1
|
Wang Y, Gao Y, Shi H, Gao R, Yang J, Qu Y, Hu S, Zhang J, Wang J, Cao J, Zhang F, Ge J. CCL11 released by GSDMD-mediated macrophage pyroptosis regulates angiogenesis after hindlimb ischemia. Cell Death Discov 2024; 10:294. [PMID: 38906863 PMCID: PMC11192718 DOI: 10.1038/s41420-023-01764-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 12/02/2023] [Accepted: 12/06/2023] [Indexed: 06/23/2024] Open
Abstract
Peripheral vascular disease (PVD) is an emerging public health burden with a high rate of disability and mortality. Gasdermin D (GSDMD) has been reported to exert pyroptosis and play a critical role in the pathophysiology of many cardiovascular diseases. We ought to determine the role of GSDMD in the regulation of perfusion recovery after hindlimb ischemia (HLI). Our study revealed that GSDMD-mediated pyroptosis occurred in HLI. GSDMD deletion aggravated perfusion recovery and angiogenesis in vitro and in vivo. However, how GSDMD regulates angiogenesis after ischemic injury remains unclear. We then found that GSDMD-mediated pyroptosis exerted the angiogenic capacity in macrophages rather than endothelial cells after HLI. GSDMD deletion led to a lower level of CCL11 in mice serum. GSDMD knockdown in macrophages downregulated the expression and decreased the releasing level of CCL11. Furthermore, recombinant CCL11 improved endothelial functions and angiogenesis, which was attenuated by CCL11 antibody. Taken together, these results demonstrate that GSDMD promotes angiogenesis by releasing CCL11, thereby improving blood flow perfusion recovery after hindlimb ischemic injury. Therefore, CCL11 may be a novel target for prevention and treatment of vascular ischemic diseases.
Collapse
Affiliation(s)
- Yiwen Wang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
| | - Yang Gao
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
| | - Huairui Shi
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
| | - Rifeng Gao
- Department of Cardiology, Shanghai Fifth People's Hospital, Fudan University, 200240, Shanghai, China
| | - Ji'e Yang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
| | - Ya'nan Qu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
| | - Shiyu Hu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
| | - Jian Zhang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
| | - Jingpu Wang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
| | - Jiatian Cao
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
| | - Feng Zhang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 200032, Shanghai, China.
| | - Junbo Ge
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 200032, Shanghai, China.
- Key Laboratory of Viral Heart Diseases, National Health Commission, 200032, Shanghai, China.
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, 200032, Shanghai, China.
- National Clinical Research Center for Interventional Medicine, 200032, Shanghai, China.
- Institutes of Biomedical Sciences, Fudan University, 200032, Shanghai, China.
| |
Collapse
|
2
|
Sastriques-Dunlop S, Elizondo-Benedetto S, Arif B, Meade R, Zaghloul MS, Luehmann H, Heo GS, English SJ, Liu Y, Zayed MA. Ketosis prevents abdominal aortic aneurysm rupture through C-C chemokine receptor type 2 downregulation and enhanced extracellular matrix balance. Sci Rep 2024; 14:1438. [PMID: 38228786 PMCID: PMC10791699 DOI: 10.1038/s41598-024-51996-7] [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: 01/12/2024] [Indexed: 01/18/2024] Open
Abstract
Abdominal aortic aneurysms (AAAs) are prevalent with aging, and AAA rupture is associated with increased mortality. There is currently no effective medical therapy to prevent AAA rupture. The monocyte chemoattractant protein (MCP-1)/C-C chemokine receptor type 2 (CCR2) axis critically regulates AAA inflammation, matrix-metalloproteinase (MMP) production, and extracellular matrix (ECM) stability. We therefore hypothesized that a diet intervention that can modulate CCR2 axis may therapeutically impact AAA risk of rupture. Since ketone bodies (KBs) can trigger repair mechanisms in response to inflammation, we evaluated whether systemic ketosis in vivo could reduce CCR2 and AAA progression. Male Sprague-Dawley rats underwent surgical AAA formation using porcine pancreatic elastase and received daily β-aminopropionitrile to promote AAA rupture. Rats with AAAs received either a standard diet, ketogenic diet (KD), or exogenous KBs (EKB). Rats receiving KD and EKB reached a state of ketosis and had significant reduction in AAA expansion and incidence of rupture. Ketosis also led to significantly reduced aortic CCR2 content, improved MMP balance, and reduced ECM degradation. Consistent with these findings, we also observed that Ccr2-/- mice have significantly reduced AAA expansion and rupture. In summary, this study demonstrates that CCR2 is essential for AAA expansion, and that its modulation with ketosis can reduce AAA pathology. This provides an impetus for future clinical studies that will evaluate the impact of ketosis on human AAA disease.
