1
|
Wei P, Li K, Li M, Liu H, Xia C, Chen Y, Zhao S, Li Y. The C57BL/6N mouse substrain is a viable model of elastase-induced abdominal aortic aneurysm. Front Cardiovasc Med 2024; 11:1462032. [PMID: 39399516 PMCID: PMC11466807 DOI: 10.3389/fcvm.2024.1462032] [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: 07/23/2024] [Accepted: 09/19/2024] [Indexed: 10/15/2024] Open
Abstract
Aim Compared with the C57BL/6N substrain, the C57BL/6J substrain is more susceptible to the angiotensin II (Ang II)-induced development of dissected abdominal aortic aneurysms (AAAs). The aim of this study was to elucidate whether the widely used C57BL/6N mouse substrain is as susceptible as the C57BL/6J mouse substrain to porcine pancreatic elastase (PPE) infusion-induced experimental nondissected AAA development. Methods Experimental nondissected AAAs were induced in C57BL/6J and C57BL/6N mice via transient aortic luminal infusion of PPE. On Day 0 (baseline) and Day 14 after PPE infusion, the abdominal aortic diameter was directly measured. Aortic aneurysmal segment samples were collected, and histopathological analysis was performed. Results On Day 14 after PPE infusion, aortic diameters were significantly increased in both mouse substrains (from approximately 0.51 to 1.24 mm in C57BL/6J mice and from 0.51 to 1.18 mm in C57BL/6N mice). The increase in diameter of all the mice exceeded 50% and met the criteria for AAA model establishment (143% and 135% in C57BL/6J mice and C57BL/6N mice, respectively). PPE infusion also induced obvious local aortic wall macrophage and T-cell infiltration, elastin degradation, smooth muscle cell depletion and high metallopeptidase (MMP)-2 and MMP-9 expression levels in C57BL/6N mice, but these differences were not significant compared with those in C57BL/6J mice. However, PPE infusion led to the recruitment of more B cells and the sprouting of more neovessels at the aneurysmal lesion site in C57BL/6J mice than in C57BL/6N mice. Conclusion The C57BL/6N mouse substrain is suitable for establishing a model of AAA via elastase infusion.
Collapse
Affiliation(s)
- Panpan Wei
- Department of Vascular Surgery, The Second Hospital of Tianjin Medical University, Tianjin, China
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Kexin Li
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Meng Li
- Department of Vascular Surgery, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Haole Liu
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Congcong Xia
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Yajie Chen
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, Guangdong, China
| | - Sihai Zhao
- Department of Vascular Surgery, The Second Hospital of Tianjin Medical University, Tianjin, China
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Yankui Li
- Department of Vascular Surgery, The Second Hospital of Tianjin Medical University, Tianjin, China
| |
Collapse
|
2
|
Cui Y, Tan C, Zhang W, Jiang P, Sun J, Mei F. Establishment of Mouse Models of Abdominal Aortic Aneurysm. Angiology 2024:33197241284848. [PMID: 39268808 DOI: 10.1177/00033197241284848] [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: 09/15/2024]
Abstract
Abdominal aortic aneurysm (AAA) is a chronic vascular disease that commonly affects elderly individuals but has recently increased in younger populations. As the aneurysm grows, it can cause compression symptoms such as abdominal pain, rupture, and bleeding, which are absent in the early stages. Once an AAA ruptures and causes bleeding, the mortality rate is alarmingly high. Currently, the pathogenesis for AAA is unknown, and therapeutic options are limited, necessitating improvement in treatment efficacy. An essential research method for studying the processes and potential treatment of AAA is establishing animal models using mice. The present study provides a detailed overview of the widely used AAA mouse animal models and their construction strategies, advantages, disadvantages, scope of applications, and prospects.
Collapse
Affiliation(s)
- Yongpan Cui
- Department of Vascular Surgery, Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, Hubei, China
| | - Chengpeng Tan
- Department of Vascular Surgery, Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, Hubei, China
| | - Wuming Zhang
- Department of Vascular Surgery, Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, Hubei, China
| | - Peng Jiang
- Department of Vascular Surgery, Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, Hubei, China
| | - Jianfeng Sun
- Department of Vascular Surgery, Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, Hubei, China
| | - Fei Mei
- Department of Vascular Surgery, Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, Hubei, China
| |
Collapse
|
3
|
Zhao J, Xiong Y, Liu Y, Ling J, Liu S, Wang W. Endothelium Piezo1 deletion alleviates experimental varicose veins by attenuating perivenous inflammation. Mol Cell Biochem 2024:10.1007/s11010-024-05115-9. [PMID: 39249600 DOI: 10.1007/s11010-024-05115-9] [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: 03/28/2024] [Accepted: 08/31/2024] [Indexed: 09/10/2024]
Abstract
Previous large-scale genetic studies have prioritized the causal genes piezo type mechanosensitive ion channel component 1 (PIEZO1) and castor zinc finger 1 (CASZ1) associated with varicose veins (VVs). This study aims to evaluate their roles in both clinical and experimental VVs. In this study, we investigated abundance of PIEZO1 and CASZ1 in both varicose and normal veins from the same patients. Yoda1 (a selective PIEZO1 agonist, 2.6 mg/kg/day) or vehicle was administered intraperitoneally for 3 weeks to evaluate the effect of PIEZO1 activation on experimental VVs. Subsequently, endothelial Piezo1 deletion mice (Piezo1iΔEC mice) were generated to explored the role of endothelial PIEZO1 on VVs. Laser speckle imaging, flow cytometry, cell tracing with Evans blue or rhodamine-6G, and histopathological staining were utilized to evaluate the pathophysiology of VVs. Our results showed that mRNA expression of PIEZO1, but not CASZ1, was abundant and increased in clinical VVs. The Piezo1tP1-td mice revealed endothelium-specific expression of PIEZO1 in mice veins. By establishing iliac vein ligation-induced VVs in mice, Yoda1 exacerbated experimental VVs with increased inflammatory cell infiltration. Subsequently, endothelial Piezo1 deletion (Piezo1iΔEC mice) alleviated experimental VVs and vascular remodeling by directly reducing vascular permeability and leukocyte-endothelium interactions compared to the control (Piezo1fl/fl mice). PIEZO1 is highly expressed in clinical VVs, meanwhile, activation or inhibition of PIEZO1 exerts a remarkable effect on experimental VVs. Furthermore, Piezo1 may constitute a potential therapeutic approach for the medical treatment of VVs.
Collapse
Affiliation(s)
- Jiani Zhao
- Department of General and Vascular Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yacheng Xiong
- Department of General and Vascular Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yu Liu
- Department of General and Vascular Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Jin Ling
- Department of General and Vascular Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Shuai Liu
- Department of General and Vascular Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China.
| | - Wei Wang
- Department of General and Vascular Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
4
|
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
|
5
|
Zhu J, Meganathan I, MacAruthur R, Kassiri Z. Inflammation in Abdominal Aortic Aneurysm: Cause or Comorbidity? Can J Cardiol 2024:S0828-282X(24)00926-7. [PMID: 39181326 DOI: 10.1016/j.cjca.2024.08.274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/12/2024] [Accepted: 08/15/2024] [Indexed: 08/27/2024] Open
Abstract
Aortic aneurysm is a potentially deadly disease. It is chronic degeneration of the aortic wall that involves an inflammatory response and the immune system, aberrant remodelling of the extracellular matrix, and maladaptive transformation of the aortic cells. This review article focuses on the role of the inflammatory cells in abdominal aortic aneurysm. Studies in human aneurysmal specimens and animal models have identified various inflammatory cell types that could contribute to formation or expansion of aneurysms. These include the commonly studied leukocytes (neutrophils and macrophages) as well as the less commonly explored natural killer cells, dendritic cells, T cells, and B cells. Despite the well-demonstrated contribution of inflammatory cells and the related signalling pathways to development and expansion of aneurysms, anti-inflammatory therapy approaches have demonstrated limitations and may require additional considerations such as a combinational approach in targeting multiple pathways for significant beneficial outcomes.
Collapse
Affiliation(s)
- Jiechun Zhu
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Ilamaran Meganathan
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Roderick MacAruthur
- Department of Cardiac Surgery, Mazankowski Alberta Heart Institute, University of Alberta Hospital, Edmonton, Alberta, Canada
| | - Zamaneh Kassiri
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
6
|
Wu Z, Zhang P, Yue J, Wang Q, Zhuang P, Jehan S, Fan L, Xue J, Zhou W, Wang H. Tea polyphenol nanoparticles enable targeted siRNA delivery and multi-bioactive therapy for abdominal aortic aneurysms. J Nanobiotechnology 2024; 22:471. [PMID: 39118143 PMCID: PMC11308685 DOI: 10.1186/s12951-024-02756-2] [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: 03/31/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024] Open
Abstract
Abdominal aortic aneurysm (AAA) is a life-threatening vascular disease, while there is a lack of pharmaceutical interventions to halt AAA progression presently. To address the multifaceted pathology of AAA, this work develops a novel multifunctional gene delivery system to simultaneously deliver two siRNAs targeting MMP-2 and MMP-9. The system (TPNs-siRNA), formed through the oxidative polymerization and self-assembly of epigallocatechin gallate (EGCG), efficiently encapsulates siRNAs during self-assembly. TPNs-siRNA safeguards siRNAs from biological degradation, facilitates intracellular siRNA transfection, promotes lysosomal escape, and releases siRNAs to silence MMP-2 and MMP-9. Additionally, TPNs, serving as a multi-bioactive material, mitigates oxidative stress and inflammation, fosters M1-to-M2 repolarization of macrophages, and inhibits cell calcification and apoptosis. In experiments with AAA mice, TPNs-siRNA accumulated and persisted in aneurysmal tissue after intravenous delivery, demonstrating that TPNs-siRNA can be significantly distributed in macrophages and VSMCs relevant to AAA pathogenesis. Leveraging the carrier's intrinsic multi-bioactive properties, the targeted siRNA delivery by TPNs exhibits a synergistic effect for enhanced AAA therapy. Furthermore, TPNs-siRNA is gradually metabolized and excreted from the body, resulting in excellent biocompatibility. Consequently, TPNs emerges as a promising multi-bioactive nanotherapy and a targeted delivery nanocarrier for effective AAA therapy.
Collapse
Affiliation(s)
- Zhen Wu
- Department of Vascular and Interventional Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
| | - Peng Zhang
- Department of Vascular Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Jie Yue
- Department of Vascular Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Qingshan Wang
- Department of Vascular Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Peipei Zhuang
- Department of Vascular Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Shah Jehan
- Department of Vascular Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Liyuan Fan
- Department of Vascular Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Jiarun Xue
- Department of Vascular Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Wenhu Zhou
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China
| | - Haiyang Wang
- Department of Vascular and Interventional Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China.
| |
Collapse
|
7
|
Zhu X, Yu G, Lv Y, Yang N, Zhao Y, Li F, Zhao J, Chen Z, Lai Y, Chen L, Wang X, Xiao J, Cai Y, Feng Y, Ding J, Gao W, Zhou K, Xu H. Neuregulin-1, a member of the epidermal growth factor family, mitigates STING-mediated pyroptosis and necroptosis in ischaemic flaps. BURNS & TRAUMA 2024; 12:tkae035. [PMID: 38855574 PMCID: PMC11162832 DOI: 10.1093/burnst/tkae035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 05/22/2024] [Accepted: 05/22/2024] [Indexed: 06/11/2024]
Abstract
Background Ensuring the survival of the distal end of a random flap during hypoperfusion (ischaemia) is difficult in clinical practice. Effective prevention of programmed cell death is a potential strategy for inhibiting ischaemic flap necrosis. The activation of stimulator of interferon genes (STING) pathway promotes inflammation and leads to cell death. The epidermal growth factor family member neuregulin-1 (NRG1) reduces cell death by activating the protein kinase B (AKT) signalling pathway. Moreover, AKT signalling negatively regulates STING activity. We aimed to verify the efficacy of NRG1 injection in protecting against flap necrosis. Additionally, we investigated whether NRG1 effectively enhances ischemic flap survival by inhibiting pyroptosis and necroptosis through STING suppression. Methods A random-pattern skin flap model was generated on the backs of C57BL/6 mice. The skin flap survival area was determined. The blood supply and vascular network of the flap was assessed by laser Doppler blood flow analysis. Cluster of differentiation 34 immunohistochemistry (IHC) and haematoxylin and eosin (H&E) staining of the flap sections revealed microvessels. Transcriptome sequencing analysis revealed the mechanism by which NRG1 promotes the survival of ischaemic flaps. The levels of angiogenesis, oxidative stress, necroptosis, pyroptosis and indicators associated with signalling pathways in flaps were examined by IHC, immunofluorescence and Western blotting. Packaging adeno-associated virus (AAV) was used to activate STING in flaps. Results NRG1 promoted the survival of ischaemic flaps. An increased subcutaneous vascular network and neovascularization were found in ischaemic flaps after the application of NRG1. Transcriptomic gene ontology enrichment analysis and protein level detection indicated that necroptosis, pyroptosis and STING activity were reduced in the NRG1 group. The phosphorylation of AKT and forkhead box O3a (FOXO3a) were increased after NRG1 treatment. The increased expression of STING in flaps induced by AAV reversed the therapeutic effect of NRG1. The ability of NRG1 to phosphorylate AKT-FOXO3a, inhibit STING and promote flap survival was abolished after the application of the AKT inhibitor MK2206. Conclusions NRG1 inhibits pyroptosis and necroptosis by activating the AKT-FOXO3a signalling pathway to suppress STING activation and promote ischaemic flap survival.
Collapse
Affiliation(s)
- Xuwei Zhu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
| | - Gaoxiang Yu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
| | - Ya Lv
- The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang Street, Ouhai District, Wenzhou 325000, China
| | - Ningning Yang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
| | - Yinuo Zhao
- School of Pharmaceutical Science of Zhejiang Chinese Medical University, NO. 548 Binwen Road, Binjiang District, Hangzhou 310000, China
| | - Feida Li
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
| | - Jiayi Zhao
- The Second Clinical Medical College of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
| | - Zhuliu Chen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
| | - Yingying Lai
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
| | - Liang Chen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
| | - Xiangyang Wang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
| | - Jian Xiao
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Chashan University Town, Ouhai District, Wenzhou, 325000, China
| | - Yuepiao Cai
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Chashan University Town, Ouhai District, Wenzhou, 325000, China
| | | | - Jian Ding
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
| | - Weiyang Gao
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
| | - Kailiang Zhou
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
| | - Hui Xu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou 325027, China
| |
Collapse
|
8
|
Liu Y, Sun X, Gou Z, Deng Z, Zhang Y, Zhao P, Sun W, Bai Y, Jing Y. Epigenetic modifications in abdominal aortic aneurysms: from basic to clinical. Front Cardiovasc Med 2024; 11:1394889. [PMID: 38895538 PMCID: PMC11183338 DOI: 10.3389/fcvm.2024.1394889] [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: 03/11/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
Abdominal Aortic Aneurysm (AAA) is a disease characterized by localized dilation of the abdominal aorta, involving multiple factors in its occurrence and development, ultimately leading to vessel rupture and severe bleeding. AAA has a high mortality rate, and there is a lack of targeted therapeutic drugs. Epigenetic regulation plays a crucial role in AAA, and the treatment of AAA in the epigenetic field may involve a series of related genes and pathways. Abnormal expression of these genes may be a key factor in the occurrence of the disease and could potentially serve as promising therapeutic targets. Understanding the epigenetic regulation of AAA is of significant importance in revealing the mechanisms underlying the disease and identifying new therapeutic targets. This knowledge can contribute to offering AAA patients better clinical treatment options beyond surgery. This review systematically explores various aspects of epigenetic regulation in AAA, including DNA methylation, histone modification, non-coding RNA, and RNA modification. The analysis of the roles of these regulatory mechanisms, along with the identification of relevant genes and pathways associated with AAA, is discussed comprehensively. Additionally, a comprehensive discussion is provided on existing treatment strategies and prospects for epigenetics-based treatments, offering insights for future clinical interventions.
