and norepinephrine (NE) content in regional PVAT and subsequently affect the regulation of specific adipokine signaling pathways in regional PVAT, resulting in differences in the regulation of vascular function. The aim of this review was to explore the potential factors that influence the anticontractile function of regional PVAT in the vasculature, including the heterogeneity of regional PVAT, the anticontractile function mediated by endothelial nitric oxide synthase (eNOS) in regional PVAT, the activity of the adiponectin-eNOS pathway in regional PVAT adipocytes, and the concentration of the sympathetic neurotransmitter NE in regional PVAT.
Collapse
Affiliation(s)
- Erling Guo
- College of Physical Education, Guangzhou College of Commerce, Guangzhou, China
| | - Dan Liu
- Primary Department International Division, Tsinghua International School Daoxiang Lake, Beijing, China
| | - Ziming Zhu
- International College, Guangzhou College of Commerce, Guangzhou, China
| |
Collapse
|
8
|
Ginzburg D, Nowak S, Attenberger U, Luetkens J, Sprinkart AM, Kuetting D. Computer tomography-based assessment of perivascular adipose tissue in patients with abdominal aortic aneurysms. Sci Rep 2024; 14:20512. [PMID: 39227666 PMCID: PMC11372190 DOI: 10.1038/s41598-024-71283-9] [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: 10/05/2023] [Accepted: 08/27/2024] [Indexed: 09/05/2024] Open
Abstract
This retrospective study investigates perivascular adipose tissue (PVAT) alterations in CT as a marker of inflammation in patients with abdominal aortic aneurysms (AAA). 100 abdominal CT scans of patients with abdominal aortic aneurysms and 100 age and sex matched controls without underlying aortic disease were included. Artificial Intelligence (AI) assisted segmentation of the aorta and the surrounding adipose tissue was performed. Adipose tissue density was measured in Hounsfield units (HU) close (2-5mm, HUclose) and distant (10-12mm, HUdistant) to the aortic wall. To investigate alterations in adipose tissue density close to the aorta (HUclose) as a potential marker of inflammation, we calculated the difference HUΔ = HUclose-HUdistant and the fat attenuation ratio HUratio = HUclose/HUdistant as normalized attenuation measures. These two markers were compared i) inter-individually between AAA patients and controls and ii) intra-individually between the aneurysmal and non-aneurysmal segments in AAA patients. Since most AAAs are generally observed infrarenal, the aneurysmal section of the AAA patients was compared with the infrarenal section of the aorta of the control patients. In inter-individual comparisons, higher HUΔ and a lower HUratio were observed (aneurysmal: 8.9 ± 5.1 HU vs. control: 6.9 ± 4.8 HU, p-value = 0.006; aneurysmal: 89.8 ± 5.7% vs. control: 92.1 ± 5.5% p-value = 0.004). In intra-individual comparisons, higher HUΔ and lower HUratio were observed (aneurysmal: 8.9 ± 5.1 HU vs. non-aneurysmal: 5.5 ± 4.1 HU, p-value < 0.001; aneurysmal: 89.8 ± 5.7% vs. non-aneurysmal 93.3 ± 4.9%, p-value < 0.001). The results indicate PVAT density alterations in AAA patients. This motivates further research to establish non-invasive imaging markers for vascular and perivascular inflammation in AAA.
Collapse
Affiliation(s)
- Daniel Ginzburg
- Department of Diagnostic and Interventional Radiology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Sebastian Nowak
- Department of Diagnostic and Interventional Radiology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Ulrike Attenberger
- Department of Diagnostic and Interventional Radiology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Julian Luetkens
- Department of Diagnostic and Interventional Radiology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Alois Martin Sprinkart
- Department of Diagnostic and Interventional Radiology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Daniel Kuetting
- Department of Diagnostic and Interventional Radiology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany.
| |
Collapse
|
9
|
Wang Y, Wang X, Chen Y, Zhang Y, Zhen X, Tao S, Dou J, Li P, Jiang G. Perivascular fat tissue and vascular aging: A sword and a shield. Pharmacol Res 2024; 203:107140. [PMID: 38513826 DOI: 10.1016/j.phrs.2024.107140] [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: 10/21/2023] [Revised: 02/16/2024] [Accepted: 03/11/2024] [Indexed: 03/23/2024]
Abstract
The understanding of the function of perivascular adipose tissue (PVAT) in vascular aging has significantly changed due to the increasing amount of information regarding its biology. Adipose tissue surrounding blood vessels is increasingly recognized as a key regulator of vascular disorders. It has significant endocrine and paracrine effects on the vasculature and is mediated by the production of a variety of bioactive chemicals. It also participates in a number of pathological regulatory processes, including oxidative stress, immunological inflammation, lipid metabolism, vasoconstriction, and dilation. Mechanisms of homeostasis and interactions between cells at the local level tightly regulate the function and secretory repertoire of PVAT, which can become dysregulated during vascular aging. The PVAT secretion group changes from being reducing inflammation and lowering cholesterol to increasing inflammation and increasing cholesterol in response to systemic or local inflammation and insulin resistance. In addition, the interaction between the PVAT and the vasculature is reciprocal, and the biological processes of PVAT are directly influenced by the pertinent indicators of vascular aging. The architectural and biological traits of PVAT, the molecular mechanism of crosstalk between PVAT and vascular aging, and the clinical correlation of vascular age-related disorders are all summarized in this review. In addition, this paper aims to elucidate and evaluate the potential benefits of therapeutically targeting PVAT in the context of mitigating vascular aging. Furthermore, it will discuss the latest advancements in technology used for targeting PVAT.