Collapse
Affiliation(s)
- Sergio Sastriques-Dunlop
- Section of Vascular Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Santiago Elizondo-Benedetto
- Section of Vascular Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Batool Arif
- Section of Vascular Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Rodrigo Meade
- Section of Vascular Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Mohamed S Zaghloul
- Section of Vascular Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Hannah Luehmann
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Gyu S Heo
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Sean J English
- Section of Vascular Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Yongjian Liu
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Mohamed A Zayed
- Section of Vascular Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA.
- Division of Molecular Cell Biology, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University, St. Louis, MO, USA.
- Veterans Affairs St. Louis Health Care System, St. Louis, MO, USA.
| |
Collapse
|
3
|
Zayed M, Sastriques-Dunlop S, Elizondo-Benedetto S, Arif B, Meade R, Zaghloul M, Luehmann H, Heo G, English S, Liu Y. Ketosis Prevents Abdominal Aortic Aneurysm Rupture Through C-C Chemokine Receptor Type 2 Downregulation and Enhanced MMP Balance. RESEARCH SQUARE 2023:rs.3.rs-3054767. [PMID: 37461581 PMCID: PMC10350122 DOI: 10.21203/rs.3.rs-3054767/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/24/2023]
Abstract
Abdominal aortic aneurysms (AAAs) are prevelant with aging, and AAA rupture is associated with high mortality. There is currently no effective medical therapy for AAA rupture. Previous work demonstrated that the monocyte chemoattractant protein (MCP-1) / C-C chemokine receptor type 2 (CCR2) axis critically regulates AAA inflammation, matrix-metalloproteinase (MMP) production, and extracellular matrix (ECM) stability. Here we similarly observed that Ccr2-/- mice have significantly reduced AAA expansion and rupture. We therefore hypothesized that a dietary modulation of the CCR2 axis may therapeutically impact AAA risk of rupture. Since ketone bodies (KBs) can trigger repair mechanisms in response to inflammation, we specifically evaluated whether systemic ketosis in vivo can reduce CCR2 and AAA progression. Male Sprague-Dawley rats underwent surgical AAA formation using porcine pancreatic elastase (PPE), and received daily β-aminopropionitrile (BAPN) to promote AAA rupture. Animals with AAAs received either a standard diet (SD), ketogenic diet (KD), or exogenous KBs (EKB). Animals recieving KD and EKB reached a state of ketosis, and had significant reduction in AAA expansion and incidence of rupture. Ketosis also led to significantly reduced aortic CCR2 content, improved MMP balance, and reduced ECM degradation. In summary, this study demonstrates that ketosis plays a crucial role in AAA pathobiology, and provides the impetus for future clinical studies investigating the potential benefit of ketosis for prevention of AAA expansion and rupture.