Collapse
Affiliation(s)
- YuChen Liu
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China
| | - XiaoYun Sun
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China
| | - Zhen Gou
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China
| | - ZhenKun Deng
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China
| | - YunRui Zhang
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China
| | - PingPing Zhao
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China
| | - Wei Sun
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China
| | - Yang Bai
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China
| | - YuChen Jing
- Department of Vascular Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
9
|
Li M, Wei P, Li K, Liu H, Alam N, Hou H, Deng J, Xu B, Liu E, Zhao S, Li Y. The incidence rate and histological characteristics of intimal hyperplasia in elastase-induced experimental abdominal aortic aneurysms in mice. Animal Model Exp Med 2024; 7:388-395. [PMID: 38017222 PMCID: PMC11228087 DOI: 10.1002/ame2.12362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 10/26/2023] [Indexed: 11/30/2023] Open
Abstract
Intimal hyperplasia (IH) is a negative vascular remodeling after arterial injury. IH occasionally occurs in elastase-induced abdominal aortic aneurysm (AAA) mouse models. This study aims to clarify the incidence and histological characteristics of IH in aneurysmal mice. A retrospective study was conducted by including 42 male elastase-induced mouse AAA models. The IH incidence, aortic diameters with or without IH, and hyperplasia lesional features of mice were analyzed. Among 42 elastase-induced AAA mouse models, 10 mice developed mild IH (24%) and severe IH was found in only 2 mice (5%). The outer diameters of the AAA segments in mice with and without IH did not show significant difference. Both mild and severe IH lesions show strong smooth muscle cell positive staining, but endothelial cells were occasionally observed in severe IH lesions. There was obvious macrophage infiltration in the IH lesions of the AAA mouse models, especially in mice with severe IH. However, only a lower numbers of T cells and B cells were found in the IH lesion. Local cell-secreted matrix metalloproteinases (MMP) 2 was highly expressed in all IH lesions, but MMP9 was only overexpressed in severe lesions. In conclusion, this study is the first to demonstrate the occurrence of aneurysmal IH and its histological characteristics in an elastase-induced mouse AAA model. This will help researchers better understand this model, and optimize it for use in AAA-related research.
Collapse
Affiliation(s)
- Meng Li
- Laboratory Animal CenterXi'an Jiaotong UniversityXi'anChina
- Department of Vascular SurgeryThe Second Hospital of Tianjin Medical UniversityTianjinChina
| | - Panpan Wei
- Laboratory Animal CenterXi'an Jiaotong UniversityXi'anChina
| | - Kexin Li
- Laboratory Animal CenterXi'an Jiaotong UniversityXi'anChina
| | - Haole Liu
- Laboratory Animal CenterXi'an Jiaotong UniversityXi'anChina
| | - Naqash Alam
- Laboratory Animal CenterXi'an Jiaotong UniversityXi'anChina
| | - Haiwen Hou
- Laboratory Animal CenterXi'an Jiaotong UniversityXi'anChina
| | - Jie Deng
- Department of CardiologyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Baohui Xu
- Department of Vascular SurgeryStanford University School of MedicineStanfordCaliforniaUSA
| | - Enqi Liu
- Laboratory Animal CenterXi'an Jiaotong UniversityXi'anChina
| | - Sihai Zhao
- Laboratory Animal CenterXi'an Jiaotong UniversityXi'anChina
- Department of CardiologyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Yankui Li
- Department of Vascular SurgeryThe Second Hospital of Tianjin Medical UniversityTianjinChina
| |
Collapse
|
10
|
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: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [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
|
11
|
Huang Y, Herbst EB, Xie Y, Yin L, Islam ZH, Kent EW, Wang B, Klibanov AL, Hossack JA. In Vivo Validation of Modulated Acoustic Radiation Force-Based Imaging in Murine Model of Abdominal Aortic Aneurysm Using VEGFR-2-Targeted Microbubbles. Invest Radiol 2023; 58:865-873. [PMID: 37433074 PMCID: PMC10784413 DOI: 10.1097/rli.0000000000001000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2023]
Abstract
OBJECTIVES The objective of this study is to validate the modulated acoustic radiation force (mARF)-based imaging method in the detection of abdominal aortic aneurysm (AAA) in murine models using vascular endothelial growth factor receptor 2 (VEGFR-2)-targeted microbubbles (MBs). MATERIALS AND METHODS The mouse AAA model was prepared using the subcutaneous angiotensin II (Ang II) infusion combined with the β-aminopropionitrile monofumarate solution dissolved in drinking water. The ultrasound imaging session was performed at 7 days, 14 days, 21 days, and 28 days after the osmotic pump implantation. For each imaging session, 10 C57BL/6 mice were implanted with Ang II-filled osmotic pumps, and 5 C57BL/6 mice received saline infusion only as the control group. Biotinylated lipid MBs conjugated to either anti-mouse VEGFR-2 antibody (targeted MBs) or isotype control antibody (control MBs) were prepared before each imaging session and were injected into mice via tail vein catheter. Two separate transducers were colocalized to image the AAA and apply ARF to translate MBs simultaneously. After each imaging session, tissue was harvested and the aortas were used for VEGFR-2 immunostaining analysis. From the collected ultrasound image data, the signal magnitude response of the adherent targeted MBs was analyzed, and a parameter, residual-to-saturation ratio ( Rres - sat ), was defined to measure the enhancement in the adherent targeted MBs signal after the cessation of ARF compared with the initial signal intensity. Statistical analysis was performed with the Welch t test and analysis of variance test. RESULTS The Rres - sat of abdominal aortic segments from Ang II-challenged mice was significantly higher compared with that in the saline-infused control group ( P < 0.001) at all 4 time points after osmotic pump implantation (1 week to 4 weeks). In control mice, the Rres - sat values were 2.13%, 1.85%, 3.26%, and 4.85% at 1, 2, 3, and 4 weeks postimplantation, respectively. In stark contrast, the Rres - sat values for the mice with Ang II-induced AAA lesions were 9.20%, 20.6%, 22.7%, and 31.8%, respectively. It is worth noting that there was a significant difference between the Rres - sat for Ang II-infused mice at all 4 time points ( P < 0.005), a finding not present in the saline-infused mice. Immunostaining results revealed the VEGFR-2 expression was increased in the abdominal aortic segments of Ang II-infused mice compared with the control group. CONCLUSIONS The mARF-based imaging technique was validated in vivo using a murine model of AAA and VEGFR-2-targeted MBs. Results in this study indicated that the mARF-based imaging technique has the ability to detect and assess AAA growth at early stages based on the signal intensity of adherent targeted MBs, which is correlated with the expression level of the desired molecular biomarker. The results may suggest, in very long term, a pathway toward eventual clinical implementation for an ultrasound molecular imaging-based approach to AAA risk assessment in asymptomatic patients.
Collapse
Affiliation(s)
- Yi Huang
- From the Department of Biomedical Engineering, University of Virginia, Charlottesville, VA (Y.H., Y.X., J.A.H.); Philips Research North America, Cambridge, MA (E.B.H.); Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA (L.Y., Z.H.I., E.W.K., B.W.); and Division of Cardiovascular Medicine, Cardiovascular Research Center and Department of Biomedical Engineering, University of Virginia, Charlottesville, VA (A.L.K.)
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Luo Y, Yang J, Zhang L, Tai Z, Huang H, Xu Z, Zhang H. Phosphoglycerate kinase (PGK) 1 succinylation modulates epileptic seizures and the blood-brain barrier. Exp Anim 2023; 72:475-489. [PMID: 37258131 PMCID: PMC10658094 DOI: 10.1538/expanim.23-0019] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 05/22/2023] [Indexed: 06/02/2023] Open
Abstract
Epilepsy is the most common chronic disorder in the nervous system, mainly characterized by recurrent, periodic, unpredictable seizures. Post-translational modifications (PTMs) are important protein functional regulators that regulate various physiological and pathological processes. It is significant for cell activity, stability, protein folding, and localization. Phosphoglycerate kinase (PGK) 1 has traditionally been studied as an important adenosine triphosphate (ATP)-generating enzyme of the glycolytic pathway. PGK1 catalyzes the reversible transfer of a phosphoryl group from 1, 3-bisphosphoglycerate (1, 3-BPG) to ADP, producing 3-phosphoglycerate (3-PG) and ATP. In addition to cell metabolism regulation, PGK1 is involved in multiple biological activities, including angiogenesis, autophagy, and DNA repair. However, the exact role of PGK1 succinylation in epilepsy has not been thoroughly investigated. The expression of PGK1 succinylation was analyzed by Immunoprecipitation. Western blots were used to assess the expression of PGK1, angiostatin, and vascular endothelial growth factor (VEGF) in a rat model of lithium-pilocarpine-induced acute epilepsy. Behavioral experiments were performed in a rat model of lithium-pilocarpine-induced acute epilepsy. ELISA method was used to measure the level of S100β in serum brain biomarkers' integrity of the blood-brain barrier. The expression of the succinylation of PGK1 was decreased in a rat model of lithium-pilocarpine-induced acute epilepsy compared with the normal rats in the hippocampus. Interestingly, the lysine 15 (K15), and the arginine (R) variants of lentivirus increased the susceptibility in a rat model of lithium-pilocarpine-induced acute epilepsy, and the K15 the glutamate (E) variants, had the opposite effect. In addition, the succinylation of PGK1 at K15 affected the expression of PGK1 succinylation but not the expression of PGK1total protein. Furthermore, the study found that the succinylation of PGK1 at K15 may affect the level of angiostatin and VEGF in the hippocampus, which also affects the level of S100β in serum. In conclusion, the mutation of the K15 site of PGK1 may alter the expression of the succinylation of PGK1 and then affect the integrity of the blood-brain barrier through the angiostatin / VEGF pathway altering the activity of epilepsy, which may be one of the new mechanisms of treatment strategies.
Collapse
Affiliation(s)
- Yuemei Luo
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Zunyi, Guizhou 563003, P.R. China
| | - Juan Yang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Zunyi, Guizhou 563003, P.R. China
| | - Lijia Zhang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Zunyi, Guizhou 563003, P.R. China
| | - Zhenzhen Tai
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Zunyi, Guizhou 563003, P.R. China
| | - Hao Huang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Zunyi, Guizhou 563003, P.R. China
| | - Zucai Xu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Zunyi, Guizhou 563003, P.R. China
| | - Haiqing Zhang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Zunyi, Guizhou 563003, P.R. China
| |
Collapse
|
13
|
Kang S, Yeon B, Kim MS, Yoo M, Kim B, Yu YM. Aneurysm and Artery Dissection After Oral VEGFR-TKI Use in Adults With Cancer. JAMA Netw Open 2023; 6:e2345977. [PMID: 38019511 PMCID: PMC10687660 DOI: 10.1001/jamanetworkopen.2023.45977] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 10/23/2023] [Indexed: 11/30/2023] Open
Abstract
Importance The association of tyrosine kinase inhibitors targeting vascular endothelial growth factor receptors (VEGFR-TKIs) with aneurysm and artery dissection (AAD) has been frequently reported in spontaneous reporting databases. Objective To investigate the risk and incidence of AAD occurrence in patients with cancer treated with oral VEGFR-TKIs, with capecitabine as an active comparator. Design, Setting, and Participants This national, historical cohort study was conducted using national claims data from the National Health Insurance Service in Korea from 2007 to 2020, with a 1-year follow-up. Patients with cancer aged 40 years or older prescribed oral VEGFR-TKIs or capecitabine were enrolled. Data were analyzed from September 2022 through April 2023. Exposure Oral VEGFR-TKIs (sorafenib, regorafenib, vandetanib, sunitinib, lenvatinib, axitinib, and pazopanib) or capecitabine as a comparator. Main Outcomes and Measures Hazard ratios (HRs) were used to investigate the association between VEGFR-TKI use and AAD after propensity score matching. The primary outcome was AAD, and secondary outcomes were aortic aneurysm and dissection and AAD with rupture. Outcomes were defined using International Statistical Classification of Diseases and Related Health Problems, Tenth Revision (ICD-10) diagnosis codes. Results Among 127 710 patients with cancer eligible for the study (80 386 males [62.9%]; mean [SD] age, 62.6 [10.9] years), 37 308 patients received VEGFR-TKIs and 90 402 patients received capecitabine. Among 27 535 matched patients receiving VEGFR-TKIs, the incidence of AAD within 1 year of treatment initiation was 6.0 per 1000 person-years. The median (IQR) time to AAD onset in the matched AAD group was 114 (67-257) days after treatment initiation, with the highest incidence observed during the first 3 months (45 incidents vs 31, 17, and 16 incidents during 3- to 6-month, 6- to 9-month, and 9- to 12-month periods, respectively). Cox regression modeling showed that the risk of AAD occurrence was significantly higher among patients prescribed VEGFR-TKIs than those receiving capecitabine (HR, 1.48; 95% CI, 1.08-2.02); similar results were obtained among females (HR, 2.08; 95% CI, 1.26-3.42), older adults (aged ≥65 years; HR, 1.42; 95% CI, 1.01-1.99), and patients with dyslipidemia (HR, 1.58; 95% CI, 1.11-2.24). Conclusions and Relevance In this study, the use of oral VEGFR-TKIs was associated with an increased risk of AAD occurrence. These findings elucidate vascular toxic effects and may provide a substantial reference for reducing the socioeconomic burden of adverse events associated with VEGFR-TKI use.
Collapse
Affiliation(s)
- Soyoung Kang
- Department of Drug Safety Information, Korea Institute of Drug Safety and Risk Management, Gyeonggi-do, Republic of Korea
| | - Bora Yeon
- Department of Drug Safety Information, Korea Institute of Drug Safety and Risk Management, Gyeonggi-do, Republic of Korea
| | - Myo-Song Kim
- Department of Drug Safety Information, Korea Institute of Drug Safety and Risk Management, Gyeonggi-do, Republic of Korea
| | - Myungsik Yoo
- Department of Drug Safety Information, Korea Institute of Drug Safety and Risk Management, Gyeonggi-do, Republic of Korea
| | - Bonggi Kim
- Department of Drug Safety Information, Korea Institute of Drug Safety and Risk Management, Gyeonggi-do, Republic of Korea
| | - Yun Mi Yu
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, Republic of Korea
- Department of Pharmaceutical Medicine and Regulatory Sciences, College of Pharmacy, Yonsei University, Incheon, Republic of Korea
| |
Collapse
|
14
|
Fu Y, Liu H, Li K, Wei P, Alam N, Deng J, Li M, Wu H, He X, Hou H, Xia C, Wang R, Wang W, Bai L, Xu B, Li Y, Wu Y, Liu E, Zhao S. C-reactive protein deficiency ameliorates experimental abdominal aortic aneurysms. Front Immunol 2023; 14:1233807. [PMID: 37753091 PMCID: PMC10518468 DOI: 10.3389/fimmu.2023.1233807] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 08/25/2023] [Indexed: 09/28/2023] Open
Abstract
Background C-reactive protein (CRP) levels are elevated in patients with abdominal aortic aneurysms (AAA). However, it has not been investigated whether CRP contributes to AAA pathogenesis. Methods CRP deficient and wild type (WT) male mice were subjected to AAA induction via transient intra-aortic infusion of porcine pancreatic elastase. AAAs were monitored by in situ measurements of maximal infrarenal aortic external diameters immediately prior to and 14 days following elastase infusion. Key AAA pathologies were assessed by histochemical and immunohistochemical staining procedures. The influence of CRP deficiency on macrophage activation was evaluated in peritoneal macrophages in vitro. Results CRP protein levels were higher in aneurysmal than that in non-aneurysmal aortas. Aneurysmal aortic dilation was markedly suppressed in CRP deficient (aortic diameter: 1.08 ± 0.11 mm) as compared to WT (1.21 ± 0.08 mm) mice on day 14 after elastase infusion. More medial elastin was retained in CRP deficient than in WT elastase-infused mice. Macrophage accumulation was significantly less in aneurysmal aorta from CRP deficient than that from WT mice. Matrix metalloproteinase 2 expression was also attenuated in CRP deficient as compared to WT aneurysmal aortas. CRP deficiency had no recognizable influence on medial smooth muscle loss, lymphocyte accumulation, aneurysmal angiogenesis, and matrix metalloproteinase 9 expression. In in vitro assays, mRNA levels for tumor necrosis factor α and cyclooxygenase 2 were reduced in lipopolysaccharide activated peritoneal macrophages from CRP deficient as compared to wild type mice. Conclusion CRP deficiency suppressed experimental AAAs by attenuating aneurysmal elastin destruction, macrophage accumulation and matrix metalloproteinase 2 expression.