Collapse
Affiliation(s)
- Yan Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xianmin Wang
- Xinjiang Uygur Autonomous Region Hospital of Traditional Chinese Medicine, Xinjiang 830000, China
| | - Yang Chen
- School of Traditional Chinese Medicine, Xinjiang Medical University, Xinjiang 830011, China
| | - Yuelin Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xianjie Zhen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Siyu Tao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jinfang Dou
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Peng Li
- Xinjiang Uygur Autonomous Region Hospital of Traditional Chinese Medicine, Xinjiang 830000, China
| | - Guangjian Jiang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China; School of Traditional Chinese Medicine, Xinjiang Medical University, Xinjiang 830011, China.
| |
Collapse
|
10
|
Gao JP, Zhang HP, Wei R, Guo W. A Novel Method for the Rat Model of Abdominal Aortic Aneurysm Induced by Retroperitoneal Implantation of an Osmotic Pump System With Lipopolysaccharide. Ann Vasc Surg 2024; 101:41-52. [PMID: 38154490 DOI: 10.1016/j.avsg.2023.11.025] [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: 06/23/2023] [Revised: 09/17/2023] [Accepted: 11/17/2023] [Indexed: 12/30/2023]
Abstract
BACKGROUND Few methods can cocurrently mimic the pathological characteristics and nature history of human abdominal aortic aneurysms (AAAs), especially for the exist of the self-healing tendency of rodents. This study tested a novel method for the AAA rat model induced by retroperitoneal implantation of an osmotic pump system with lipopolysaccharide (LPS) based on the hypothesis that chronic inflammation of perivascular adipose tissue directly influenced the development and progression of AAAs. METHODS 20 male Sprague-Dawley rats (10-month-old) fed with the Paigen diet were randomly divided into 4 groups: the blank group ×2, the sham group ×4, the empty capsule group ×4, and the LPS capsule group ×10. The LPS capsule group received implantations of the ALZET® osmotic pump capsule with LPS (3.6 μg/day) parallel to the abdominal aorta through a retroperitoneal approach. Two weeks later, 6 rats were randomly selected from the LPS capsule group to form the anti-inflammatory group and received implantations of another osmotic pump capsule with interleukin (IL)-10 (75 ng/day) through the same approach. The changes in abdominal aortic diameter were observed by ultrasound every 2 weeks, and samples were harvested for histopathologic and immunohistochemical analysis 6 weeks later. RESULTS Within the 6 weeks after modeling, the LPS capsule group showed sustained and significant aortic dilatation (P < 0.01), while the anti-inflammatory group showed a rapid and obvious shrinkage 2 weeks after the IL-10 osmotic pump capsule implantation (P < 0.01). The LPS capsule group presented excellent pathological mimicking of human AAAs and showed severe medial degeneration with the least elastic content among the 5 groups at the end of the sixth week (P < 0.05). Notably, the anti-inflammatory group showed perfect medial preservation with the most elastic content (P < 0.05) and the highest elastin/collagen ratio (P < 0.01) at the end of the study. Matrix metalloproteinases (MMP) 2 and 9 and toll-like receptor 2 showed strong expression in the LPS capsule group at the end of the sixth week, which was significantly higher than that in the blank group and sham group. Interestingly, the anti-inflammatory group showed a slightly higher MMP9 expression than the LPS capsule group though there was no statistical difference between them. CONCLUSIONS This novel method for the rat AAA model induced by retroperitoneal implantation of an osmotic pump capsule with LPS can concurrently mimic the histological characteristics and natural history of human AAAs. Further studies were needed to improve the osmotic pump system.