Collapse
Affiliation(s)
- Mohamed Zayed
- Washington University in St. Louis School of Medicine
| | | | | | - Batool Arif
- Washington University in St. Louis School of Medicine
| | - Rodrigo Meade
- Washington University in St. Louis School of Medicine
| | | | | | - Gyu Heo
- ashington University in St. Louis School of Medicine
| | - Sean English
- ashington University in St. Louis School of Medicine
| | | |
Collapse
|
4
|
Li J, Ma S, Jia X, Bu Y, Zhou T, Zhang L, Qiu M, Wang X. Rivaroxaban in patients with abdominal aortic aneurysm and high-sensitivity C-reactive protein elevation (BANBOO): study protocol for a randomized, controlled trial. Trials 2023; 24:419. [PMID: 37337298 DOI: 10.1186/s13063-023-07461-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 06/13/2023] [Indexed: 06/21/2023] Open
Abstract
BACKGROUND Abdominal aortic aneurysm (AAA) is a fatal disease due to the tendency to rupture. The drug treatment for small AAA without surgical indications has been controversial. Previous studies showed that high-sensitivity C-reactive protein (hs-CRP) had become a potential biomarker of the disease, and the anti-inflammatory effect of rivaroxaban for AAA had been well established. Thus, we hypothesized that rivaroxaban could control the progression of AAA in patients with hs-CRP elevation. METHODS The study is a prospective, open-label, randomized, controlled clinical trial. Sixty subjects are recruited from the General Hospital of Northern Theatre Command of China. Subjects are randomly assigned (1:1) to the intervention arm (rivaroxaban) or control arm (aspirin). The primary efficacy outcome is the level of serum hs-CRP at 6 months. The secondary outcomes include imaging examination (the maximal diameter of AAA, the maximal thickness of mural thrombus, and the length of aneurysm), major adverse cardiovascular and cerebrovascular events (MACCE, including AAA transformation, non-fatal myocardial infarction, acute congestive heart failure, stent thrombosis, ischemia-driven target vessel revascularization, vascular amputation, stroke, cardiovascular death, and all-cause death), and other laboratory tests (troponin T, interleukin 6, D-dimer, and coagulation function). DISCUSSION The BANBOO trial tested the effect of rivaroxaban on the progression of AAA in patients with elevated Hs-CRP for the first time. TRIAL REGISTRATION ChiCTR2100051990, ClinicalTrials.gov, registered on 12 October 2021.
Collapse
Affiliation(s)
- Jingyuan Li
- College of Life Science and Biopharmaceutical, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, Liaoning, China
- Department of Cardiology, General Hospital of Northern Theatre Command, Liaoning, 110016, Shenyang, China
| | - Sicong Ma
- Department of Cardiology, General Hospital of Northern Theatre Command, Liaoning, 110016, Shenyang, China
| | - Xiu Jia
- Department of Cardiology, General Hospital of Northern Theatre Command, Liaoning, 110016, Shenyang, China
| | - Yingzhen Bu
- Department of Cardiology, General Hospital of Northern Theatre Command, Liaoning, 110016, Shenyang, China
| | - Tienan Zhou
- Department of Cardiology, General Hospital of Northern Theatre Command, Liaoning, 110016, Shenyang, China
| | - Lei Zhang
- Department of Cardiology, General Hospital of Northern Theatre Command, Liaoning, 110016, Shenyang, China
| | - Miaohan Qiu
- Department of Cardiology, General Hospital of Northern Theatre Command, Liaoning, 110016, Shenyang, China
| | - Xiaozeng Wang
- College of Life Science and Biopharmaceutical, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, Liaoning, China.