Collapse
Affiliation(s)
- Yu Fu
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Haole Liu
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Kexin Li
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Panpan Wei
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Naqash Alam
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Jie Deng
- Department of Cardiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Meng Li
- Department of Vascular Surgery, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Haibin Wu
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Xue He
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Haiwen Hou
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Congcong Xia
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Rong Wang
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Weirong Wang
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Liang Bai
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Baohui Xu
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Yankui Li
- Department of Vascular Surgery, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Yi Wu
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an, Shaanxi, China
| | - Enqi Liu
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Sihai Zhao
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
- Department of Cardiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| |
Collapse
|
15
|
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
|
16
|
Zalewski D, Chmiel P, Kołodziej P, Borowski G, Feldo M, Kocki J, Bogucka-Kocka A. Dysregulations of Key Regulators of Angiogenesis and Inflammation in Abdominal Aortic Aneurysm. Int J Mol Sci 2023; 24:12087. [PMID: 37569462 PMCID: PMC10418409 DOI: 10.3390/ijms241512087] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/21/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
Abdominal aortic aneurysm (AAA) is a chronic vascular disease caused by localized weakening and broadening of the abdominal aorta. AAA is a clearly underdiagnosed disease and is burdened with a high mortality rate (65-85%) from AAA rupture. Studies indicate that abnormal regulation of angiogenesis and inflammation contributes to progression and onset of this disease; however, dysregulations in the molecular pathways associated with this disease are not yet fully explained. Therefore, in our study, we aimed to identify dysregulations in the key regulators of angiogenesis and inflammation in patients with AAA in peripheral blood mononuclear cells (using qPCR) and plasma samples (using ELISA). Expression levels of ANGPT1, CXCL8, PDGFA, TGFB1, VEGFB, and VEGFC and plasma levels of TGF-alpha, TGF-beta 1, VEGF-A, and VEGF-C were found to be significantly altered in the AAA group compared to the control subjects without AAA. Associations between analyzed factors and risk factors or biochemical parameters were also explored. Any of the analyzed factors was associated with the size of the aneurysm. The presented study identified dysregulations in key angiogenesis- and inflammation-related factors potentially involved in AAA formation, giving new insight into the molecular pathways involved in the development of this disease and providing candidates for biomarkers that could serve as diagnostic or therapeutic targets.
Collapse
Affiliation(s)
- Daniel Zalewski
- Chair and Department of Biology and Genetics, Medical University of Lublin, 4a Chodźki St., 20-093 Lublin, Poland; (P.C.); (P.K.); (A.B.-K.)
| | - Paulina Chmiel
- Chair and Department of Biology and Genetics, Medical University of Lublin, 4a Chodźki St., 20-093 Lublin, Poland; (P.C.); (P.K.); (A.B.-K.)
| | - Przemysław Kołodziej
- Chair and Department of Biology and Genetics, Medical University of Lublin, 4a Chodźki St., 20-093 Lublin, Poland; (P.C.); (P.K.); (A.B.-K.)
| | - Grzegorz Borowski
- Chair and Department of Vascular Surgery and Angiology, Medical University of Lublin, 11 Staszica St., 20-081 Lublin, Poland; (G.B.); (M.F.)
| | - Marcin Feldo
- Chair and Department of Vascular Surgery and Angiology, Medical University of Lublin, 11 Staszica St., 20-081 Lublin, Poland; (G.B.); (M.F.)
| | - Janusz Kocki
- Department of Clinical Genetics, Chair of Medical Genetics, Medical University of Lublin, 11 Radziwiłłowska St., 20-080 Lublin, Poland;
| | - Anna Bogucka-Kocka
- Chair and Department of Biology and Genetics, Medical University of Lublin, 4a Chodźki St., 20-093 Lublin, Poland; (P.C.); (P.K.); (A.B.-K.)
| |
Collapse
|
17
|
Guo J, Shi J, Qin M, Wang Y, Li Z, Shoji T, Ikezoe T, Ge Y, Xu B. Pharmacological Inhibition of Gasdermin D Suppresses Angiotensin II-Induced Experimental Abdominal Aortic Aneurysms. Biomolecules 2023; 13:899. [PMID: 37371479 PMCID: PMC10295961 DOI: 10.3390/biom13060899] [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: 04/10/2023] [Revised: 05/08/2023] [Accepted: 05/11/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND Gasdermin D, a molecule downstream of the nucleotide-binding oligomerization domain-like receptor family pyrin domain containing inflammasome, forms the membrane pore for the secretion of interleukin (IL)-1β and IL-18, and also mediates pyroptosis. This study was to explore the influence of treatment with disulfiram, a small molecule inhibitor to gasdermin D, on the formation and progression of experimental abdominal aortic aneurysms (AAA). METHODS AAAs were induced in 10-week-old male apolipoprotein E deficient mice by subcutaneous infusion of angiotensin II (1000 ng/min/kg body weight) for 28 days via osmotic minipumps. Three days prior to angiotensin II infusion, disulfiram (50 mg/kg) or an equal volume of saline as the vehicle control was administered daily via oral gavage. The influence on experimental AAAs was analyzed by serial measurements of aortic diameters via ultrasonography, grading AAA severity and histopathology at sacrifice. Serum IL-1β and IL-18 levels, systolic blood pressure, total cholesterol, and triglyceride were also measured. Additional experiments assayed the influences on the cell viability and IL-1β secretion of in vitro activated macrophages. RESULTS Disulfiram significantly reduced the enlargement, incidence, and severity of angiotensin II-induced experimental AAAs with attenuation of medial elastin breaks, mural macrophage accumulation, and systolic blood pressure. The AAA suppression was also associated with reduced systemic levels of IL-1β but not IL-18. However, disulfiram treatment had no impact on body weight gain and lipid levels in aneurysmal mice. Additionally, disulfiram treatment also markedly reduced the secretion of IL-1β from activated macrophages with a limited effect on cell viability in vitro. CONCLUSIONS Gasdermin D inhibition by disulfiram attenuated angiotensin II-induced experimental AAAs with reduced systemic IL-1β levels and in vitro activated macrophage IL-1β secretion. Our study suggests that pharmacological gasdermin D inhibition may have translational potential for limiting clinical AAA progression.
Collapse
Affiliation(s)
- Jia Guo
- Center for Hypertension Care, Shanxi Medical University First Hospital, Taiyuan 030001, China; (J.S.); (M.Q.)
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; (T.S.); (T.I.); (B.X.)
| | - Jinyun Shi
- Center for Hypertension Care, Shanxi Medical University First Hospital, Taiyuan 030001, China; (J.S.); (M.Q.)
| | - Min Qin
- Center for Hypertension Care, Shanxi Medical University First Hospital, Taiyuan 030001, China; (J.S.); (M.Q.)
| | - Yan Wang
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing 100191, China;
| | - Zhidong Li
- Department of Pharmacology, Shanxi Medical University, Taiyuan 030001, China;
| | - Takahiro Shoji
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; (T.S.); (T.I.); (B.X.)
| | - Toru Ikezoe
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; (T.S.); (T.I.); (B.X.)
| | - Yingbin Ge
- Department of Physiology, Nanjing Medical University, Nanjing 211166, China;
| | - Baohui Xu
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; (T.S.); (T.I.); (B.X.)
| |
Collapse
|
18
|
Li Y, Zheng X, Guo J, Samura M, Ge Y, Zhao S, Li G, Chen X, Shoji T, Ikezoe T, Miyata M, Xu B, Dalman RL. Treatment With Small Molecule Inhibitors of Advanced Glycation End-Products Formation and Advanced Glycation End-Products-Mediated Collagen Cross-Linking Promotes Experimental Aortic Aneurysm Progression in Diabetic Mice. J Am Heart Assoc 2023; 12:e028081. [PMID: 37158066 PMCID: PMC10227285 DOI: 10.1161/jaha.122.028081] [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: 09/05/2022] [Accepted: 04/14/2023] [Indexed: 05/10/2023]
Abstract
Background Although diabetes attenuates abdominal aortic aneurysms (AAAs), the mechanisms by which diabetes suppresses AAAs remain incompletely understood. Accumulation of advanced glycation end- (AGEs) reduces extracellular matrix (ECM) degradation in diabetes. Because ECM degradation is critical for AAA pathogenesis, we investigated whether AGEs mediate experimental AAA suppression in diabetes by blocking AGE formation or disrupting AGE-ECM cross-linking using small molecule inhibitors. Methods and Results Male C57BL/6J mice were treated with streptozotocin and intra-aortic elastase infusion to induce diabetes and experimental AAAs, respectively. Aminoguanidine (AGE formation inhibitor, 200 mg/kg), alagebrium (AGE-ECM cross-linking disrupter, 20 mg/kg), or vehicle was administered daily to mice from the last day following streptozotocin injection. AAAs were assessed via serial aortic diameter measurements, histopathology, and in vitro medial elastolysis assays. Treatment with aminoguanidine, not alagebrium, diminished AGEs in diabetic AAAs. Treatment with both inhibitors enhanced aortic enlargement in diabetic mice as compared with vehicle treatment. Neither enhanced AAA enlargement in nondiabetic mice. AAA enhancement in diabetic mice by aminoguanidine or alagebrium treatment promoted elastin degradation, smooth muscle cell depletion, mural macrophage accumulation, and neoangiogenesis without affecting matrix metalloproteinases, C-C motif chemokine ligand 2, or serum glucose concentration. Additionally, treatment with both inhibitors reversed suppression of diabetic aortic medial elastolysis by porcine pancreatic elastase in vitro. Conclusions Inhibiting AGE formation or AGE-ECM cross-linking enhances experimental AAAs in diabetes. These findings support the hypothesis that AGEs attenuate experimental AAAs in diabetes. These findings underscore the potential translational value of enhanced ECM cross-linking as an inhibitory strategy for early AAA disease.
Collapse
Affiliation(s)
- Yankui Li
- Department of SurgeryStanford University School of MedicineStanfordCAUSA
- Department of Vascular SurgeryTianjin Medical University Second HospitalTianjinChina
| | - Xiaoya Zheng
- Department of SurgeryStanford University School of MedicineStanfordCAUSA
- Department of EndocrinologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Jia Guo
- Department of SurgeryStanford University School of MedicineStanfordCAUSA
| | - Makoto Samura
- Department of SurgeryStanford University School of MedicineStanfordCAUSA
| | - Yingbin Ge
- Department of PhysiologyNanjing Medical UniversityNanjingChina
| | - Sihai Zhao
- Department of SurgeryStanford University School of MedicineStanfordCAUSA
| | - Gang Li
- Department of SurgeryStanford University School of MedicineStanfordCAUSA
| | - Xiaofeng Chen
- Department of Radiation OncologyIndiana University School of MedicineIndianapolisINUSA
| | - Takahiro Shoji
- Department of SurgeryStanford University School of MedicineStanfordCAUSA
| | - Toru Ikezoe
- Department of SurgeryStanford University School of MedicineStanfordCAUSA
| | - Masaaki Miyata
- School of Health SciencesKagoshima University Faculty of MedicineKagoshimaJapan
| | - Baohui Xu
- Department of SurgeryStanford University School of MedicineStanfordCAUSA
| | - Ronald L. Dalman
- Department of SurgeryStanford University School of MedicineStanfordCAUSA
| |
Collapse
|
19
|
Zhuang J, Zhu H, Cheng Z, Hu X, Yu X, Li J, Liu H, Tang P, Zhang Y, Xiong X, Deng H. PCSK9, a novel immune and ferroptosis related gene in abdominal aortic aneurysm neck. Sci Rep 2023; 13:6054. [PMID: 37055467 PMCID: PMC10102181 DOI: 10.1038/s41598-023-33287-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 04/11/2023] [Indexed: 04/15/2023] Open
Abstract
The gene expression profile of abdominal aortic aneurysm (AAA) neck is not fully understood. The etiology of AAA is considered to be related to atherosclerosis and the inflammatory response, involving congenital, genetic, metabolic, and other factors. The level of proprotein convertase subtilisin/kexin type 9 (PCSK9) is related to those of cholesterol, oxidized low-density lipoprotein, and triglycerides. PCSK9 inhibitors have significant effects on lowering LDL-cholesterol, reversing atherosclerotic plaques, and reducing the risk of cardiovascular events and have been approved by several lipid-lowering guidelines. This work was aimed to investigate the potential role of PCSK9 in the neck of AAA. We extracted the expression dataset (GSE47472) containing 14 AAA patients and 8 donors and single-cell RNAseq (scRNA-seq) data (GSE164678) of CaCl2-induced (AAA) samples from the Gene Expression Omnibus dataset. Through bioinformatics methods, we found that PCSK9 was up-regulated in the proximal neck of human AAA. In AAA, PCSK9 was mainly expressed in fibroblasts. Additionally, immune check-point PDCD1LG2 was also expressed higher in AAA neck than donor, while CTLA4, PDCD1, and SIGLEC15 were down-regulated in AAA neck. The expression of PCSK was correlated with PDCD1LG2, LAG3, and CTLA4 in AAA neck. Additionally, some ferroptosis-related genes were also down-regulated in AAA neck. PCSK9 was also correlated with ferroptosis-related genes in AAA neck. In conclusion, PCSK9 was highly expressed in AAA neck, and may exert its role through interacting with immune check-points and ferroptosis-related genes.
Collapse
Affiliation(s)
- Junli Zhuang
- Department of Vascular Surgery, Renmin Hospital of Wuhan University, NO. 99 Zhang Zhidong Road, Wuchang District, Wuhan, 430060, China
| | - Hua Zhu
- Department of Vascular Surgery, Renmin Hospital of Wuhan University, NO. 99 Zhang Zhidong Road, Wuchang District, Wuhan, 430060, China
- Department of Neurosurgery, The Affiliated Huzhou Hospital, Zhejiang University School of Medicine (Huzhou Central Hospital), NO. 1558 North Sanhuan Road, Huzhou, 313003, Zhejiang, China
- Department of Neurosurgery, Renmin Hospital of Wuhan University, NO. 99 Zhang Zhidong Road, Wuchang District, Wuhan, 430060, China
| | - Ziqi Cheng
- Department of Neurosurgery, Renmin Hospital of Wuhan University, NO. 99 Zhang Zhidong Road, Wuchang District, Wuhan, 430060, China
| | - Xinyao Hu
- Cancer Center, Renmin Hospital of Wuhan University, NO. 99 Zhang Zhidong Road, Wuchang District, Wuhan, 430060, China
| | - Xiaohui Yu
- Department of Vascular Surgery, Renmin Hospital of Wuhan University, NO. 99 Zhang Zhidong Road, Wuchang District, Wuhan, 430060, China
| | - Jie Li
- Department of Vascular Surgery, Renmin Hospital of Wuhan University, NO. 99 Zhang Zhidong Road, Wuchang District, Wuhan, 430060, China
| | - Huagang Liu
- Department of Vascular Surgery, Renmin Hospital of Wuhan University, NO. 99 Zhang Zhidong Road, Wuchang District, Wuhan, 430060, China
| | - Peng Tang
- Department of Vascular Surgery, Renmin Hospital of Wuhan University, NO. 99 Zhang Zhidong Road, Wuchang District, Wuhan, 430060, China
| | - Ying Zhang
- Department of Vascular Surgery, Renmin Hospital of Wuhan University, NO. 99 Zhang Zhidong Road, Wuchang District, Wuhan, 430060, China
| | - Xiaoxing Xiong
- Department of Vascular Surgery, Renmin Hospital of Wuhan University, NO. 99 Zhang Zhidong Road, Wuchang District, Wuhan, 430060, China.
- Department of Neurosurgery, The Affiliated Huzhou Hospital, Zhejiang University School of Medicine (Huzhou Central Hospital), NO. 1558 North Sanhuan Road, Huzhou, 313003, Zhejiang, China.