Collapse
Affiliation(s)
- Jiang-Ping Gao
- Department of Vascular Surgery, Chinese PLA General Hospital, Beijing, China; Medical School of Chinese PLA, Beijing, China
| | - Hong-Peng Zhang
- Department of Vascular Surgery, Chinese PLA General Hospital, Beijing, China
| | - Ren Wei
- Department of Vascular Surgery, Chinese PLA General Hospital, Beijing, China
| | - Wei Guo
- Department of Vascular Surgery, Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
11
|
Wu Y, Zhang H, Jiang D, Yin F, Guo P, Zhang X, Zhang J, Han Y. Body mass index and the risk of abdominal aortic aneurysm presence and post-operative mortality: a systematic review and dose-response meta-analysis. Int J Surg 2024; 110:01279778-990000000-01023. [PMID: 38320094 PMCID: PMC11020033 DOI: 10.1097/js9.0000000000001125] [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: 11/14/2023] [Accepted: 01/09/2024] [Indexed: 02/08/2024]
Abstract
BACKGROUND The clinical data regarding the relationships between body mass index (BMI) and abdominal aortic aneurysm (AAA) are inconsistent, especially for the obese and overweight patients. The aims of this study were to determine whether obesity is associated with the presence of AAA and to investigate the quantitative relationship between BMI and the risk of AAA presence and post-operative mortality. MATERIALS AND METHODS PubMed, Web of Science and Embase databases were used to search for pertinent studies updated to December 2023. The pooled relative risk (RR) with 95% confidence interval (CI) was estimated by conventional meta-analysis based on random effects model. Dose-response meta-analyses using robust-error meta-regression (REMR) model were conducted to quantify the associations between BMI and AAA outcome variables. Subgroup analysis, sensitivity analysis and publication bias analysis were performed according to the characteristics of participants. RESULTS 18 studies were included in our study. The meta-analysis showed a higher prevalence of AAA with a RR of 1.07 in patients with obesity. The dose-response meta-analysis revealed a non-linear relationship between BMI and the risk of AAA presence. A "U" shape curve reflecting the correlation between BMI and the risk of post-operative mortality in AAA patients was also uncovered, suggesting the "safest" BMI interval [28.55, 31.05] with the minimal RR. CONCLUSIONS Obesity is positively but nonlinearly correlated with the increased risk of AAA presence. BMI is related to AAA post-operative mortality in a "U" shaped curve, with the lowest RR observed among patients suffering from overweight and obesity. These findings offer a preventive strategy for AAA morbidity and provide guidance for improving the prognosis in patients undergone AAA surgical repair.
Collapse
Affiliation(s)
- Yihao Wu
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin
| | - Hao Zhang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin
| | - Deying Jiang
- Department of Vascular Surgery, Central Hospital of Dalian University of Technology Dalian
| | - Fanxing Yin
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin
| | - Panpan Guo
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin
| | - Xiaoxu Zhang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin
| | - Jian Zhang
- Department of Vascular Surgery, The First Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Yanshuo Han
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin
- Department of Vascular Surgery, Central Hospital of Dalian University of Technology Dalian
| |
Collapse
|
12
|
Domagała D, Data K, Szyller H, Farzaneh M, Mozdziak P, Woźniak S, Zabel M, Dzięgiel P, Kempisty B. Cellular, Molecular and Clinical Aspects of Aortic Aneurysm-Vascular Physiology and Pathophysiology. Cells 2024; 13:274. [PMID: 38334666 PMCID: PMC10854611 DOI: 10.3390/cells13030274] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 01/27/2024] [Accepted: 01/30/2024] [Indexed: 02/10/2024] Open
Abstract
A disturbance of the structure of the aortic wall results in the formation of aortic aneurysm, which is characterized by a significant bulge on the vessel surface that may have consequences, such as distention and finally rupture. Abdominal aortic aneurysm (AAA) is a major pathological condition because it affects approximately 8% of elderly men and 1.5% of elderly women. The pathogenesis of AAA involves multiple interlocking mechanisms, including inflammation, immune cell activation, protein degradation and cellular malalignments. The expression of inflammatory factors, such as cytokines and chemokines, induce the infiltration of inflammatory cells into the wall of the aorta, including macrophages, natural killer cells (NK cells) and T and B lymphocytes. Protein degradation occurs with a high expression not only of matrix metalloproteinases (MMPs) but also of neutrophil gelatinase-associated lipocalin (NGAL), interferon gamma (IFN-γ) and chymases. The loss of extracellular matrix (ECM) due to cell apoptosis and phenotype switching reduces tissue density and may contribute to AAA. It is important to consider the key mechanisms of initiating and promoting AAA to achieve better preventative and therapeutic outcomes.
Collapse
Affiliation(s)
- Dominika Domagała
- Division of Anatomy, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland; (D.D.); (K.D.); (H.S.); (S.W.)
| | - Krzysztof Data
- Division of Anatomy, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland; (D.D.); (K.D.); (H.S.); (S.W.)
| | - Hubert Szyller
- Division of Anatomy, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland; (D.D.); (K.D.); (H.S.); (S.W.)
| | - Maryam Farzaneh
- Fertility, Infertility and Perinatology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran;
| | - Paul Mozdziak
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, NC 27607, USA;
- Physiology Graduate Faculty, North Carolina State University, Raleigh, NC 27613, USA
| | - Sławomir Woźniak
- Division of Anatomy, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland; (D.D.); (K.D.); (H.S.); (S.W.)
| | - Maciej Zabel
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland; (M.Z.); (P.D.)