- Department of Cardiology, General Hospital of Northern Theatre Command, Liaoning, 110016, Shenyang, China.
| |
Collapse
|
5
|
Demirci G, Demir AR, Uygur B, Bulut U, Avci Y, Tükenmez Karakurt S, Memiç Sancar K, Aktemur T, Ersoy B, Celik O, Erturk M. C-reactive protein to albumin ratio provides important long-term prognostic information in patients undergoing endovascular abdominal aortic repair. Vascular 2023; 31:270-278. [PMID: 35014591 DOI: 10.1177/17085381211062736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND The prognostic value of C-reactive protein/albumin ratio (CAR) is of import in cardiovascular diseases. Our aim was to evaluate the impact of the CAR in patients with asymptomatic abdominal aortic aneurysm (AAA) undergoing endovascular aneurysm repair (EVAR). MATERIAL AND METHOD We retrospectively evaluated 127 consecutive patients who underwent technically successful elective EVAR procedure between December 2014 and September 2020. The optimal CAR cut-off value was determined by using receiver operating characteristic (ROC) curve analysis. Based on the cut-off value, we investigated the association of CAR with long-term all-cause mortality. RESULTS 32 (25.1%) of the patients experienced all-cause mortality during a mean 32.7 ± 21.7 months' follow-up. In the group with mortality, CAR was significantly higher than in the survivor group (4.63 (2.60-11.88) versus 1.63 (0.72-3.24), p < 0.001). Kaplan-Meier curves showed a higher incidence of all-cause mortality in patients with high CAR compared to patients with low CAR (log-rank test, p < 0.001). Multivariable Cox regression analysis revealed that glucose ≥ 110 mg/dL (HR: 2.740; 95% CI: 1.354-5.542; p = 0.005), creatinine ≥ 0.99 mg/dL (HR: 2.957, 95% CI: 1.282-6.819, p = 0.011) and CAR > 2.05 (HR: 8.190, 95% CI: 1.899-35.320, p = 0.005) were the independent predictors of mortality. CONCLUSION CAR was associated with a significant increase in postoperative long-term mortality in patients who underwent EVAR. Preoperatively calculated CAR can be used as an important prognostic factor.
Collapse
Affiliation(s)
- Gökhan Demirci
- Department of Cardiology, Training and Research Hospital, Mehmet Akif Ersoy Thoracic and Cardiovascular Surgery Center, 484473University of Health Sciences, Istanbul, Turkey
| | - Ali Riza Demir
- Department of Cardiology, Training and Research Hospital, Mehmet Akif Ersoy Thoracic and Cardiovascular Surgery Center, 484473University of Health Sciences, Istanbul, Turkey
| | - Begüm Uygur
- Department of Cardiology, Training and Research Hospital, Mehmet Akif Ersoy Thoracic and Cardiovascular Surgery Center, 484473University of Health Sciences, Istanbul, Turkey
| | - Umit Bulut
- Department of Cardiology, Training and Research Hospital, Mehmet Akif Ersoy Thoracic and Cardiovascular Surgery Center, 484473University of Health Sciences, Istanbul, Turkey
| | - Yalcin Avci
- Department of Cardiology, Training and Research Hospital, Mehmet Akif Ersoy Thoracic and Cardiovascular Surgery Center, 484473University of Health Sciences, Istanbul, Turkey
| | - Seda Tükenmez Karakurt
- Department of Cardiology, Training and Research Hospital, Mehmet Akif Ersoy Thoracic and Cardiovascular Surgery Center, 484473University of Health Sciences, Istanbul, Turkey
| | - Kadriye Memiç Sancar
- Department of Cardiology, Training and Research Hospital, Mehmet Akif Ersoy Thoracic and Cardiovascular Surgery Center, 484473University of Health Sciences, Istanbul, Turkey
| | - Tugba Aktemur
- Department of Cardiology, Training and Research Hospital, Mehmet Akif Ersoy Thoracic and Cardiovascular Surgery Center, 484473University of Health Sciences, Istanbul, Turkey
| | - Burak Ersoy
- Department of Cardiology, Training and Research Hospital, Mehmet Akif Ersoy Thoracic and Cardiovascular Surgery Center, 484473University of Health Sciences, Istanbul, Turkey
| | - Omer Celik
- Department of Cardiology, Training and Research Hospital, Mehmet Akif Ersoy Thoracic and Cardiovascular Surgery Center, 484473University of Health Sciences, Istanbul, Turkey
| | - Mehmet Erturk