- Department of Neurosurgery, Renmin Hospital of Wuhan University, NO. 99 Zhang Zhidong Road, Wuchang District, Wuhan, 430060, China.
| | - Hongping Deng
- Department of Vascular Surgery, Renmin Hospital of Wuhan University, NO. 99 Zhang Zhidong Road, Wuchang District, Wuhan, 430060, China.
| |
Collapse
|
20
|
Fu W, Liu H, Wei P, Xia C, Yu Q, Tian K, Li Y, Liu E, Xu B, Miyata M, Wang R, Zhao S. Genetic deficiency of protein inhibitor of activated STAT3 suppresses experimental abdominal aortic aneurysms. Front Cardiovasc Med 2023; 10:1092555. [PMID: 37008329 PMCID: PMC10050368 DOI: 10.3389/fcvm.2023.1092555] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 02/27/2023] [Indexed: 03/17/2023] Open
Abstract
AimSignal transducer and activator of transcription (STAT) signaling is critical for the pathogenesis of abdominal aortic aneurysms (AAAs). Though protein inhibitor of activated STAT3 (PIAS3) negatively modulates STAT3 activity, but its role in AAA disease remains undefined.MethodAAAs were induced in PIAS3 deficient (PIAS3−/−) and wild type (PIAS3+/+) male mice via transient intra-aortic elastase infusion. AAAs were assessed by in situ measurements of infrarenal aortic external diameters prior to (day 0) and 14 days after elastase infusion. Characteristic aneurysmal pathologies were evaluated by histopathology.ResultsFourteen days following elastase infusion, aneurysmal aortic diameter was reduced by an approximately 50% in PIAS3−/− as compared to PIAS3+/+ mice. On histological analyses, PIAS3−/− mice showed less medial elastin degradation (media score: 2.5) and smooth muscle cell loss (media score: 3.0) than those in PIAS3+/+ mice (media score: 4 for both elastin and SMC destruction). Aortic wall leukocyte accumulation including macrophages, CD4+ T cells, CD8+ T cells and B cells as well as mural neovessel formation were significantly reduced in PIAS3−/− as compared to PIAS3+/+ mice. Additionally, PIAS3 deficiency also downregulated the expression levels of matrix metalloproteinases 2 and 9 by 61% and 70%, respectively, in aneurysmal lesion.ConclusionPIAS3 deficiency ameliorated experimental AAAs in conjunction with reduced medial elastin degradation and smooth muscle cell depletion, mural leukocyte accumulation and angiogenesis.
Collapse
Affiliation(s)
- Weilai Fu
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi’an, China
- Department of Vascular Surgery, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Haole Liu
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi’an, China
| | - Panpan Wei
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi’an, China
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi’an, China
| | - Congcong Xia
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi’an, China
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi’an, China
| | - Qingqing Yu
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi’an, China
| | - Kangli Tian
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi’an, China
| | - Yankui Li
- Department of Vascular Surgery, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Enqi Liu
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi’an, China
| | - Baohui Xu
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Masaaki Miyata
- School of Health Science, Faculty of Medicine, Kagoshima University, Kagoshima, Japan
| | - Rong Wang
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi’an, China
- Correspondence: Rong Wang Sihai Zhao
| | - Sihai Zhao
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi’an, China
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi’an, China
- Correspondence: Rong Wang Sihai Zhao
| |
Collapse
|
21
|
Xu B, Li G, Li Y, Deng H, Cabot A, Guo J, Samura M, Zheng X, Chen T, Zhao S, Fujimura N, Dalman RL. Mechanisms and efficacy of metformin-mediated suppression of established experimental abdominal aortic aneurysms. JVS Vasc Sci 2023; 4:100102. [PMID: 37168662 PMCID: PMC10165270 DOI: 10.1016/j.jvssci.2023.100102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 02/16/2023] [Indexed: 04/03/2023] Open
Abstract
Objective Metformin treatment attenuates experimental abdominal aortic aneurysm (AAA) formation, as well as reduces clinical AAA diameter enlargement in patients with diabetes. The mechanisms of metformin-mediated aneurysm suppression, and its efficacy in suppressing established experimental aneurysms, remain uncertain. Methods Experimental AAAs were created in male C57BL/6J mice via intra-aortic infusion of porcine pancreatic elastase. Metformin alone (250 mg/kg), or metformin combined with the 5' AMP-activated protein kinase (AMPK) antagonist Compound C (10 mg/kg), were administered to respective mouse cohorts daily beginning 4 days following AAA induction. Further AAA cohorts received either the AMPK agonist AICA riboside (500 mg/kg) as positive, or vehicle (saline) as negative, controls. AAA progression in all groups was assessed via serial in vivo ultrasonography and histopathology at sacrifice. Cytokine-producing T cells and myeloid cellularity were determined by flow cytometric analyses. Results Metformin limited established experimental AAA progression at 3 (-85%) and 10 (-68%) days following treatment initiation compared with saline control. Concurrent Compound C treatment reduced this effect by approximately 50%. In metformin-treated mice, reduced AAA progression was associated with relative elastin preservation, smooth muscle cell preservation, and reduced mural leukocyte infiltration and neoangiogenesis compared with vehicle control group. Metformin also resulted in reduced interferon-γ-, but not interleukin-10 or -17, producing splenic T cells in aneurysmal mice. Additionally, metformin therapy increased circulating and splenic inflammatory monocytes (CD11b+Ly-6Chigh), but not neutrophils (CD11b+Ly-6G+), with no effect on respective bone marrow cell populations. Conclusions Metformin treatment suppresses existing experimental AAA progression in part via AMPK agonist activity, limiting interferon-γ-producing T cell differentiation while enhancing circulating and splenic inflammatory monocyte retention.
Collapse
|
22
|
Sophocleous F, De Garate E, Bigotti MG, Anwar M, Jover E, Chamorro-Jorganes A, Rajakaruna C, Mitrousi K, De Francesco V, Wilson A, Stoica S, Parry A, Benedetto U, Chivasso P, Gill F, Hamilton MCK, Bucciarelli-Ducci C, Caputo M, Emanueli C, Biglino G. A Segmental Approach from Molecular Profiling to Medical Imaging to Study Bicuspid Aortic Valve Aortopathy. Cells 2022; 11:cells11233721. [PMID: 36496981 PMCID: PMC9737804 DOI: 10.3390/cells11233721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/01/2022] [Accepted: 11/14/2022] [Indexed: 11/25/2022] Open
Abstract
Bicuspid aortic valve (BAV) patients develop ascending aortic (AAo) dilation. The pathogenesis of BAV aortopathy (genetic vs. haemodynamic) remains unclear. This study aims to identify regional changes around the AAo wall in BAV patients with aortopathy, integrating molecular data and clinical imaging. BAV patients with aortopathy (n = 15) were prospectively recruited to surgically collect aortic tissue and measure molecular markers across the AAo circumference. Dilated (anterior/right) vs. non-dilated (posterior/left) circumferential segments were profiled for whole-genomic microRNAs (next-generation RNA sequencing, miRCURY LNA PCR), protein content (tandem mass spectrometry), and elastin fragmentation and degeneration (histomorphometric analysis). Integrated bioinformatic analyses of RNA sequencing and proteomic datasets identified five microRNAs (miR-128-3p, miR-210-3p, miR-150-5p, miR-199b-5p, and miR-21-5p) differentially expressed across the AAo circumference. Among them, three miRNAs (miR-128-3p, miR-150-5p, and miR-199b-5p) were predicted to have an effect on eight common target genes, whose expression was dysregulated, according to proteomic analyses, and involved in the vascular-endothelial growth-factor signalling, Hippo signalling, and arachidonic acid pathways. Decreased elastic fibre levels and elastic layer thickness were observed in the dilated segments. Additionally, in a subset of patients n = 6/15, a four-dimensional cardiac magnetic resonance (CMR) scan was performed. Interestingly, an increase in wall shear stress (WSS) was observed at the anterior/right wall segments, concomitantly with the differentially expressed miRNAs and decreased elastic fibres. This study identified new miRNAs involved in the BAV aortic wall and revealed the concomitant expressional dysregulation of miRNAs, proteins, and elastic fibres on the anterior/right wall in dilated BAV patients, corresponding to regions of elevated WSS.
Collapse
Affiliation(s)
- Froso Sophocleous
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol BS8 1TH, UK
- Bristol Heart Institute, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol BS1 3NU, UK
| | - Estefania De Garate
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol BS8 1TH, UK
- Bristol Heart Institute, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol BS1 3NU, UK
| | - Maria Giulia Bigotti
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol BS8 1TH, UK
- School of Biochemistry, Faculty of Life Sciences, University of Bristol, Bristol BS8 1TH, UK
| | - Maryam Anwar
- National Heart and Lung Institute, Imperial College London, London SW7 2BX, UK
| | - Eva Jover
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol BS8 1TH, UK
- Instituto de Investigación Sanitaria de Navarra, 31008 Pamplona, Spain
| | | | - Cha Rajakaruna
- Bristol Heart Institute, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol BS1 3NU, UK
| | - Konstantina Mitrousi
- Bristol Heart Institute, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol BS1 3NU, UK
| | - Viola De Francesco
- Bristol Heart Institute, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol BS1 3NU, UK
| | - Aileen Wilson
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol BS8 1TH, UK
- Bristol Heart Institute, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol BS1 3NU, UK
| | - Serban Stoica
- Bristol Heart Institute, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol BS1 3NU, UK
| | - Andrew Parry
- Bristol Heart Institute, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol BS1 3NU, UK
| | - Umberto Benedetto
- Bristol Heart Institute, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol BS1 3NU, UK
| | - Pierpaolo Chivasso
- Bristol Heart Institute, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol BS1 3NU, UK
| | - Frances Gill
- Bristol Heart Institute, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol BS1 3NU, UK
| | - Mark C. K. Hamilton
- Department of Clinical Radiology, University Hospitals Bristol, Bristol Royal Infirmary, Bristol BS2 8EJ, UK
| | - Chiara Bucciarelli-Ducci
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol BS8 1TH, UK
- Bristol Heart Institute, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol BS1 3NU, UK
- Royal Brompton & Harefield Hospitals, Guy’s and St Thomas’ NHS Foundation Trust, London SW3 6NP, UK
| | - Massimo Caputo
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol BS8 1TH, UK
- Bristol Heart Institute, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol BS1 3NU, UK
| | - Costanza Emanueli
- National Heart and Lung Institute, Imperial College London, London SW7 2BX, UK
| | - Giovanni Biglino
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol BS8 1TH, UK
- National Heart and Lung Institute, Imperial College London, London SW7 2BX, UK
- Correspondence: ; Tel.: +44-117-342-3287
| |
Collapse
|
23
|
Type I Interferon Receptor Subunit 1 Deletion Attenuates Experimental Abdominal Aortic Aneurysm Formation. Biomolecules 2022; 12:biom12101541. [PMID: 36291750 PMCID: PMC9599283 DOI: 10.3390/biom12101541] [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: 09/07/2022] [Revised: 10/14/2022] [Accepted: 10/17/2022] [Indexed: 11/16/2022] Open
Abstract
Objective: Type I interferon receptor signaling contributes to several autoimmune and vascular diseases such as lupus, atherosclerosis and stroke. The purpose of this study was to assess the influence of type I interferon receptor deficiency on the formation and progression of experimental abdominal aortic aneurysms (AAAs). Methods: AAAs were induced in type I interferon receptor subunit 1 (IFNAR1)-deficient and wild type control male mice via intra-infrarenal aortic infusion of porcine pancreatic elastase. Immunostaining for IFNAR1 was evaluated in experimental and clinical aneurysmal abdominal aortae. The initiation and progression of experimental AAAs were assessed via ultrasound imaging prior to (day 0) and days 3, 7 and 14 following elastase infusion. Aneurysmal histopathology was analyzed at sacrifice. Results: Increased aortic medial and adventitial IFNAR1 expression was present in both clinical AAAs harvested at surgery and experimental AAAs. Following AAA induction, wild type mice experienced progressive, time-dependent infrarenal aortic enlargement. This progression was substantially attenuated in IFNAR1-deficient mice. On histological analyses, medial elastin degradation, smooth muscle cell depletion, leukocyte accumulation and neoangiogenesis were markedly diminished in IFNAR1-deficient mice in comparison to wild type mice. Conclusion: IFNAR1 deficiency limited experimental AAA progression in response to intra-aortic elastase infusion. Combined with clinical observations, these results suggest an important role for IFNAR1 activity in AAA pathogenesis.
Collapse
|
24
|
Sikorski V, Vento A, Kankuri E. Emerging roles of the RNA modifications N6-methyladenosine and adenosine-to-inosine in cardiovascular diseases. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 29:426-461. [PMID: 35991314 PMCID: PMC9366019 DOI: 10.1016/j.omtn.2022.07.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Cardiovascular diseases lead the mortality and morbidity disease metrics worldwide. A multitude of chemical base modifications in ribonucleic acids (RNAs) have been linked with key events of cardiovascular diseases and metabolic disorders. Named either RNA epigenetics or epitranscriptomics, the post-transcriptional RNA modifications, their regulatory pathways, components, and downstream effects substantially contribute to the ways our genetic code is interpreted. Here we review the accumulated discoveries to date regarding the roles of the two most common epitranscriptomic modifications, N6-methyl-adenosine (m6A) and adenosine-to-inosine (A-to-I) editing, in cardiovascular disease.
Collapse
Affiliation(s)
- Vilbert Sikorski
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Antti Vento
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland
| | - Esko Kankuri
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - IHD-EPITRAN Consortium
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland
| |
Collapse
|
25
|
Becker von Rose A, Kobus K, Bohmann B, Lindquist-Lilljequist M, Eilenberg W, Bassermann F, Reeps C, Eckstein HH, Trenner M, Maegdefessel L, Neumayer C, Brostjan C, Roy J, Hultgren R, Schwaiger BJ, Busch A. Radiation and chemotherapeutics are associated with altered aortic aneurysm growth in cancer patients. Eur J Vasc Endovasc Surg 2022; 64:255-264. [PMID: 35853577 DOI: 10.1016/j.ejvs.2022.07.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 06/26/2022] [Accepted: 07/10/2022] [Indexed: 11/26/2022]
Abstract
INTRODUCTION Co-prevalence of aorto-iliac aneurysm (i.e. AAA) and cancer confronts patients and physicians with two life-threatening diseases. Modern chemotherapeutics and target therapies might impact the aneurysm wall integrity and subsequently affect growth. The purpose of this study was to assess associations between malignancy, therapeutic regimens and the growth rates of aorto-iliac aneurysms. PATIENTS AND METHODS A retrospective single-center analysis identified patients with aneurysm + cancer. Patients with ≥2 CT angiograms over ≥6 months and additional malignancy were included. Clinical data and aneurysm diameters were analyzed. AAA growth under cancer therapy (chemotherapy/radiation) was compared to a non-cancer AAA control cohort and to meta-analysis data. Statistics included t-tests and a linear regression model with correction for initial aortic diameter and type of treatment. RESULTS From 2003 - 2020, 217 patients (median age 70 years; 92% male) with 246 aneurysms (58.8% AAA) and 238 malignancies were identified. Prostate (27%) and lung (16%) cancer were most frequently seen, 157 patients (72%) received chemotherapy and 105 patients (48%) radiation, thereof 79 (36.4%) both. Annual AAA growth was not significantly different for cancer and non-cancer patients (2.0±2.3 vs. 2.8±2.1mm/y, p=0.20). However, subgroup analyses revealed that radiation was associated with a significantly reduced aneurysm growth rate compared to cancer patients without radiation (1.1±1.3 vs. 1.6±2.1 mm/y, p=0.046) and to the non-cancer control cohort (1.7±1.9 vs. 2.8±2.1 mm/y, p=0.007). Administration of antimetabolites showed significantly increased AAA growth (+0.9mm/year, p=0.011), while e.g. topoisomerase inhibitors (-0.8mm/year, p=0.17) and anti-androgens (-0.5mm/year, p=0.27) showed a possible trend for reduced growth. Similar was observed for iliac aneurysms (n=85). Additionally, effects were persistent in combinations of chemotherapies (2.6±1.4 substances/patient). CONCLUSION Cancer patients with concomitant aortic aneurysms may require intensified monitoring when undergoing specific therapies, such as antimetabolites, since they may experience an increased aneurysm growth rate. Radiation may be associated with reduced aneurysm growth.