- Division of Anatomy and Histology, University of Zielona Góra, 65-046 Zielona Góra, Poland
| | - Piotr Dzięgiel
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland; (M.Z.); (P.D.)
- Department of Physiotherapy, University School of Physical Education, 51-612 Wroclaw, Poland
| | - Bartosz Kempisty
- Division of Anatomy, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland; (D.D.); (K.D.); (H.S.); (S.W.)
- Physiology Graduate Faculty, North Carolina State University, Raleigh, NC 27613, USA
- Institute of Veterinary Medicine, Nicolaus Copernicus University, 87-100 Torun, Poland
- Department of Obstetrics and Gynecology, University Hospital and Masaryk University, 602 00 Brno, Czech Republic
| |
Collapse
|
13
|
Zhang K, Zhang J, Kan C, Tian H, Ma Y, Huang N, Han F, Hou N, Sun X. Role of dysfunctional peri-organ adipose tissue in metabolic disease. Biochimie 2023; 212:12-20. [PMID: 37019205 DOI: 10.1016/j.biochi.2023.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 03/21/2023] [Accepted: 03/30/2023] [Indexed: 04/07/2023]
Abstract
Metabolic disease is a complex disorder defined by a group with interrelated factors. There is growing evidence that obesity can lead to a variety of metabolic diseases, including diabetes and cardiovascular disease. Excessive adipose tissue (AT) deposition and ectopic accumulation can lead to increased peri-organ AT thickness. Dysregulation of peri-organ (perivascular, perirenal, and epicardial) AT is strongly associated with metabolic disease and its complications. The mechanisms include secretion of cytokines, activation of immunocytes, infiltration of inflammatory cells, involvement of stromal cells, and abnormal miRNA expression. This review discusses the associations and mechanisms by which various types of peri-organ AT affect metabolic diseases while addressing it as a potential future treatment strategy.
Collapse
Affiliation(s)
- Kexin Zhang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Jingwen Zhang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Chengxia Kan
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Hongzhan Tian
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Yanhui Ma
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China; Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Na Huang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Fang Han
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China; Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Ningning Hou
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China.
| | - Xiaodong Sun
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China.
| |
Collapse
|
14
|
Huang CL, Huang YN, Yao L, Li JP, Zhang ZH, Huang ZQ, Chen SX, Zhang YL, Wang JF, Chen YX, Liu ZY. Thoracic perivascular adipose tissue inhibits VSMC apoptosis and aortic aneurysm formation in mice via the secretome of browning adipocytes. Acta Pharmacol Sin 2023; 44:345-355. [PMID: 35945313 PMCID: PMC9889802 DOI: 10.1038/s41401-022-00959-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 07/12/2022] [Indexed: 02/04/2023]
Abstract
Abdominal aortic aneurysm (AAA) is a dangerous vascular disease without any effective drug therapies so far. Emerging evidence suggests the phenotypic differences in perivascular adipose tissue (PVAT) between regions of the aorta are implicated in the development of atherosclerosis evidenced by the abdominal aorta more vulnerable to atherosclerosis than the thoracic aorta in large animals and humans. The prevalence of thoracic aortic aneurysms (TAA) is much less than that of abdominal aortic aneurysms (AAA). In this study we investigated the effect of thoracic PVAT (T-PVAT) transplantation on aortic aneurysm formation and the impact of T-PVAT on vascular smooth muscle cells. Calcium phosphate-induced mouse AAA model was established. T-PVAT (20 mg) was implanted around the abdominal aorta of recipient mice after removal of endogenous abdominal PVAT (A-PVAT) and calcium phosphate treatment. Mice were sacrificed two weeks after the surgery and the maximum external diameter of infrarenal aorta was measured. We found that T-PVAT displayed a more BAT-like phenotype than A-PVAT; transplantation of T-PVAT significantly attenuated calcium phosphate-induced abdominal aortic dilation and elastic degradation as compared to sham control or A-PVAT transplantation. In addition, T-PVAT transplantation largely preserved smooth muscle cell content in the abdominal aortic wall. Co-culture of T-PVAT with vascular smooth muscle cells (VSMCs) significantly inhibited H2O2- or TNFα plus cycloheximide-induced VSMC apoptosis. RNA sequencing analysis showed that T-PVAT was enriched by browning adipocytes and anti-apoptotic secretory proteins. We further verified that the secretome of mature adipocytes isolated from T-PVAT significantly inhibited H2O2- or TNFα plus cycloheximide-induced VSMC apoptosis. Using proteomic and bioinformatic analyses we identified cartilage oligomeric matrix protein (COMP) as a secreted protein significantly increased in T-PVAT. Recombinant COMP protein significantly inhibited VSMC apoptosis. We conclude that T-PVAT exerts anti-apoptosis effect on VSMCs and attenuates AAA formation, which is possibly attributed to the secretome of browning adipocytes.