- Department of Cardiology, Training and Research Hospital, Mehmet Akif Ersoy Thoracic and Cardiovascular Surgery Center, 484473University of Health Sciences, Istanbul, Turkey
| |
Collapse
|
6
|
Expression of TLR4 Is Upregulated in Patients with Sporadic Acute Stanford Type A Aortic Dissection. Cardiol Res Pract 2022; 2022:3806462. [DOI: 10.1155/2022/3806462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 08/31/2022] [Indexed: 11/17/2022] Open
Abstract
Sporadic acute Stanford type A aortic dissection (TAAD) is a serious condition that requires urgent treatment to avoid catastrophic consequences. The purpose of the present study was to explore, firstly, whether TLR4-regulated immune signalling molecules were activated in TAAD patients and, secondly, whether TLR4-regulated inflammatory products interleukin-1β (IL-1β) and CC chemokine ligand 5 (CCL5) could be a promising biomarker for diagnosis in patients with TAAD. Full-thickness ascending aortic wall specimens from TAAD patients (n = 12) and control donors (n = 12) were examined for the expression of TLR4 and its major signalling molecules, in terms of immunity and inflammation. Blood samples from TAAD (n = 49) and control patients (n = 53) were collected to detect the circulating plasma cytokine levels of IL-1β and CCL5. We demonstrated that expression levels of TLR4 and its downstream signalling cascade molecules were significantly elevated. Furthermore, receiver operating characteristic curve analyses showed that elevated IL-1β levels and decreased plasma CCL5 may have diagnostic value for TAAD. In summary, this current study suggests a more generalized pattern of inflammation in TAAD. In addition, TLR4-mediated inflammatory product, such as IL-1β and CCL5, could be novel and promising biomarkers with important diagnostic and predictive value in the identification of sporadic TAAD diseases.
Collapse
|
7
|
Wang H, Zhong Z, Jiang D, Zhang H, Yin F, Guo P, Chen J, Zhu X, You K, Han Y, Liu K. Circulating Inflammatory Mediators and Genetic Polymorphisms of Inflammation Mediators and Their Association with Factors Related to Abdominal Aortic Aneurysm: A Systemic Review and Meta-Analysis. Rev Cardiovasc Med 2022; 23:270. [PMID: 39076643 PMCID: PMC11266967 DOI: 10.31083/j.rcm2308270] [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: 01/17/2022] [Revised: 04/23/2022] [Accepted: 05/17/2022] [Indexed: 07/31/2024] Open
Abstract
Background This study aimed to explore the levels of circulating inflammatory factors CRP, IL-6, IL-10 and TNF- α based on the literature review. This study also examined the influence of single nucleotide polymorphism (SNP) sites on the susceptibility of abdominal aortic aneurysm (AAA) using meta-analysis and intended to provide additional information on pathogenesis of AAA research. Methods Electronic databases including PubMed and Web of Science were systemically searched to collect the information on AAA, inflammatory factors such as CRP, IL-6, IL-10, TNF- α and the SNP sites for data extraction. Altogether six SNPs in four genes (rs3091244, CRP; rs1800947, CRP; rs1205, CRP; rs1800795, IL-6; rs1800896, IL-10; and rs1800629, TNF) were assessed. Results This study enrolled altogether 41 relevant investigations involving 9,007 AAA patients to carry out meta-analysis. According to pooled analysis, circulating CRP and IL-6 levels were shown to be related to the AAA, while plasma IL-10 and TNF- α levels were not associated with AAA. The circulating CRP level standard mean difference (SMD) was 0.30 (95% confidence interval (CI): 0.17-0.43), the IL-6 level SMD was 0.34 (95% CI: 0.20-0.49), the IL-10 level SMD was -0.01 (95% CI: -0.09-0.06), and the TNF- α level SMD was 0.09 (95% CI: 0.00-0.19). Similarly, the odds ratio (OR) of rs3091244 (CRP) under the recessive gene model was 1.70 (95% CI: 1.13-2.57). In addition, individuals with A and T mutant genes at locus rs3091244 might have a higher tendency of AAA susceptibility than those with C allele. Consecutively, the OR was 0.91 (95% CI: 0.51-0.97) for rs1800795 (IL-6) locus in the allele model, and individuals with G mutant gene at locus rs1800795 (IL-6) might be less susceptible to AAA than those with C allele. Meanwhile, the