Collapse
Affiliation(s)
- Aaron Becker von Rose
- III. Medical Department for Hematology and Oncology, Klinikum rechts der Isar Technical University Munich, Munich, Germany
| | - Kathrin Kobus
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Bianca Bohmann
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Moritz Lindquist-Lilljequist
- Stockholm Aneurysm Research Group (STAR), Department of Vascular Surgery, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Wolf Eilenberg
- Division of Vascular Surgery, Department of General Surgery, Medical University of Vienna
| | - Florian Bassermann
- III. Medical Department for Hematology and Oncology, Klinikum rechts der Isar Technical University Munich, Munich, Germany
| | - Christian Reeps
- Division of Vascular and Endovascular Surgery, Department for Visceral-, Thoracic and Vascular Surgery, Medical Faculty Carl Gustav Carus and University Hospital, Technische Universität Dresden, Dresden, Germany
| | - Hans-Henning Eckstein
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Matthias Trenner
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, Munich, Germany; Division of Vascular Medicine, St. Josefs-Hospital Wiesbaden, Wiesbaden, Germany
| | - Lars Maegdefessel
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Christoph Neumayer
- Division of Vascular Surgery, Department of General Surgery, Medical University of Vienna
| | - Christine Brostjan
- Division of Vascular Surgery, Department of General Surgery, Medical University of Vienna
| | - Joy Roy
- Stockholm Aneurysm Research Group (STAR), Department of Vascular Surgery, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Rebecka Hultgren
- Stockholm Aneurysm Research Group (STAR), Department of Vascular Surgery, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Benedikt J Schwaiger
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Albert Busch
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, Munich, Germany; Division of Vascular and Endovascular Surgery, Department for Visceral-, Thoracic and Vascular Surgery, Medical Faculty Carl Gustav Carus and University Hospital, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
26
|
Between the devil and the deep blue sea: new insights of cancer therapies effects on abdominal aortic aneurysm growth. Eur J Vasc Endovasc Surg 2022; 64:265. [PMID: 35697235 DOI: 10.1016/j.ejvs.2022.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 06/05/2022] [Indexed: 11/23/2022]
|
27
|
Zhu JX, Tang QQ, Zhou C, Shi XC, Yi SY, Yang Y. Establishment of a New Abdominal Aortic Aneurysm Model in Rats by a Retroperitoneal Approach. Front Cardiovasc Med 2022; 9:808732. [PMID: 35282381 PMCID: PMC8905142 DOI: 10.3389/fcvm.2022.808732] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 01/17/2022] [Indexed: 12/17/2022] Open
Abstract
Background Constructing an ideal model of abdominal aortic aneurysm (AAA) is of great significance to elucidate its complex pathogenesis. Therefore, we introduce a new and simple method to simulate human AAA and construct a rat AAA model through a retroperitoneal approach. Methods Forty healthy adult Sprague Dawley (SD) rats were randomly divided into a control group, elastase + calcium chloride group (PPE+CaCl2), elastase group (PPE), and elastase + beta aminopropionitrile group (PPE+BAPN) according to a male-female ratio of 1:1, with 10 rats in each group. A retroperitoneal approach was used to free the infrarenal abdominal aorta in all four groups. In the PPE + CaCl2 group, 0.1 ml of elastase (approximately 5 U) was perfused into the arterial cavity for 20 min, and 1.0 mol/L calcium chloride was infiltrated out of the arterial cavity for 10 min. In the PPE group, 0.1 mL of elastase (approximately 5U) was perfused into the arterial cavity for 20 min, and normal saline was infiltrated out of arterial cavity for 10 min; the PPE + BAPN group combined with 0.3% BAPN drinking water/day on the basis of PPE group; the control group was treated with saline instead of elastase and calcium chloride. Abdominal aortic specimens were collected after 4 weeks of feeding. The diagnostic criteria of AAA were 50% dilation of the abdominal aorta or rupture of the aneurysm at 4 weeks after the operation. Histopathology, immunohistochemistry, quantitative PCR (qPCR), western blotting assay, gelatine zymogram, and other methods were used. Results The operation time of the four groups was controlled at approximately 40 min, and the success rate of the operation was 100%. Survival rate: Control Group (100%) = PPE Group (100%) > PPE + CaCl2 Group (90%) > PPE + BAPN Group (40%); Aneurysm formation rate: PPE + BAPN Group (100%) > PPE + CaCl2 Group (80%) > PPE Group (60%) > Control Group (0%); Aneurysm rupture rate: PPE + BAPN group (60%) > PPE + CaCl2 group (12.5%) > PPE group (0%);Inflammatory cells (macrophages, T cells, B cells, dendritic cells) infiltrated in different degrees in the PPE + CaCl2, PPE and PPE + BAPN groups. Vascular thickness, elastic fiber content, collagen fiber content, and vascular smooth muscle cell content in the PPE + CaCl2 group and PPE + BNPA group were significantly lower than those in Control group (P < 0.05). The content of elastic fibers and vascular smooth muscle cells in the PPE group were significantly lower than that in Control group (P < 0.05). The expression and activity of matrix metalloproteinase 2 (MMP2) and MMP9 in the PPE + CaCl2 group, PPE group, and PPE + BNPA group were significantly higher than those in the control group (P < 0.05). Conclusions A new, simple, and reproducible rat AAA model can be constructed by a retroperitoneal approach. The pathological features of the three models are effective simulation of human AAA inflammatory cell infiltration, protease activity enhancement, and extracellular matrix destruction. The PPE+ CaCl2 model has the advantages of a high survival rate, high aneurysm formation rate, good stability, and reproducibility. It is an ideal animal model for studying the pathogenesis of AAA. The PPE + BAPN model can simulate the characteristics of spontaneous rupture of aneurysms. It is an ideal animal model to study the mechanism of AAA rupture.
Collapse
Affiliation(s)
- Jun-Xing Zhu
- Department of Clinical Medicine, North Sichuang Medical College, Nanchong, China
- Department of Cardiovascular Disease, Affiliated Hospital, North Sichuang Medical College, Nanchong, China
| | - Quan-Qiao Tang
- Department of Clinical Medicine, North Sichuang Medical College, Nanchong, China
- Department of Cardiovascular Disease, Affiliated Hospital, North Sichuang Medical College, Nanchong, China
| | - Can Zhou
- Department of Clinical Medicine, North Sichuang Medical College, Nanchong, China
- Department of Cardiovascular Disease, Affiliated Hospital, North Sichuang Medical College, Nanchong, China
| | - Xing-Chi Shi
- Department of Clinical Medicine, North Sichuang Medical College, Nanchong, China
- Department of Cardiovascular Disease, Affiliated Hospital, North Sichuang Medical College, Nanchong, China
| | - Si-Yi Yi
- Department of Clinical Medicine, North Sichuang Medical College, Nanchong, China
- Department of Cardiovascular Disease, Affiliated Hospital, North Sichuang Medical College, Nanchong, China
| | - Ying Yang
- Department of Clinical Medicine, North Sichuang Medical College, Nanchong, China
- Department of Cardiovascular Disease, Affiliated Hospital, North Sichuang Medical College, Nanchong, China
- *Correspondence: Ying Yang
| |
Collapse
|
28
|
Imaging Techniques for Aortic Aneurysms and Dissections in Mice: Comparisons of Ex Vivo, In Situ, and Ultrasound Approaches. Biomolecules 2022; 12:biom12020339. [PMID: 35204838 PMCID: PMC8869425 DOI: 10.3390/biom12020339] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/14/2022] [Accepted: 02/16/2022] [Indexed: 01/04/2023] Open
Abstract
Aortic aneurysms and dissections are life-threatening conditions that have a high risk for lethal bleeding and organ malperfusion. Many studies have investigated the molecular basis of these diseases using mouse models. In mice, ex vivo, in situ, and ultrasound imaging are major approaches to evaluate aortic diameters, a common parameter to determine the severity of aortic aneurysms. However, accurate evaluations of aortic dimensions by these imaging approaches could be challenging due to pathological features of aortic aneurysms. Currently, there is no standardized mode to assess aortic dissections in mice. It is important to understand the characteristics of each approach for reliable evaluation of aortic dilatations. In this review, we summarize imaging techniques used for aortic visualization in recent mouse studies and discuss their pros and cons. We also provide suggestions to facilitate the visualization of mouse aortas.
Collapse
|
29
|
Liu H, Tian K, Xia C, Wei P, Xu B, Fu W, Li Y, Li Y, Bai L, Wang R, Wang W, Xu B, Liu E, Zhao S. Kunming mouse strain is less susceptible to elastase-induced abdominal aortic aneurysms. Animal Model Exp Med 2022; 5:72-80. [PMID: 35229996 PMCID: PMC8879628 DOI: 10.1002/ame2.12197] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 11/04/2021] [Accepted: 11/22/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Porcine pancreatic elastase (PPE) is successfully used to induce abdominal aortic aneurysm (AAA) in mice. However, differences between mouse strains in susceptibility to PPE induction have been reported. Kunming mouse is one of the most frequently used strains in China but whether it is suitable for induction of AAA by PPE application remains unclear. METHODS PPE infusion (1.5 units/ml) in temporary controlled aorta was performed to induce AAAs in both C57BL/6J and Kunming mice. Phosphate-buffered saline (PBS) application was used as vehicle control. The aorta diameters of all mice were measured at days 0 and 14 after surgery to evaluate the AAA formation. RESULTS After 14 days of PPE or PBS infusion, all mice were sacrificed and aorta tissues were collected for histological staining analysis. At the 14th day after infusion, PPE successfully induced aortic dilation in Kunming mice and typical AAA in C57BL/6J mice. The aorta diameter increased by 0.23 mm in Kunming mice after PPE infusion, while it was 0.72 mm in the C57BL/6J strain. PPE induced mild elastin degradation, smooth muscle cell (SMC) depletion and mural leucocyte infiltration in Kunming mice, but in PPE-sensitive C57BL/6J mice, it induced total loss of SMCs, elastin disappearance and diffused infiltrated leucocytes in aortic aneurysmal segments. The effects of PPE in inducing angiogenesis and upregulating matrix metalloproteinase 2 and 9 expression in Kunming mice were also weaker than that in C57BL/6J mice. CONCLUSION At the reported dose of PPE, Kunming mouse is not as susceptible to AAA formation as C57BL/6J mice. The failure of PPE to induce AAA formation in Kunming mice may be associated to its inability to boost a strong inflammatory response.
Collapse
Affiliation(s)
- Haole Liu
- Institute of Cardiovascular ScienceSchool of Basic Medical SciencesXi'an Jiaotong University Health Science CenterXi'anChina
| | - Kangli Tian
- Laboratory Animal CenterXi'an Jiaotong UniversityXi'anChina
| | - Congcong Xia
- Laboratory Animal CenterXi'an Jiaotong UniversityXi'anChina
| | - Panpan Wei
- Laboratory Animal CenterXi'an Jiaotong UniversityXi'anChina
| | - Boyu Xu
- Laboratory Animal CenterXi'an Jiaotong UniversityXi'anChina
| | - Weilai Fu
- Department of Vascular SurgeryThe Second Hospital of Tianjin Medical UniversityTianjinChina
| | - Yankui Li
- Department of Vascular SurgeryThe Second Hospital of Tianjin Medical UniversityTianjinChina
| | - Yafeng Li
- Pain Rehabilitation Department of TCM Orthopedic CenterXi'an Honghui HospitalXi'anChina
| | - Liang Bai
- Institute of Cardiovascular ScienceSchool of Basic Medical SciencesXi'an Jiaotong University Health Science CenterXi'anChina
| | - Rong Wang
- Institute of Cardiovascular ScienceSchool of Basic Medical SciencesXi'an Jiaotong University Health Science CenterXi'anChina
| | - Weirong Wang
- Institute of Cardiovascular ScienceSchool of Basic Medical SciencesXi'an Jiaotong University Health Science CenterXi'anChina
| | - Baohui Xu
- Department of Vascular SurgeryStanford University School of MedicineStanfordCaliforniaUSA
| | - Enqi Liu
- Institute of Cardiovascular ScienceSchool of Basic Medical SciencesXi'an Jiaotong University Health Science CenterXi'anChina
- Laboratory Animal CenterXi'an Jiaotong UniversityXi'anChina
| | - Sihai Zhao
- Institute of Cardiovascular ScienceSchool of Basic Medical SciencesXi'an Jiaotong University Health Science CenterXi'anChina
- Laboratory Animal CenterXi'an Jiaotong UniversityXi'anChina
| |
Collapse
|
30
|
Pan T, Lu G, Ge L, Jiang Y, Wan H, Xu S, Zhang X. A low-grade cerebral arteriovenous malformation suspected of being a metastatic tumor: A case report and literature review. J Interv Med 2022; 5:40-45. [PMID: 35586284 PMCID: PMC8947987 DOI: 10.1016/j.jimed.2021.12.010] [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: 11/09/2021] [Revised: 12/29/2021] [Accepted: 12/30/2021] [Indexed: 11/23/2022] Open
Abstract
Cases of low-grade cerebral arteriovenous malformations (cAVMs) showing dynamic changes and large areas of brain edema on short-term MRI follow-up have rarely been reported. This report describes an incidentally discovered and initially misdiagnosed cAVM in a patient with malignancies. The presence of abnormal signals surrounded by large areas of brain edema combined with tortuous or dilated vessels indicates the possibility of an AVM, especially in young people.
Collapse
Affiliation(s)
- Ting Pan
- Department of Radiology, Affiliated Huashan Hospital, Fudan University, Shanghai, China
| | - Gang Lu
- Department of Radiology, Affiliated Huashan Hospital, Fudan University, Shanghai, China
| | - Liang Ge
- Department of Radiology, Affiliated Huashan Hospital, Fudan University, Shanghai, China
| | - Yeqing Jiang
- Department of Radiology, Affiliated Huashan Hospital, Fudan University, Shanghai, China
| | - Hailin Wan
- Department of Radiology, Affiliated Huashan Hospital, Fudan University, Shanghai, China
| | - Shu Xu
- Department of Radiology, Affiliated Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaolong Zhang
- Department of Radiology, Affiliated Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
31
|
Liu H, Wei P, Fu W, Xia C, Li Y, Tian K, Li Y, Cheng D, Sun J, Xu Y, Lu M, Xu B, Zhang Y, Wang R, Wang W, Xu B, Liu E, Zhao S. Dapagliflozin Ameliorates the Formation and Progression of Experimental Abdominal Aortic Aneurysms by Reducing Aortic Inflammation in Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8502059. [PMID: 35126822 PMCID: PMC8816542 DOI: 10.1155/2022/8502059] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 01/03/2022] [Accepted: 01/10/2022] [Indexed: 02/08/2023]
Abstract
BACKGROUND Dapagliflozin, a sodium glucose transporter protein-2 (SGLT-2) inhibitor, reduces the risk for cardiovascular diseases. However, the influence of dapagliflozin on nondissecting abdominal aortic aneurysms (AAAs) remains unclear. METHODS AAAs were created in male C57BL/6 mice via intra-aortic porcine pancreatic elastase (PPE) infusion. Mice were daily treated with dapagliflozin (1 or 5 mg/kg body weight) or an equal volume of vehicle through oral gavage beginning one day prior to PPE infusion for 14 days. To investigate its translational value, dapagliflozin or vehicle was also administered to mice with existing AAAs in another cohort. Aortic diameters were measured prior to (day 0 for baseline) and 14 days after PPE infusion. After sacrifice, mice aortae were collected, and following histological analyses were performed. RESULTS Dapagliflozin treatment significantly reduced aneurysmal aortic expansion following PPE infusion as compared to vehicle treatment especially at 5 mg/kg body weight (approximately 21% and 33% decreases in 1 and 5 mg/kg treatment groups, respectively). The dose-dependent attenuation of AAAs by dapagliflozin was also confirmed on histological analyses. Dapagliflozin remarkably reduced aortic accumulation of macrophages, CD4+ T cells, and B cells particularly following dapagliflozin treatment at 5 mg/kg. Dapagliflozin treatment also markedly attenuated medial SMC loss. Though the difference was not significant, dapagliflozin treatment tended to attenuate CD8+ T cells and elastin degradation. Dapagliflozin treatment at 5 mg/kg caused a 53% reduction in neovessel density. Furthermore, dapagliflozin treatment mitigated further progress of existing AAAs. CONCLUSION Dapagliflozin treatment ameliorated PPE-induced AAAs by inhibiting aortic leukocytes infiltration and angiogenesis.