Collapse
Affiliation(s)
- Chun-Ling Huang
- Medical Research Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Yu-Na Huang
- Department of Cardiology, Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Lei Yao
- Medical Research Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Jun-Ping Li
- Department of Cardiology, Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Zeng-Hui Zhang
- Department of Cardiology, Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Zhao-Qi Huang
- Department of Cardiology, Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Si-Xu Chen
- Department of Cardiology, Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Yu-Ling Zhang
- Department of Cardiology, Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Jing-Feng Wang
- Department of Cardiology, Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
| | - Yang-Xin Chen
- Department of Cardiology, Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
| | - Zhao-Yu Liu
- Medical Research Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
| |
Collapse
|
15
|
Hu J, Xue S, Xu Z, Wu Z, Xu X, Wang X, Liu G, Lu X, Li B, Liu X. Identification of core cuprotosis-correlated biomarkers in abdominal aortic aneurysm immune microenvironment based on bioinformatics. Front Immunol 2023; 14:1138126. [PMID: 37138870 PMCID: PMC10150024 DOI: 10.3389/fimmu.2023.1138126] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 03/30/2023] [Indexed: 05/05/2023] Open
Abstract
Background The occurrence of abdominal aortic aneurysms (AAAs) is related to the disorder of immune microenvironment. Cuprotosis was reported to influence the immune microenvironment. The objective of this study is to identify cuprotosis-related genes involved in the pathogenesis and progression of AAA. Methods Differentially expressed lncRNAs (DElncRNAs) and mRNAs (DEmRNAs) in mouse were identified following AAA through high-throughput RNA sequencing. The enrichment analyses of pathway were selected through Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG). The validation of cuprotosis-related genes was conducted through immunofluorescence and western blot analyses. Results Totally, 27616 lncRNAs and 2189 mRNAs were observed to be differentially expressed (|Fold Change| ≥ 2 and q< 0.05) after AAA, including 10424 up-regulated and 17192 down-regulated lncRNAs, 1904 up-regulated and 285 down-regulated mRNAs. Gene ontology and KEGG pathway analysis showed that the DElncRNAs and DEmRNAs were implicated in many different biological processes and pathways. Furthermore, Cuprotosis-related genes (NLRP3, FDX1) were upregulated in the AAA samples compared with the normal one. Conclusion Cuprotosis-related genes (NLRP3,FDX1) involved in AAA immune environment might be critical for providing new insight into identification of potential targets for AAA therapy.
Collapse
Affiliation(s)
- Jiateng Hu
- Department of Vascular Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Vascular Centre of Shanghai Jiao Tong University, Shanghai, China
| | - Song Xue
- Department of Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhijue Xu
- Department of Vascular Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Vascular Centre of Shanghai Jiao Tong University, Shanghai, China
| | - Zhaoyu Wu
- Department of Vascular Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Vascular Centre of Shanghai Jiao Tong University, Shanghai, China
| | - Xintong Xu
- Department of Vascular Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Vascular Centre of Shanghai Jiao Tong University, Shanghai, China
| | - Xin Wang
- Department of Vascular Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Vascular Centre of Shanghai Jiao Tong University, Shanghai, China
| | - Guang Liu
- Department of Vascular Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Vascular Centre of Shanghai Jiao Tong University, Shanghai, China
| | - Xinwu Lu
- Department of Vascular Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Vascular Centre of Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Xinwu Lu, ; Bo Li, ; Xiaobing Liu,
| | - Bo Li
- Department of Vascular Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Vascular Centre of Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Xinwu Lu, ; Bo Li, ; Xiaobing Liu,
| | - Xiaobing Liu
- Department of Vascular Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Vascular Centre of Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Xinwu Lu, ; Bo Li, ; Xiaobing Liu,
| |
Collapse
|
16
|
Jadli AS, Ballasy NN, Gomes KP, Mackay CDA, Meechem M, Wijesuriya TM, Belke D, Thompson J, Fedak PWM, Patel VB. Attenuation of Smooth Muscle Cell Phenotypic Switching by Angiotensin 1-7 Protects against Thoracic Aortic Aneurysm. Int J Mol Sci 2022; 23:ijms232415566. [PMID: 36555207 PMCID: PMC9779869 DOI: 10.3390/ijms232415566] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 11/30/2022] [Accepted: 12/02/2022] [Indexed: 12/13/2022] Open
Abstract
Thoracic aortic aneurysm (TAA) involves extracellular matrix (ECM) remodeling of the aortic wall, leading to reduced biomechanical support with risk of aortic dissection and rupture. Activation of the renin-angiotensin system, and resultant angiotensin (Ang) II synthesis, is critically involved in the onset and progression of TAA. The current study investigated the effects of angiotensin (Ang) 1-7 on a murine model of TAA. Male 8-10-week-old ApoEKO mice were infused with Ang II (1.44 mg/kg/day) and treated with Ang 1-7 (0.576 mg/kg/day). ApoEKO mice developed advanced TAA in response to four weeks of Ang II infusion. Echocardiographic and histological analyses demonstrated increased aortic dilatation, excessive structural remodelling, perivascular fibrosis, and inflammation in the thoracic aorta. Ang 1-7 infusion led to attenuation of pathological phenotypic alterations associated with Ang II-induced TAA. Smooth muscle cells (SMCs) isolated from adult murine thoracic aorta exhibited excessive mitochondrial fission, oxidative stress, and hyperproliferation in response to Ang II. Treatment with Ang 1-7 resulted in inhibition of mitochondrial fragmentation, ROS generation, and hyperproliferation. Gene expression profiling used for characterization of the contractile and synthetic phenotypes of thoracic aortic SMCs revealed preservation of the contractile phenotype with Ang 1-7 treatment. In conclusion, Ang 1-7 prevented Ang II-induced vascular remodeling and the development of TAA. Enhancing Ang 1-7 actions may provide a novel therapeutic strategy to prevent or delay the progression of TAA.