rs1800896 (IL-10) locus had a positive association under the five statistical models, and individuals with A mutant gene at locus rs1800896 might have a higher susceptibility to AAA than those with G allele. Nevertheless, the rs1800947 (CRP), rs1205 (CRP), and rs1800629 (TNF) loci did not have positive correlation under the five statistical models, with no statistical significance. The results indicate that the gene polymorphisms at rs1800629, rs1800947, and rs1205 loci were not related to the AAA susceptibility. Conclusions Gene polymorphisms in certain known inflammatory mediators related to AAA susceptibility might serve as potential predictive biomarkers for clinical applications. Moreover, SNP of inflammatory mediators relevant to abdominal aortic aneurysmal formation and progression need extensive investigations to confirm these results.
Collapse
Affiliation(s)
- Hecheng Wang
- School of Life and Pharmaceutical Science, Dalian University of Technology, 124221 Panjin, Liaoning, China
| | - Zhenwu Zhong
- School of Life and Pharmaceutical Science, Dalian University of Technology, 124221 Panjin, Liaoning, China
| | - Deying Jiang
- Department of Vascular Surgery, Dalian Municipal Central Hospital, 116003 Dalian, Liaoning, China
| | - Hao Zhang
- School of Life and Pharmaceutical Science, Dalian University of Technology, 124221 Panjin, Liaoning, China
| | - Fanxing Yin
- School of Life and Pharmaceutical Science, Dalian University of Technology, 124221 Panjin, Liaoning, China
| | - Panpan Guo
- School of Life and Pharmaceutical Science, Dalian University of Technology, 124221 Panjin, Liaoning, China
| | - Junyu Chen
- School of Life and Pharmaceutical Science, Dalian University of Technology, 124221 Panjin, Liaoning, China
| | - Xinyu Zhu
- School of Life and Pharmaceutical Science, Dalian University of Technology, 124221 Panjin, Liaoning, China
| | - Kui You
- School of Ocean Science and Technology, Dalian University of Technology, 124221 Panjin, Liaoning, China
| | - Yanshuo Han
- School of Life and Pharmaceutical Science, Dalian University of Technology, 124221 Panjin, Liaoning, China
| | - Kun Liu
- Department of Cardiac Surgery, Affiliated Hospital of Guizhou Medical University, 550001 Guiyang, Guizhou, China
| |
Collapse
|
8
|
Zhu H, Tan J, Zhao Y, Wang Z, Wu Z, Li M. Potential Role of the Chemotaxis System in Formation and Progression of Intracranial Aneurysms Through Weighted Gene Co-Expression Network Analysis. Int J Gen Med 2022; 15:2217-2231. [PMID: 35250300 PMCID: PMC8893157 DOI: 10.2147/ijgm.s347420] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 02/22/2022] [Indexed: 12/21/2022] Open
Abstract
Background Intracranial aneurysm (IA) is the most common and is the main cause of spontaneous subarachnoid hemorrhage (SAH). The underlying molecular mechanisms for preventing IA progression have not been fully identified. Our research aimed to identify the key genes and critical pathways of IA through gene co-expression networks. Methods Gene Expression Omnibus (GEO) datasets GSE13353, GSE54083 and GSE75436 were used in the study. The genetic data were analyzed by weighted gene co-expression network analysis (WGCNA). Then the clinically significant modules were identified and the differentially expressed genes (DEGs) with the genes were intersected in these modules. GO (gene ontology) and KEGG (Kyoto Gene and Genomic Encyclopedia) were used for gene enrichment analysis to determine the function or pathway. In addition, the composition of immune cells was analyzed by CIBERSORT algorithm. Finally, the hub genes and key genes were identified by GSE122897. Results A total of 266 DEGs and two modules with clinical significance were identified. The inflammatory response and immune response were identified by GO and KEGG. CCR5, CCL4, CCL20, and FPR3 were the key genes in the module correlated with IA. The proportions of infiltrating immune cells in IA and normal tissues were different, especially in terms of macrophages and mast cells. Conclusion The chemotactic system has been identified as a key pathway of IA, and interacting macrophages may regulate this pathological process.