Collapse
Affiliation(s)
- Haole Liu
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
- Laboratory Animal Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Panpan Wei
- Laboratory Animal Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Weilai Fu
- Department of Vascular Surgery, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Congcong Xia
- Laboratory Animal Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Yankui Li
- Department of Vascular Surgery, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Kangli Tian
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Yafeng Li
- Laboratory Animal Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Daxin Cheng
- Laboratory Animal Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Jiaying Sun
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Yangwei Xu
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Ming Lu
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Boyu Xu
- Laboratory Animal Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Yali Zhang
- Laboratory Animal Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Rong Wang
- Laboratory Animal Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Weirong Wang
- Laboratory Animal Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Baohui Xu
- Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Enqi Liu
- Laboratory Animal Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Sihai Zhao
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
- Laboratory Animal Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| |
Collapse
|
32
|
Guo J, Shoji T, Ge Y, Zheng X, Li Y, Zhao S, Ikezoe T, Liu S, Huang J, Wang W, Xu B, Dalman RL. Treatment with the Prolyl Hydroxylase Inhibitor JNJ Promotes Abdominal Aortic Aneurysm Progression in Diabetic Mice. Eur J Vasc Endovasc Surg 2021; 63:484-494. [PMID: 34872812 DOI: 10.1016/j.ejvs.2021.10.030] [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: 10/12/2020] [Revised: 09/21/2021] [Accepted: 10/09/2021] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Prolyl hydroxylase domain containing proteins (PHD) rigorously regulate intracellular hypoxia inducible factor-1 (HIF-1) protein expression and activity. Diabetes impairs PHD activity and attenuates abdominal aortic aneurysm (AAA) progression. The extent to which dysregulated PHD activity contributes to diabetes mediated AAA suppression remains undetermined. METHODS AAAs were induced in diabetic and non-diabetic male C57BL/6J mice via intra-aortic elastase infusion. A PHD inhibitor (JNJ-42041935, aka "JNJ", 150 mmol/kg) or vehicle alone was administered daily starting one day prior to AAA induction for 14 days. Influences on AAA progression was assessed via ultrasonography and histopathology. Expression of aortic HIF-1α, three of its target genes and macrophage derived mediators were assayed via quantitative reverse transcription polymerase chain reaction. Aneurysmal sections from AAA patients with and without diabetes (two patients in each group) were immunostained for HIF-1α and vascular endothelial growth factor (VEGF)-A. RESULTS Expression of HIF-1α target genes (erythropoietin, VEGF-A, and glucose transporter-1) was reduced by 45% - 95% in experimental diabetic aortas. Diameter enlargement was similarly limited, as were mural elastin degradation, leukocyte infiltration, and neo-angiogenesis (reduced capillary density and length) on histopathology. Pre-treatment with JNJ prior to AAA initiation augmented aortic HIF-1α target gene expression and aneurysm progression in diabetic mice, along with macrophage VEGF-A and matrix metalloproteinase 2 mRNA expression. No differences were noted in HIF-1α or VEGF-A expression on aortic immunohistochemical staining of human aortic tissue as a function of diabetes status. CONCLUSION Small molecule PHD inhibitor treatment reduces or offsets impairment of experimental AAA progression in hyperglycemic mice, highlighting the potential contribution of dysregulated PHD activity to diabetes mediated aneurysm suppression.
Collapse
Affiliation(s)
- Jia Guo
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA; Centre for Hypertension Care, Shanxi Medical University First Hospital, Taiyuan, Shanxi Province, P. R. China
| | - Tahakiro Shoji
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA; Department of Emergency Medicine, Saiseikai Central Hospital, Minatoku, Tokyo, Japan
| | - Yingbin Ge
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu Province, P. R. China
| | - Xiaoya Zheng
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Yankui Li
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Sihai Zhao
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Toru Ikezoe
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Shuai Liu
- Department of Vascular Surgery, Central South University School of Medicine, Changsha, Hunan Province, P. R. China
| | - Jianhua Huang
- Department of Vascular Surgery, Central South University School of Medicine, Changsha, Hunan Province, P. R. China
| | - Wei Wang
- Department of Vascular Surgery, Central South University School of Medicine, Changsha, Hunan Province, P. R. China
| | - Baohui Xu
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA.
| | - Ronald L Dalman
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
33
|
Temporal and Quantitative Analysis of Aortic Immunopathologies in Elastase-Induced Mouse Abdominal Aortic Aneurysms. J Immunol Res 2021; 2021:6297332. [PMID: 34825008 PMCID: PMC8610647 DOI: 10.1155/2021/6297332] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 11/02/2021] [Indexed: 02/06/2023] Open
Abstract
Objective Elastase-induced abdominal aortic aneurysm (AAA) model is widely used for aneurysmal pathogenesis and translational research. However, temporal alternations in aneurysmal histologies remain unknown. This study is aimed at analyzing temporal immunopathologies of aneurysmal aorta following experimental AAA induction. Methods Male C57BL/6J mice at the age of 10-14 weeks received intra-aortic infusion of elastase to induce AAAs. Aortic diameters at the baseline and indicated days after AAA induction were measured, and aortae were collected for histopathological analysis. Results Aorta diameters increased from 0.52 mm at the baseline levels to 0.99 mm, 1.34 mm, and 1.41 mm at days 7, 14, and 28, respectively, corresponding 90%, 158%, and 171% increases over the baseline level. Average aortic diameters did not differ between days 14 and 28. Severe elastin degradation and smooth muscle cell depletion were found at days 14 and 28 as compared to the baseline and day 7. No difference in the scores of medial elastin and SMC destruction was noted between days 14 and 28. Consistent results were found for leukocyte accumulation, neoangiogenesis, and matrix metalloproteinase expression. Twenty-eight days after AAA induction, all aneurysmal pathologies showed an attenuated trend, although most histopathological parameters did no differ between days 14 and 28. Conclusion Our data suggest that almost aneurysmal immunohistopathologies reach maximal 14 days following AAA induction. Analysis of day 14 histologies is sufficient for AAA pathogenesis and translational studies in elastase-induced mouse experimental AAAs.
Collapse
|
34
|
Nicotine Exacerbates TAAD Formation Induced by Smooth Muscle-Specific Deletion of the TGF- β Receptor 2. J Immunol Res 2021; 2021:6880036. [PMID: 34646889 PMCID: PMC8505064 DOI: 10.1155/2021/6880036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 09/04/2021] [Indexed: 01/22/2023] Open
Abstract
Tobacco smoke is an established risk factor for thoracic aortic aneurysms and dissections (TAAD). However, little is known about its underlying mechanisms due to the lack of validated animal models. The present study developed a mouse model that may be utilized to investigate exacerbation of TAAD formation by mimetics of tobacco smoke. TAADs were created via inducible deletion of smooth muscle cell-specific Tgfbr2 receptors. Using this model, the first set of experiments evaluated the efficacy of nicotine salt (34.0 mg/kg/day), nicotine free base (NFB, 5.0 mg 90-day pellets), and cigarette smoke extract (0.1 ml/mouse/day). Compared with their respective control groups, only NFB pellets promoted TAAD dilation (23 ± 3% vs. 12 ± 2%, P = 0.014), and this efficacy was achieved at a cost of >50% acute mortality. Infusion of NFB with osmotic minipumps at extremely high, but nonlethal, doses (15.0 or 45.0 mg/kg/day) failed to accelerate TAAD dilation. Interestingly, costimulation with β-aminopropionitrile (BAPN) promoted TAAD dilation and aortic rupture at dosages of 3.0 and 45.0 mg/kg/day, respectively, indicating that BAPN sensitizes the response of TAADs to NFB. In subsequent analyses, the detrimental effects of NFB were associated with clustering of macrophages, neutrophils, and T-cells in areas with structural destruction, enhanced matrix metalloproteinase- (MMP-) 2 production, and pathological angiogenesis with attenuated fibrosis in the adventitia. In conclusion, modeling nicotine exacerbation of TAAD formation requires optimization of chemical form, route of delivery, and dosage of the drug as well as the pathologic complexity of TAADs. Under the optimized conditions of the present study, chronic inflammation and adventitial mal-remodeling serve as critical pathways through which NFB exacerbates TAAD formation.
Collapse
|
35
|
Sun P, Zhang L, Gu Y, Wei S, Wang Z, Li M, Wang W, Wang Z, Bai H. Immune checkpoint programmed death-1 mediates abdominal aortic aneurysm and pseudoaneurysm progression. Biomed Pharmacother 2021; 142:111955. [PMID: 34339918 DOI: 10.1016/j.biopha.2021.111955] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 07/18/2021] [Accepted: 07/20/2021] [Indexed: 12/20/2022] Open
Abstract
PURPOSE The causes and pathogenetic mechanisms underlying abdominal aortic aneurysms (AAAs) and pseudoaneurysms are not fully understood. We hypothesized that inhibiting programmed death-1 (PD-1) can decrease AAA and pseudoaneurysm formation in mouse and rat models. METHODS Human AAA samples were examined in conjunction with an adventitial calcium chloride (CaCl2) application mouse model and an aortic patch angioplasty rat model. Single-dose PD-1 antibody (4 mg/kg) or BMS-1 (PD-1 inhibitor-1) (1 mg/kg) was administered by intraperitoneal (IP) or intraluminal injection. In the intramural injection group, PD-1 antibody was injected after CaCl2 incubation. The rats were divided into three groups: (1) the control group was only decellularized without other special treatment, (2) the PD-1 antibody-coated patch group, and (3) the BMS-1 coated patch group. Patches implanted in the rat abdominal aorta were harvested on day 14 after implantation and analyzed. RESULTS Immunohistochemical analysis showed PD-1-positive cells, PD-1 and CD3, PD-1 and CD68, and PD-1 and α-actin co-expressed in the human AAA samples. Intraperitoneal (IP) injection or intraluminal injection of PD-1antibody/BMS-1 significantly inhibited AAA progression. PD-1 antibody and BMS-1 were each successfully conjugated to decellularized rat thoracic artery patches, respectively, by hyaluronic acid. Patches coated with either humanized PD-1 antibody or BMS-1 can also inhibit pseudoaneurysm progression and inflammatory cell infiltration. CONCLUSION PD-1 pathway inhibition may be a promising therapeutic strategy for inhibiting AAA and pseudoaneurysm progression.
Collapse
MESH Headings
- Aneurysm, False/drug therapy
- Aneurysm, False/metabolism
- Aneurysm, False/pathology
- Angioplasty/methods
- Animals
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibodies, Monoclonal, Humanized/therapeutic use
- Aortic Aneurysm, Abdominal/drug therapy
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/pathology
- B7-H1 Antigen/antagonists & inhibitors
- B7-H1 Antigen/metabolism
- Calcium Chloride/toxicity
- Coated Materials, Biocompatible/pharmacology
- Coated Materials, Biocompatible/therapeutic use
- Disease Models, Animal
- Disease Progression
- Humans
- Immune Checkpoint Inhibitors/pharmacology
- Immune Checkpoint Inhibitors/therapeutic use
- Injections, Intraperitoneal
- Lymphocytes/immunology
- Macrophages/immunology
- Male
- Mice
- Programmed Cell Death 1 Receptor/antagonists & inhibitors
- Programmed Cell Death 1 Receptor/immunology
- Programmed Cell Death 1 Receptor/metabolism
- Rats, Sprague-Dawley
- Rats
Collapse
Affiliation(s)
- Peng Sun
- Department of Vascular and Endovascular Surgery, First Affiliated Hospital of Zhengzhou University, 450052 Henan, China
| | - Liwei Zhang
- Department of Vascular and Endovascular Surgery, First Affiliated Hospital of Zhengzhou University, 450052 Henan, China
| | - Yulei Gu
- Emergency Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, 450052 Henan, China
| | - Shunbo Wei
- Department of Vascular and Endovascular Surgery, First Affiliated Hospital of Zhengzhou University, 450052 Henan, China
| | - Zhiwei Wang
- Department of Vascular and Endovascular Surgery, First Affiliated Hospital of Zhengzhou University, 450052 Henan, China
| | - Mingxing Li
- Department of Vascular and Endovascular Surgery, First Affiliated Hospital of Zhengzhou University, 450052 Henan, China
| | - Wang Wang
- Key Vascular Physiology and Applied Research Laboratory of Zhengzhou City, 450002 Henan, China; Department of Physiology, Medical School of Zhengzhou University, 450002 Henan, China
| | - Zhiju Wang
- Department of Physiology, Medical School of Zhengzhou University, 450002 Henan, China
| | - Hualong Bai
- Department of Vascular and Endovascular Surgery, First Affiliated Hospital of Zhengzhou University, 450052 Henan, China; Key Vascular Physiology and Applied Research Laboratory of Zhengzhou City, 450002 Henan, China.
| |
Collapse
|
36
|
Ji L, Chen S, Gu G, Wang W, Ren J, Xu F, Li F, Wu J, Yang D, Zheng Y. Discovery of potential biomarkers for human atherosclerotic abdominal aortic aneurysm through untargeted metabolomics and transcriptomics. J Zhejiang Univ Sci B 2021; 22:733-745. [PMID: 34514753 DOI: 10.1631/jzus.b2000713] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Abdominal aortic aneurysm (AAA) and atherosclerosis (AS) have considerable similarities in clinical risk factors and molecular pathogenesis. The aim of our study was to investigate the differences between AAA and AS from the perspective of metabolomics, and to explore the potential mechanisms of differential metabolites via integration analysis with transcriptomics. Plasma samples from 32 AAA and 32 AS patients were applied to characterize the metabolite profiles using untargeted liquid chromatography-mass spectrometry (LC-MS). A total of 18 remarkably different metabolites were identified, and a combination of seven metabolites could potentially serve as a biomarker to distinguish AAA and AS, with an area under the curve (AUC) of 0.93. Subsequently, we analyzed both the metabolomics and transcriptomics data and found that seven metabolites, especially 2'-deoxy-D-ribose (2dDR), were significantly correlated with differentially expressed genes. In conclusion, our study presents a comprehensive landscape of plasma metabolites in AAA and AS patients, and provides a research direction for pathogenetic mechanisms in atherosclerotic AAA.
Collapse
Affiliation(s)
- Lei Ji
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Siliang Chen
- School of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Guangchao Gu
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Wei Wang
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Jinrui Ren
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Fang Xu
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Fangda Li
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Jianqiang Wu
- Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Dan Yang
- Department of Computational Biology and Bioinformatics, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Yuehong Zheng
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China.
| |
Collapse
|
37
|
Li T, Wang T, Jing J, Sun L. Expression Pattern and Clinical Value of Key m6A RNA Modification Regulators in Abdominal Aortic Aneurysm. J Inflamm Res 2021; 14:4245-4258. [PMID: 34511965 PMCID: PMC8412829 DOI: 10.2147/jir.s327152] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/19/2021] [Indexed: 12/17/2022] Open
Abstract
Background Aberrant expression of N6-methyladenosine (m6A) RNA modification regulators plays a critical role in a variety of human diseases. However, their implication in abdominal aortic aneurysm (AAA) remains largely unknown. Herein, we sought to explore the general expression pattern and potential functions of m6A regulators in AAA. Methods We analyzed gene expression data of m6A regulators in human AAA and normal tissues from public GEO database. The R package and other tools such as m6A2Target database, Gene ontology (GO) functional and Kyoto encyclopedia of genes and genomes (KEGG) pathway analyses, gene set variation analysis (GSVA), Search Tool for the Retrieval of Interacting Genes (STRING), starBase, miRDB and Cytoscape software were applied for bioinformatics analysis to investigate the downstream molecular mechanisms and upstream regulatory mechanisms for distinctly expressed regulators. Quantitative real-time PCR (qRT-PCR) and enzyme-linked immunosorbent assay (ELISA) were performed to validate the expression of key m6A regulators in our collected human AAA specimens. Results We found that METTL14 and HNRNPC were the downregulated m6A regulators, and RBM15B was the upregulated methylation transferase in human AAA. The modified genes were primarily enriched in RNA catabolic process, regulation of translation, focal adhesion, transcription coregulator activity, ribosome, RNA transport, cell cycle, et al. METTL14, HNRNPC and RBM15B levels were correlated with the immune infiltration degree of Tcm, macrophages, mast cells, Tgd and NK CD56bright cells. A total of 154 and 76 target genes of three regulators were separately involved in body metabolism and autophagy in AAA disease, and their interactive relationships and hub genes were identified. The lncRNA-miRNA-mRNA interaction regulatory networks were also constructed for METTL14, HNRNPC and RBM15B. Based on our clinical tissue and serum samples, METTL14 exhibited lower expression levels in AAA and its rupture type, and low METTL14 expression was associated with high levels of WBC and CRP (all P < 0.05). Conclusion Our study presents an overview of the expression pattern and functional significance of m6A regulators in human AAA. Our findings will provide a valuable resource that may guide both mechanistic and therapeutic analyses about the role of key m6A regulators in AAA.