Collapse
Affiliation(s)
- Anshul S. Jadli
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Libin Cardiovascular Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Noura N. Ballasy
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Libin Cardiovascular Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Karina P. Gomes
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Libin Cardiovascular Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Cameron D. A. Mackay
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Libin Cardiovascular Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Megan Meechem
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Libin Cardiovascular Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Tishani Methsala Wijesuriya
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Libin Cardiovascular Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Darrell Belke
- Libin Cardiovascular Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
- Section of Cardiac Surgery, Department of Cardiac Sciences, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Jennifer Thompson
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Libin Cardiovascular Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
- Alberta Children’s Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Paul W. M. Fedak
- Libin Cardiovascular Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
- Section of Cardiac Surgery, Department of Cardiac Sciences, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Vaibhav B. Patel
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Libin Cardiovascular Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
- Correspondence: or ; Tel.: +1-(403)-220-3446
| |
Collapse
|
17
|
FOS gene associated immune infiltration signature in perivascular adipose tissues of abdominal aortic aneurysm. Gene X 2022; 831:146576. [PMID: 35568340 DOI: 10.1016/j.gene.2022.146576] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 04/19/2022] [Accepted: 05/09/2022] [Indexed: 11/23/2022] Open
Abstract
Abdominal aortic aneurysms (AAA) are pathological dilations in local aortic wall. The inflammatory infiltrates of the perivascular adipose tissue (PAT) surrounding AAAs were associated with AAAs and have been shown to contribute vascular pathology. However, the mechanism by which PAT inflammation contributes to vascular pathology in AAA remains to be clarified. This study aimed to explore the association between immune cell infiltration and key gene expression profile in PAT of AAA. For that, a gene expression dataset of human dilated perivascular adipose tissue (dPAT), non-dilated perivascular adipose tissue (ndPAT), subcutaneous abdominal fat (SAF) and omental-visceral fat (OVF) samples, as well as another microarray dataset of the abdominal perivascular adipose tissue in peripheral artery disease patients were downloaded from GEO database for analysis in this study. The CIBERSORT algorithm, weighted gene co-expression network analysis (WGCNA) and LASSO algorithm were used for the identification of immune infiltration, immune-related genes and the development of diagnostic signature. Our data discovered a significant higher proportion of activated mast cells and follicular helper T (Tfh) cells in dPAT than ndPAT, OVT and SAF samples. Moreover, AP-1 family members (FOS, FOSB, ATF3, JUN and JUNB) were found to compose the hub genes of purple module in WGCNA. Among them, FOS gene acts as a higher efficient marker to discriminate dPAT from ndPAT, OVT and SAF in AAA. Meanwhile, the expression profiles of the AP-1 family members are all significantly positive correlated with activated mast cell, plasma cell and Tfh cell infiltration in dPAT of AAA. Therefore, in the PAT surrounding AAA, the signature of inflammatory infiltration might be represented by a FOS-dominated cell network consist of activated mast cell, plasma cell and Tfh cell. Given the complicated etiology of AAA, our results are likely to shed new light on the pathophysiologic mechanism of AAA influenced by the local dPAT.
Collapse
|
18
|
Dias-Neto M, Meekel JP, Oderich G, Yeung KK. Assessment of periadventitial adipose tissue in abdominal aortic aneurysm with computed tomography and its challenges : Invited commentary. THE INTERNATIONAL JOURNAL OF CARDIOVASCULAR IMAGING 2022; 38:1635-1637. [PMID: 38819549 DOI: 10.1007/s10554-022-02580-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 02/23/2022] [Indexed: 11/29/2022]
Affiliation(s)
- Marina Dias-Neto
- Department of Cardiothoracic and Vascular Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States.
| | - Jorn P Meekel
- Department of Vascular Surgery, Amsterdam University Medical Centres location VUmc, Amsterdam, the Netherlands
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centres, Amsterdam, the Netherlands
- Department of Vascular Surgery, Amsterdam University Medical Centres, location AMC, Amsterdam, the Netherlands
- Department of Surgery, Zaans Medisch Centrum, Zaandam, The Netherlands
| | - Gustavo Oderich
- Department of Cardiothoracic and Vascular Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Kak K Yeung
- Department of Vascular Surgery, Amsterdam University Medical Centres location VUmc, Amsterdam, the Netherlands
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centres, Amsterdam, the Netherlands
- Department of Vascular Surgery, Amsterdam University Medical Centres, location AMC, Amsterdam, the Netherlands
| |
Collapse
|
19
|
Exploring the Effect and Mechanism of Si-Miao-Yong-An Decoction on Abdominal Aortic Aneurysm Based on Mice Experiment and Bioinformatics Analysis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:4766987. [PMID: 35685724 PMCID: PMC9173986 DOI: 10.1155/2022/4766987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 04/26/2022] [Accepted: 05/19/2022] [Indexed: 12/02/2022]
Abstract
Background Abdominal aortic aneurysm (AAA) is a fatal disease characterized by high morbidity and mortality in old population. Globally, effective drugs for AAA are still limited. Si-Miao-Yong-An decoction (SMYAD), a traditional Chinese medicine (TCM) formula with a high medical value, was reported to be successfully used in an old AAA patient. Thus, we reason that SMYAD may serve as a potential anti-AAA regime. Objective The exact effects and detailed mechanisms of SMYAD on AAA were explored by using the experimental study and bioinformatics analysis. Methods Firstly, C57BL/6N mice induced by Bap and Ang II were utilized to reproduce the AAA model, and the effects of SMYAD were systematically assessed according to histology, immunohistochemistry, and enzyme-linked immunosorbent assay (ELISA). Then, network pharmacology was applied to identify the biological processes, pathways, and hub targets of SMYAD against AAA; moreover, molecular docking was utilized to identify the binding ability and action targets. Results In an animal experiment, SMYAD was found to effectively alleviate the degree of pathological expansion of abdominal aorta and reduce the incidence of Bap/Ang II-induced AAA, along with reducing the damage to elastic lamella, attenuating infiltration of macrophage, and lowering the circulating IL-6 level corresponding to the animal study, and network pharmacology revealed the detailed mechanisms of SMYAD on AAA that were related to pathways of inflammatory response, defense response, apoptotic, cell migration and adhesion, and reactive oxygen species metabolic process. Then, seven targets, IL-6, TNF, HSP90AA1, RELA, PTGS2, ESR1, and MMP9, were identified as hub targets of SMYAD against AAA. Furthermore, molecular docking verification revealed that the active compounds of SMYAD had good binding ability and clear binding site with core targets related to AAA formation. Conclusion SMYAD can suppress AAA development through multicompound, multitarget, and multipathway, which provides a research direction for further study.
Collapse
|
20
|
Hu G, Ding N, Wang Z, Jin Z. The association of body composition with abdominal aortic aneurysm growth after endovascular aneurysm repair. Insights Imaging 2022; 13:76. [PMID: 35467156 PMCID: PMC9038972 DOI: 10.1186/s13244-022-01187-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 02/19/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Body composition (BC) may be associated with abdominal aortic aneurysm (AAA) growth, but the results of previous research are contradictory. This study aimed to explore the relationship between BC and postoperative aneurysm progression.
Methods
Patients with regular postoperative follow-ups were retrospectively identified. The volume change of the aneurysm was measured to evaluate AAA progression. After segmenting different body components (subcutaneous fat, visceral fat, pure muscle, and intramuscular fat), the shape features and gray features of these tissues were extracted. Uni- and multivariable methods were used to analyze the relationship between imaging features of BC and AAA growth.
Results
A total of 94 patients (68 ± 8 years) were eligible for feature analyses. Patients with expansive aneurysms (29/94; volume change > 2%) were classified into Group(+) and others with stable or shrunken aneurysms (65/94) were classified into Group(−). Compared with Group(+), Group(−) showed a higher volume percent of pure muscle (21.85% vs 19.51%; p = .042) and a lower value of intramuscular fat (1.23% vs 1.65%; p = .025). CT attenuation of muscle tissues of Group(−) got a higher mean value (31.16 HU vs 23.92 HU; p = .019) and a lower standard deviation (36.12 vs 38.82; p = .006) than Group(+). For adipose tissue, we found no evidence of a difference between the two groups. The logistic regression model containing muscle imaging features showed better discriminative accuracy than traditional factors (84% vs 73%).
Conclusions
Muscle imaging features are associated with the volume change of postoperative aneurysms and can make an early prediction. Adipose tissue is not specifically related to AAA growth.