Collapse
Affiliation(s)
- Tan Li
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Tianlong Wang
- The First Clinical College of China Medical University, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Jingjing Jing
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Hospital of China Medical University, Shenyang, People's Republic of China.,Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Liping Sun
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Hospital of China Medical University, Shenyang, People's Republic of China.,Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, The First Hospital of China Medical University, Shenyang, People's Republic of China
| |
Collapse
|
38
|
Tanaka H, Xu B, Xuan H, Ge Y, Wang Y, Li Y, Wang W, Guo J, Zhao S, Glover KJ, Zheng X, Liu S, Inuzuka K, Fujimura N, Furusho Y, Ikezoe T, Shoji T, Wang L, Fu W, Huang J, Unno N, Dalman RL. Recombinant Interleukin-19 Suppresses the Formation and Progression of Experimental Abdominal Aortic Aneurysms. J Am Heart Assoc 2021; 10:e022207. [PMID: 34459250 PMCID: PMC8649236 DOI: 10.1161/jaha.121.022207] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Interleukin-19 is an immunosuppressive cytokine produced by immune and nonimmune cells, but its role in abdominal aortic aneurysm (AAA) pathogenesis is not known. This study aimed to investigate interleukin-19 expression in, and influences on, the formation and progression of experimental AAAs. Methods and Results Human specimens were obtained at aneurysm repair surgery or from transplant donors. Experimental AAAs were created in 10- to 12-week-old male mice via intra-aortic elastase infusion. Influence and potential mechanisms of interleukin-19 treatment on AAAs were assessed via ultrasonography, histopathology, flow cytometry, and gene expression profiling. Immunohistochemistry revealed augmented interleukin-19 expression in both human and experimental AAAs. In mice, interleukin-19 treatment before AAA initiation via elastase infusion suppressed aneurysm formation and progression, with attenuation of medial elastin degradation, smooth-muscle depletion, leukocyte infiltration, neoangiogenesis, and matrix metalloproteinase 2 and 9 expression. Initiation of interleukin-19 treatment after AAA creation limited further aneurysmal degeneration. In additional experiments, interleukin-19 treatment inhibited murine macrophage recruitment following intraperitoneal thioglycolate injection. In classically or alternatively activated macrophages in vitro, interleukin-19 downregulated mRNA expression of inducible nitric oxide synthase, chemokine C-C motif ligand 2, and metalloproteinases 2 and 9 without apparent effect on cytokine-expressing helper or cytotoxic T-cell differentiation, nor regulatory T cellularity, in the aneurysmal aorta or spleen of interleukin-19-treated mice. Interleukin-19 also suppressed AAAs created via angiotensin II infusion in hyperlipidemic mice. Conclusions Based on human evidence and experimental modeling observations, interleukin-19 may influence the development and progression of AAAs.
Collapse
Affiliation(s)
- Hiroki Tanaka
- Divison of Vascular Surgery Department of Surgery Stanford University School of Medicine Stanford CA.,Division of Vascular Surgery Hamamatsu University School of Medicine Hamamatsu Shizuoka Japan
| | - Baohui Xu
- Divison of Vascular Surgery Department of Surgery Stanford University School of Medicine Stanford CA
| | - Haojun Xuan
- Divison of Vascular Surgery Department of Surgery Stanford University School of Medicine Stanford CA
| | - Yingbin Ge
- Department of Physiology Nanjing Medical University Nanjing Jiangsu China
| | - Yan Wang
- Peking University Third HospitalMedical Research Center Haidian Beijing China
| | - Yankui Li
- Divison of Vascular Surgery Department of Surgery Stanford University School of Medicine Stanford CA
| | - Wei Wang
- Department of Surgery Xiangya HospitalSouth Central University School of Medicine Changsha Hunan China
| | - Jia Guo
- Divison of Vascular Surgery Department of Surgery Stanford University School of Medicine Stanford CA
| | - Sihai Zhao
- Divison of Vascular Surgery Department of Surgery Stanford University School of Medicine Stanford CA
| | - Keith J Glover
- Divison of Vascular Surgery Department of Surgery Stanford University School of Medicine Stanford CA
| | - Xiaoya Zheng
- Divison of Vascular Surgery Department of Surgery Stanford University School of Medicine Stanford CA
| | - Shuai Liu
- Department of Surgery Xiangya HospitalSouth Central University School of Medicine Changsha Hunan China
| | - Kazunori Inuzuka
- Division of Vascular Surgery Hamamatsu University School of Medicine Hamamatsu Shizuoka Japan
| | - Naoki Fujimura
- Divison of Vascular Surgery Department of Surgery Stanford University School of Medicine Stanford CA
| | - Yuko Furusho
- Divison of Vascular Surgery Department of Surgery Stanford University School of Medicine Stanford CA
| | - Toru Ikezoe
- Divison of Vascular Surgery Department of Surgery Stanford University School of Medicine Stanford CA
| | - Takahiro Shoji
- Divison of Vascular Surgery Department of Surgery Stanford University School of Medicine Stanford CA
| | - Lixin Wang
- Department of Vascular Surgery Zhongshan HospitalFudan University Shanghai China
| | - Weiguo Fu
- Department of Vascular Surgery Zhongshan HospitalFudan University Shanghai China
| | - Jianhua Huang
- Department of Surgery Xiangya HospitalSouth Central University School of Medicine Changsha Hunan China
| | - Naoki Unno
- Division of Vascular Surgery Hamamatsu University School of Medicine Hamamatsu Shizuoka Japan
| | - Ronald L Dalman
- Divison of Vascular Surgery Department of Surgery Stanford University School of Medicine Stanford CA
| |
Collapse
|
39
|
RNA Sequencing Data from Human Intracranial Aneurysm Tissue Reveals a Complex Inflammatory Environment Associated with Rupture. Mol Diagn Ther 2021; 25:775-790. [PMID: 34403136 DOI: 10.1007/s40291-021-00552-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/27/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Intracranial aneurysm (IA) rupture leads to deadly subarachnoid hemorrhages. However, the mechanisms leading to rupture remain poorly understood. Altered gene expression within IA tissue is linked to the pathobiology of aneurysm development and progression. Here, we analyzed expression patterns of control tissue samples and compared them to those of unruptured and ruptured IA tissue samples using data from the Gene Expression Omnibus (GEO). METHODS FASTQ files for 21 ruptured IAs, 21 unruptured IAs, and 16 control tissue samples were accessed from the GEO database. DESeq2 was used for differential expression analysis in three comparisons: unruptured IA versus control, ruptured IA versus control, and ruptured versus unruptured IA. Genes that were differentially expressed in multiple comparisons were evaluated to find those progressively increasing/decreasing from control to unruptured to ruptured. Significance was tested by either analysis of variance/Gabriel or Brown-Forsythe/Games Howell (p < 0.05 was considered significant). We used additional RNA sequencing and proteomics datasets to evaluate if our differentially expressed genes (DEGs) were present in other studies. Bioinformatics analyses were performed with g:Profiler and Ingenuity Pathway Analysis. RESULTS In total, we identified 1768 DEGs, of which 318 were found in multiple comparisons. Unruptured versus control reflected vascular remodeling processes, while ruptured versus control reflected inflammatory responses and cell activation/signaling. When comparing ruptured to unruptured IAs, we found massive activation of inflammation, inflammatory responses, and leukocyte responses. Of the 318 genes in multiple comparisons, 127 were found to be significant in the multi-cohort correlation analysis. Those that progressively increased (70 genes) were associated with immune system processes, while those that progressively decreased (38 genes) did not return any gene ontology terms. Many of our DEGs were also found in the other IA tissue sequencing studies. CONCLUSIONS We found unruptured IAs relate more to remodeling processes, while ruptured IAs reflect more inflammatory and immune responses.
Collapse
|
40
|
Rustenhoven J, Tanumihardja C, Kipnis J. Cerebrovascular Anomalies: Perspectives From Immunology and Cerebrospinal Fluid Flow. Circ Res 2021; 129:174-194. [PMID: 34166075 DOI: 10.1161/circresaha.121.318173] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Appropriate vascular function is essential for the maintenance of central nervous system homeostasis and is achieved through virtue of the blood-brain barrier; a specialized structure consisting of endothelial, mural, and astrocytic interactions. While appropriate blood-brain barrier function is typically achieved, the central nervous system vasculature is not infallible and cerebrovascular anomalies, a collective terminology for diverse vascular lesions, are present in meningeal and cerebral vasculature supplying and draining the brain. These conditions, including aneurysmal formation and rupture, arteriovenous malformations, dural arteriovenous fistulas, and cerebral cavernous malformations, and their associated neurological sequelae, are typically managed with neurosurgical or pharmacological approaches. However, increasing evidence implicates interacting roles for inflammatory responses and disrupted central nervous system fluid flow with respect to vascular perturbations. Here, we discuss cerebrovascular anomalies from an immunologic angle and fluid flow perspective. We describe immune contributions, both common and distinct, to the formation and progression of diverse cerebrovascular anomalies. Next, we summarize how cerebrovascular anomalies precipitate diverse neurological sequelae, including seizures, hydrocephalus, and cognitive effects and possible contributions through the recently identified lymphatic and glymphatic systems. Finally, we speculate on and provide testable hypotheses for novel nonsurgical therapeutic approaches for alleviating neurological impairments arising from cerebrovascular anomalies, with a particular emphasis on the normalization of fluid flow and alleviation of inflammation through manipulations of the lymphatic and glymphatic central nervous system clearance pathways.
Collapse
Affiliation(s)
- Justin Rustenhoven
- Center for Brain Immunology and Glia (J.R., J.K.), Washington University in St. Louis, St Louis, MO.,Department of Pathology and Immunology, School of Medicine (J.R., J.K.), Washington University in St. Louis, St Louis, MO
| | | | - Jonathan Kipnis
- Center for Brain Immunology and Glia (J.R., J.K.), Washington University in St. Louis, St Louis, MO.,Department of Pathology and Immunology, School of Medicine (J.R., J.K.), Washington University in St. Louis, St Louis, MO
| |
Collapse
|
41
|
Xu Y, Zhang B, Chen Y, Wang X, Li Y, Wu J, Dong H, Wang S. Simvastatin increases circulating endothelial progenitor cells and inhibits the formation of intracranial aneurysms in rats with diet-induced hyperhomocysteinemia. Neurosci Lett 2021; 760:136072. [PMID: 34147541 DOI: 10.1016/j.neulet.2021.136072] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 06/01/2021] [Accepted: 06/15/2021] [Indexed: 01/21/2023]
Abstract
BACKGROUND AND PURPOSE Endothelial dysfunction triggers early pathological changes in artery, leading to the formation of intracranial aneurysm (ICA). Increase in plasma homocysteine (Hcy) impairs endothelium and endothelial progenitor cells (EPCs) are critical in repairing damaged endothelium. The aim of this study was to assess the impact of simvastatin on ICA formation in rats with hyperhomocysteinemia (HHcy). METHODS ICAs were induced in Male Sprague-Dawley rats after surgical induction in the presence of HHcy induced by a high L-methionine diet with or without oral simvastatin treatment. The size and media thickness of ICAs were evaluated 2 months after aneurysm induction. EPCs and serum vascular endothelial grow factor (VEGF) were measured be flow cytometry and ELISA respectively. Plasma Hcy levels and expression of VEGF, endothelial nitric oxide synthase (eNOS), inducible nitric oxide synthase (iNOS), matrix metalloproteinase-2 (MMP-2), and MMP-9 in aneurysmal walls were examined and correlated with ICA formation. RESULTS HHcy accelerates ICA formation and rats treated with simvastatin exhibited a significant increase in media thickness and a reduction in aneurysmal size. Simvastatin increased levels of circulating EPCs and decreased iNOS, MMP-2, MMP-9 and VEGF mRNA levels, while increased eNOS mRNA in aneurysmal tissue. CONCLUSION In a rat model, HHcy reduces circulating EPCs and accelerates ICA formation. Simvastatin treatment increases circulating EPCs and inhabits the formation of ICA. We have shown a close association among circulating EPCs, biochemical markers related to vascular remodeling and the formation of ICA.
Collapse
Affiliation(s)
- Yong Xu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; Department of Neurosurgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Bin Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yu Chen
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xiu Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yong Li
- Department of Neurosurgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Jiangping Wu
- Department of Neurosurgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Hao Dong
- Department of Neurosurgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Shuo Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
42
|
Mahdi A, Wernly B, Pernow J, Zhou Z. Sunitinib and its effect in the cardiovascular system. Drug Discov Today 2021; 26:1773-1775. [PMID: 34023497 DOI: 10.1016/j.drudis.2021.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 05/08/2021] [Accepted: 05/11/2021] [Indexed: 11/17/2022]
Affiliation(s)
- Ali Mahdi
- Division of Cardiology, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Bernhard Wernly
- Division of Cardiology, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden; Department of Anaesthesiology, Perioperative Medicine and Intensive Care Medicine, Paracelsus Medical University of Salzburg, Austria; Department of Cardiology, Paracelsus Medical University of Salzburg, Austria; Center for Public Health and Healthcare Research, Paracelsus Medical University of Salzburg, Austria
| | - John Pernow
- Division of Cardiology, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Zhichao Zhou
- Division of Cardiology, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
43
|
Shi J, Guo J, Li Z, Xu B, Miyata M. Importance of NLRP3 Inflammasome in Abdominal Aortic Aneurysms. J Atheroscler Thromb 2021; 28:454-466. [PMID: 33678767 PMCID: PMC8193780 DOI: 10.5551/jat.rv17048] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 01/22/2021] [Indexed: 12/14/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) is a chronic inflammatory degenerative aortic disease, which particularly affects older people. Nucleotide-binding oligomerization domain-like receptor family protein 3 (NLRP3) inflammasome is a multi-protein complex and mediates inflammatory responses by activating caspase 1 for processing premature interleukin (IL)-1β and IL-18. In this review, we first summarize the principle of NLRP3 inflammasome activation and the functionally distinct classes of small molecule NLRP3 inflammasome inhibitors. Next, we provide a comprehensive literature review on the expression of NLRP3 inflammasome effector mediators (IL-1β and IL-18) and components (caspase 1, apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) and NLRP3) in clinical and experimental AAAs. Finally, we discuss the influence of genetic deficiency or pharmacological inhibition of individual effector mediators and components of NLRP3 inflammasome on experimental AAAs. Accumulating clinical and experimental evidence suggests that NLRP3 inflammasome may be a promise therapeutic target for developing pharmacological strategies for clinical AAA management.
Collapse
Affiliation(s)
- Jinyun Shi
- Center for Hypertension Care, Shanxi Medical University First Hospital, Taiyuan, Shanxi Province, P. R. China
| | - Jia Guo
- Center for Hypertension Care, Shanxi Medical University First Hospital, Taiyuan, Shanxi Province, P. R. China
| | - Zhidong Li
- Department of Pharmacology, Shanxi Medical University, Taiyuan, Shanxi Province, P. R. China
| | - Baohui Xu
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Masaaki Miyata
- School of Health Science, Faculty of Medicine, Kagoshima University, Kagoshima University, Kagoshima, Japan
| |
Collapse
|
44
|
Liraglutide, a TFEB-Mediated Autophagy Agonist, Promotes the Viability of Random-Pattern Skin Flaps. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6610603. [PMID: 33868571 PMCID: PMC8032515 DOI: 10.1155/2021/6610603] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 01/29/2021] [Accepted: 02/05/2021] [Indexed: 12/14/2022]
Abstract
Random skin flaps are commonly used in reconstruction surgery. However, distal necrosis of the skin flap remains a difficult problem in plastic surgery. Many studies have shown that activation of autophagy is an important means of maintaining cell homeostasis and can improve the survival rate of flaps. In the current study, we investigated whether liraglutide can promote the survival of random flaps by stimulating autophagy. Our results show that liraglutide can significantly improve flap viability, increase blood flow, and reduce tissue oedema. In addition, we demonstrated that liraglutide can stimulate angiogenesis and reduce pyroptosis and oxidative stress. Through immunohistochemistry analysis and Western blotting, we verified that liraglutide can enhance autophagy, while the 3-methylladenine- (3MA-) mediated inhibition of autophagy enhancement can significantly reduce the benefits of liraglutide described above. Mechanistically, we showed that the ability of liraglutide to enhance autophagy is mediated by the activation of transcription factor EB (TFEB) and its subsequent entry into the nucleus to activate autophagy genes, a phenomenon that may result from AMPK-MCOLN1-calcineurin signalling pathway activation. Taken together, our results show that liraglutide is an effective drug that can significantly improve the survival rate of random flaps by enhancing autophagy, inhibiting oxidative stress in tissues, reducing pyroptosis, and promoting angiogenesis, which may be due to the activation of TFEB via the AMPK-MCOLN1-calcineurin signalling pathway.