Collapse
|
21
|
Chen Y, Qin Z, Wang Y, Li X, Zheng Y, Liu Y. Role of Inflammation in Vascular Disease-Related Perivascular Adipose Tissue Dysfunction. Front Endocrinol (Lausanne) 2021; 12:710842. [PMID: 34456867 PMCID: PMC8385491 DOI: 10.3389/fendo.2021.710842] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 07/21/2021] [Indexed: 12/12/2022] Open
Abstract
Perivascular adipose tissue (PVAT) is the connective tissue around most blood vessels throughout the body. It provides mechanical support and maintains vascular homeostasis in a paracrine/endocrine manner. Under physiological conditions, PVAT has anti-inflammatory effects, improves free fatty acid metabolism, and regulates vasodilation. In pathological conditions, PVAT is dysfunctional, secretes many anti-vasodilator factors, and participates in vascular inflammation through various cells and mediators; thus, it causes dysfunction involving vascular smooth muscle cells and endothelial cells. Inflammation is an important pathophysiological event in many vascular diseases, such as vascular aging, atherosclerosis, and hypertension. Therefore, the pro-inflammatory crosstalk between PVAT and blood vessels may comprise a novel therapeutic target for the prevention and treatment of vascular diseases. In this review, we summarize findings concerning PVAT function and inflammation in different pathophysiological backgrounds, focusing on the secretory functions of PVAT and the crosstalk between PVAT and vascular inflammation in terms of vascular aging, atherosclerosis, hypertension, diabetes mellitus, and other diseases. We also discuss anti-inflammatory treatment for potential vascular diseases involving PVAT.
Collapse
Affiliation(s)
- Yaozhi Chen
- Center for Cardiovascular Medicine, First Hospital of Jilin University, Changchun, China
| | - Zeyu Qin
- Department of Respiratory Medicine, First Hospital of Jilin University, Changchun, China
| | - Yaqiong Wang
- Department of Endocrinology and Metabolism, First Hospital of Jilin University, Changchun, China
| | - Xin Li
- Center for Cardiovascular Medicine, First Hospital of Jilin University, Changchun, China
| | - Yang Zheng
- Center for Cardiovascular Medicine, First Hospital of Jilin University, Changchun, China
- *Correspondence: Yunxia Liu, ; Yang Zheng,
| | - Yunxia Liu
- Center for Cardiovascular Medicine, First Hospital of Jilin University, Changchun, China
- *Correspondence: Yunxia Liu, ; Yang Zheng,
| |
Collapse
|
22
|
Tedjawirja VN, Nieuwdorp M, Yeung KK, Balm R, de Waard V. A Novel Hypothesis: A Role for Follicle Stimulating Hormone in Abdominal Aortic Aneurysm Development in Postmenopausal Women. Front Endocrinol (Lausanne) 2021; 12:726107. [PMID: 34721292 PMCID: PMC8548664 DOI: 10.3389/fendo.2021.726107] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 09/02/2021] [Indexed: 12/24/2022] Open
Abstract
An abdominal aortic aneurysm (AAA) is a dilatation of the abdominal aorta, which can potentially be fatal due to exsanguination following rupture. Although AAA is less prevalent in women, women with AAA have a more severe AAA progression compared to men as reflected by enhanced aneurysm growth rates and a higher rupture risk. Women are diagnosed with AAA at an older age than men, and in line with increased osteoporosis and cardiovascular events, the delayed AAA onset has been attributed to the reduction of the protective effect of oestrogens during the menopausal transition. However, new insights have shown that a high follicle stimulating hormone (FSH) level during menopause may also play a key role in those diseases. In this report we hypothesize that FSH may aggravate AAA development and progression in postmenopausal women via a direct and/or indirect role, promoting aorta pathology. Since FSH receptors (FSHR) are reported on many other cell types than granulosa cells in the ovaries, it is feasible that FSH stimulation of FSHR-bearing cells such as aortic endothelial cells or inflammatory cells, could promote AAA formation directly. Indirectly, AAA progression may be influenced by an FSH-mediated increase in osteoporosis, which is associated with aortic calcification. Also, an FSH-mediated decrease in cholesterol uptake by the liver and an increase in cholesterol biosynthesis will increase the cholesterol level in the circulation, and subsequently promote aortic atherosclerosis and inflammation. Lastly, FSH-induced adipogenesis may lead to obesity-mediated dysfunction of the microvasculature of the aorta and/or modulation of the periaortic adipose tissue. Thus the long term increased plasma FSH levels during the menopausal transition may contribute to enhanced AAA disease in menopausal women and could be a potential novel target for treatment to lower AAA-related events in women.
Collapse
Affiliation(s)
- Victoria N. Tedjawirja
- Department of Surgery, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
- *Correspondence: Victoria N. Tedjawirja,
| | - Max Nieuwdorp
- Departments of Internal and Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Kak Khee Yeung
- Department of Surgery, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
| | - Ron Balm
- Department of Surgery, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
| | - Vivian de Waard
- Department of Medical Biochemistry, Amsterdam UMC, University of Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
| |
Collapse
|