Collapse
|
45
|
Chen L, Xu P, Xiao Q, Chen L, Li S, Jian JM, Zhong YB. Sunitinib malate inhibits intestinal tumor development in male Apc Min/+ mice by down-regulating inflammation-related factors with suppressing β-cateinin/c-Myc pathway and re-balancing Bcl-6 and Caspase-3. Int Immunopharmacol 2021; 90:107128. [PMID: 33191180 DOI: 10.1016/j.intimp.2020.107128] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 10/16/2020] [Accepted: 10/18/2020] [Indexed: 02/07/2023]
Abstract
Sunitinib is a tyrosine kinase inhibitor for many tumors. Inflammation is one of the most important factors in the development of intestinal tumors. Many inflammation-related factors are regulated by tyrosine kinase receptors. It is reasonable to hypothesize that sunitinib can regulate the development of intestinal tumors by regulating the expression and/or activity of inflammation-related factors. Here, ApcMin/+ male mouse model was used to investigate the effect and mechanism of sunitinib malate against intestinal cancer. Results show that compared to vehicle, after sunitinib malate treatment, overall survival of ApcMin/+ mice was lengthened up to 25 days, with a gain of body weight, reduction of spleen/body weight index, and RBC, WBC and HGC regulated to normal levels of wild type mice, and a number of polyps no less than 1 mm significantly reduced. Meanwhile, in the intestines, the nuclear β-Catenin protein and c-Myc mRNA were both down-regulated, and Bcl-6 was significantly reduced with Caspase-3 up regulated. Furthermore, inflammation-related factors including IL-6, TNF-α, IL-1α, IL-1β and IFN-γ were down-regulated at mRNA levels in the intestines. These results suggest that sunitinib malate can significantly improve the survival status and inhibit intestinal tumor development in male ApcMin/+ mice, through inhibiting inflammation-related factors, while suppressing β-cateinin/c-Myc pathway and re-balancing protein levels of Bcl-6 and Caspase-3.
Collapse
Affiliation(s)
- Lai Chen
- Integrated Chinese & Western Medicine Oncology Research Center, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi 330004, China; Laboratory Animal Research Center for Science and Technology, Jiangxi University of Traditional Chinese Medicine, 1688 Meiling Road, Nanchang 330004, China; Key Laboratory of Pharmacology of Traditional Chinese Medicine in Jiangxi, Nanchang 330004, China; Department of Postgraduate, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, China
| | - Pan Xu
- Integrated Chinese & Western Medicine Oncology Research Center, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi 330004, China; Key Laboratory of Pharmacology of Traditional Chinese Medicine in Jiangxi, Nanchang 330004, China; Department of Postgraduate, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, China
| | - Qiuping Xiao
- Research and Development Department of Jiangzhong Pharmaceutical Co., Ltd., Nanchang 330004, China
| | - Liling Chen
- Laboratory Animal Research Center for Science and Technology, Jiangxi University of Traditional Chinese Medicine, 1688 Meiling Road, Nanchang 330004, China; Key Laboratory of Pharmacology of Traditional Chinese Medicine in Jiangxi, Nanchang 330004, China
| | - Shanshan Li
- Laboratory Animal Research Center for Science and Technology, Jiangxi University of Traditional Chinese Medicine, 1688 Meiling Road, Nanchang 330004, China; Key Laboratory of Pharmacology of Traditional Chinese Medicine in Jiangxi, Nanchang 330004, China
| | - Ji-Mo Jian
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Wenhua West Road, Jinan, Shandong 250012, China; Department of Hematology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, Shandong 266035, China
| | - You-Bao Zhong
- Laboratory Animal Research Center for Science and Technology, Jiangxi University of Traditional Chinese Medicine, 1688 Meiling Road, Nanchang 330004, China; Key Laboratory of Pharmacology of Traditional Chinese Medicine in Jiangxi, Nanchang 330004, China; Department of Postgraduate, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, China.
| |
Collapse
|
46
|
Ye T, Zhang G, Liu H, Shi J, Qiu H, Liu Y, Han F, Hou N. Relationships Between Perivascular Adipose Tissue and Abdominal Aortic Aneurysms. Front Endocrinol (Lausanne) 2021; 12:704845. [PMID: 34194399 PMCID: PMC8236981 DOI: 10.3389/fendo.2021.704845] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 05/25/2021] [Indexed: 02/05/2023] Open
Abstract
Abdominal aortic aneurysms (AAAs) are typically asymptomatic, and there is a high mortality rate associated with aneurysm rupture. AAA pathogenesis involves extracellular matrix degradation, vascular smooth muscle cell phenotype switching, inflammation, and oxidative stress. There is increasing evidence of excessive adipocyte accumulation in ruptured AAA walls. These excessive numbers of adipocytes in the vascular wall have been closely linked with AAA progression. Perivascular adipose tissue (PVAT), a unique type of adipose tissue, can be involved in adipocyte accumulation in the AAA wall. PVAT produces various chemokines and adipocytokines around vessels to maintain vascular homeostasis through paracrine and autocrine mechanisms in normal physiological conditions. Nevertheless, PVAT loses its normal function and promotes the progression of vascular diseases in pathological conditions. There is evidence of significantly reduced AAA diameter in vessel walls of removed PVAT. There is a need to highlight the critical roles of cytokines, cells, and microRNA derived from PVAT in the regulation of AAA development. PVAT may constitute an important therapeutic target for the prevention and treatment of AAAs. In this review, we discuss the relationship between PVAT and AAA development; we also highlight the potential for PVAT-derived factors to serve as a therapeutic target in the treatment of AAAs.
Collapse
Affiliation(s)
- Tongtong Ye
- Department of Endocrinology, Affiliated Hospital of Weifang Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Guangdong Zhang
- Department of Endocrinology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Hangyu Liu
- Department of Ophthalmology, Weifang Eye Hospital, Weifang, China
| | - Junfeng Shi
- Department of Endocrinology, Affiliated Hospital of Weifang Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Hongyan Qiu
- Department of Endocrinology, Affiliated Hospital of Weifang Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Yongping Liu
- Department of Endocrinology, Affiliated Hospital of Weifang Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Fang Han
- Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang, China
- *Correspondence: Ningning Hou, ; Fang Han,
| | - Ningning Hou
- Department of Endocrinology, Affiliated Hospital of Weifang Medical University, Weifang, China
- *Correspondence: Ningning Hou, ; Fang Han,
| |
Collapse
|
47
|
Xie S, Ma L, Guan H, Guan S, Wen L, Han C. Daphnetin suppresses experimental abdominal aortic aneurysms in mice via inhibition of aortic mural inflammation. Exp Ther Med 2020; 20:221. [PMID: 33193836 PMCID: PMC7646695 DOI: 10.3892/etm.2020.9351] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 07/21/2020] [Indexed: 12/21/2022] Open
Abstract
Rupture of abdominal aortic aneurysm (AAA) is a devastating event that can be prevented by inhibiting the growth of small aneurysms. Therapeutic strategies targeting certain events that promote the development of AAA must be developed, in order to alter the course of AAA. Chronic inflammation of the aortic mural is a major characteristic of AAA and is related to AAA formation, development and rupture. Daphnetin (DAP) is a coumarin derivative with anti-inflammatory properties that is extracted from Daphne odora var. However, the effect of DAP on AAA development remains unclear. The present study investigated the effect of DAP on the formation and development of experimental AAAs and its potential underlying mechanisms. A mice AAA model was established by intra-aortic infusion of porcine pancreatic elastase (PPE), and mice were intraperitoneally injected with DAP immediately after PPE infusion. The maximum diameter of the abdominal aorta was measured by ultrasound system, and aortic mural changes were investigated by Elastica van Gieson (EVG) staining and immunohistochemical staining. The results demonstrated that DAP significantly suppressed PPE-induced AAA formation and attenuated the depletion of aortic medial elastin and smooth muscle cells in the media of the aorta. Furthermore, the density of mural macrophages, T cells and B cells were significantly attenuated in DAP-treated AAA mice. In addition, treatment with DAP resulted in a significant reduction in mural neovessels. These findings indicated that DAP may limit the formation and progression of experimental aneurysms by inhibiting mural inflammation and angiogenesis. These data confirmed the translational potential of DAP inclinical AAA inhibition strategies.
Collapse
Affiliation(s)
- Shiyun Xie
- Department of Vascular Surgery, Shandong Shanxian Central Hospital, Shanxian, Shandong 274300, P.R. China
| | - Li Ma
- Department of Vascular Surgery, Shandong Shanxian Central Hospital, Shanxian, Shandong 274300, P.R. China
| | - Hongliang Guan
- Department of Vascular Surgery, Shandong Shanxian Central Hospital, Shanxian, Shandong 274300, P.R. China
| | - Su Guan
- Department of Vascular Surgery, Shandong Shanxian Central Hospital, Shanxian, Shandong 274300, P.R. China
| | - Lijuan Wen
- Department of Vascular Surgery, Shandong Shanxian Central Hospital, Shanxian, Shandong 274300, P.R. China
| | - Chanchan Han
- Department of Ultrasound, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| |
Collapse
|
48
|
Nikol S, Mathias K, Olinic DM, Blinc A, Espinola-Klein C. Aneurysms and dissections - What is new in the literature of 2019/2020 - a European Society of Vascular Medicine annual review. VASA 2020; 49:1-36. [PMID: 32856993 DOI: 10.1024/0301-1526/a000865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
More than 6,000 publications were found in PubMed concerning aneurysms and dissections, including those Epub ahead of print in 2019, printed in 2020. Among those publications 327 were selected and considered of particular interest.
Collapse
Affiliation(s)
- Sigrid Nikol
- Department of Angiology, ASKLEPIOS Klinik St. Georg, Hamburg, Germany.,University of Münster, Germany
| | - Klaus Mathias
- World Federation for Interventional Stroke Treatment (WIST), Hamburg, Germany
| | - Dan Mircea Olinic
- Medical Clinic No. 1, University of Medicine and Pharmacy and Interventional Cardiology Department, Emergency Hospital, Cluj-Napoca, Romania
| | - Aleš Blinc
- Department of Vascular Diseases, University Medical Centre Ljubljana, Slovenia.,Faculty of Medicine, University of Ljubljana, Slovenia
| | | |
Collapse
|
49
|
Zhao M, Xu L, Qian H. Bioinformatics analysis of microRNA profiles and identification of microRNA-mRNA network and biological markers in intracranial aneurysm. Medicine (Baltimore) 2020; 99:e21186. [PMID: 32756097 PMCID: PMC7402807 DOI: 10.1097/md.0000000000021186] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Intracranial aneurysm (IA) is a kind of cerebrovascular disorder, which may result in the subarachnoid hemorrhage with high lethality and disability. The purpose of this study was to reveal the pathogenesis and identify novel biomarkers in IA.We processed the raw microRNA (miRNA) expression profile data of IA obtained from Gene Expression Omnibus. Then weighted correlation network analysis was performed to identify the hub miRNAs in IA. Target genes of hub miRNAs were predicted using multiR package. In addition, a protein-protein network as well as miRNA-mRNA network was constructed and functional and pathway enrichment analyses were done. Finally, the prediction value of hub miRNAs in IA was tested in validation set.Two modules that had relation with IA were identified and 10 hub miRNAs in each module with higher gene-module association were selected. The protein-protein network and miRNA-mRNA network contained 243 nodes and 1496 edges. Functional and pathway enrichment analyses showed that they were mainly enriched in cell cycle, cell proliferation, and PI3K/Akt signaling pathways. Besides, hsa-miR-191-3p, hsa-miR-423-5p, hsa-miR-424-5p, hsa-miR-425-3p were proven to be valuable in prediction IA occurrence.In a word, this study reveals hub miRNAs, target genes and pathways potentially participating in formation and development of IA and screens out some candidate biomarkers. Our findings provide some new perspectives for research and treatment of IA.
Collapse
Affiliation(s)
- Ming Zhao
- Department of Neurosurgery, Zhuji People's Hospital of Zhejiang Province, Zhuji Affiliated Hospital of Shaoxing University, Zhuji
| | - Longbiao Xu
- Department of Neurosurgery, Zhuji People's Hospital of Zhejiang Province, Zhuji Affiliated Hospital of Shaoxing University, Zhuji
| | - Hui Qian
- Department of Neurosurgery, Shaoxing Central Hospital, Shaoxing, Zhejiang, China
| |
Collapse
|
50
|
Colman L, Caggiani M, Leyva A, Bresque M, Liechocki S, Maya-Monteiro CM, Mazal D, Batthyany C, Calliari A, Contreras P, Escande C. The protein Deleted in Breast Cancer-1 (DBC1) regulates vascular response and formation of aortic dissection during Angiotensin II infusion. Sci Rep 2020; 10:6772. [PMID: 32317757 PMCID: PMC7174338 DOI: 10.1038/s41598-020-63841-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 04/07/2020] [Indexed: 12/25/2022] Open
Abstract
Cardiovascular diseases are among the main causes of morbimortality in the adult population. Among them, hypertension is a leading cause for stroke, heart disease and kidney failure. Also, as a result of arterial wall weakness, hypertension can lead to the development of dissecting aortic aneurysms, a rare but often fatal condition if not readily treated. In this work, we investigated the role of DBC1 in the regulation of vascular function in an ANGII-induced hypertension mouse model. We found that WT and DBC1 KO mice developed hypertension in response to ANGII infusion. However, DBC1 KO mice showed increased susceptibility to develop aortic dissections. The effect was accompanied by upregulation of vascular remodeling factors, including MMP9 and also VEGF. Consistent with this, we found decreased collagen deposition and elastic fiber fragmentation, suggesting that increased expression of MMPs in DBC1 KO mice weakens the arterial wall, promoting the formation of aortic dissections during treatment with ANGII. Finally, DBC1 KO mice had reduced cell proliferation in the intima-media layer in response to ANGII, paralleled with an impairment to increase wall thickness in response to hypertension. Furthermore, VSMC purified from DBC1 KO mice showed impaired capacity to leave quiescence, confirming the in vivo results. Altogether, our results show for the first time that DBC1 regulates vascular response and function during hypertension and protects against vascular injury. This work also brings novel insights into the molecular mechanisms of the development of aortic dissections.
Collapse
Affiliation(s)
- Laura Colman
- Laboratory of Metabolic Diseases and Aging, INDICyO Program, Institut Pasteur Montevideo, Montevideo, Uruguay
| | - Maria Caggiani
- Laboratory of Metabolic Diseases and Aging, INDICyO Program, Institut Pasteur Montevideo, Montevideo, Uruguay
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República (UdelaR), Montevideo, Uruguay
| | - Alejandro Leyva
- Laboratory of Vascular Biology and Rational Drug Design, INDICyO Program, Institut Pasteur Montevideo, Montevideo, Uruguay
- Analytical Biochemistry and Proteomics Unit, Institut Pasteur Montevideo and Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| | - Mariana Bresque
- Laboratory of Metabolic Diseases and Aging, INDICyO Program, Institut Pasteur Montevideo, Montevideo, Uruguay
| | - Sally Liechocki
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Clarissa M Maya-Monteiro
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Daniel Mazal
- Departamento de Anatomía Patológica, Facultad de Medicina, Universidad de la República (UdelaR) and C.H Pereira Rossell, Montevideo, Uruguay
| | - Carlos Batthyany
- Laboratory of Vascular Biology and Rational Drug Design, INDICyO Program, Institut Pasteur Montevideo, Montevideo, Uruguay
| | - Aldo Calliari
- Laboratory of Metabolic Diseases and Aging, INDICyO Program, Institut Pasteur Montevideo, Montevideo, Uruguay
- Departamento de Biociencias, Facultad de Veterinaria, Universidad de la República (UdelaR), Montevideo, Uruguay
| | - Paola Contreras
- Laboratory of Metabolic Diseases and Aging, INDICyO Program, Institut Pasteur Montevideo, Montevideo, Uruguay
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República (UdelaR), Montevideo, Uruguay
| | - Carlos Escande
- Laboratory of Metabolic Diseases and Aging, INDICyO Program, Institut Pasteur Montevideo, Montevideo, Uruguay.
| |
Collapse
|