Collapse
Affiliation(s)
- Huaxin Zhu
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, People’s Republic of China
| | - Jiacong Tan
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, People’s Republic of China
| | - Yeyu Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, People’s Republic of China
| | - Zhihua Wang
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, People’s Republic of China
| | - Zhiwu Wu
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, People’s Republic of China
| | - Meihua Li
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, People’s Republic of China
- Correspondence: Meihua Li, Email
| |
Collapse
|
9
|
Piqueras L, Sanz MJ. Angiotensin II and leukocyte trafficking: New insights for an old vascular mediator. Role of redox-signaling pathways. Free Radic Biol Med 2020; 157:38-54. [PMID: 32057992 DOI: 10.1016/j.freeradbiomed.2020.02.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 01/27/2020] [Accepted: 02/03/2020] [Indexed: 12/20/2022]
Abstract
Inflammation and activation of the immune system are key molecular and cellular events in the pathogenesis of cardiovascular diseases, including atherosclerosis, hypertension-induced target-organ damage, and abdominal aortic aneurysm. Angiotensin II (Ang-II) is the main effector peptide hormone of the renin-angiotensin system. Beyond its role as a potent vasoconstrictor and regulator of blood pressure and fluid homeostasis, Ang-II is intimately involved in the development of vascular lesions in cardiovascular diseases through the activation of different immune cells. The migration of leukocytes from circulation to the arterial subendothelial space is a crucial immune response in lesion development that is mediated through a sequential and coordinated cascade of leukocyte-endothelial cell adhesive interactions involving an array of cell adhesion molecules present on target leukocytes and endothelial cells and the generation and release of chemoattractants that activate and guide leukocytes to sites of emigration. In this review, we outline the key events of Ang-II participation in the leukocyte recruitment cascade, the underlying mechanisms implicated, and the corresponding redox-signaling pathways. We also address the use of inhibitor drugs targeting the effects of Ang-II in the context of leukocyte infiltration in these cardiovascular pathologies, and examine the clinical data supporting the relevance of blocking Ang-II-induced vascular inflammation.
Collapse
Affiliation(s)
- Laura Piqueras
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain; Institute of Health Research INCLIVA University Clinic Hospital of Valencia, Valencia, Spain; CIBERDEM-Spanish Biomedical Research Center in Diabetes and Associated Metabolic Disorders, Carlos III Health Institute, Spanish Ministry of Health, Madrid, Spain.
| | - Maria-Jesus Sanz
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain; Institute of Health Research INCLIVA University Clinic Hospital of Valencia, Valencia, Spain; CIBERDEM-Spanish Biomedical Research Center in Diabetes and Associated Metabolic Disorders, Carlos III Health Institute, Spanish Ministry of Health, Madrid, Spain.
| |
Collapse
|
10
|
Characterization and Significance of Monocytes in Acute Stanford Type B Aortic Dissection. J Immunol Res 2020; 2020:9670360. [PMID: 32509885 PMCID: PMC7245667 DOI: 10.1155/2020/9670360] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 02/05/2020] [Indexed: 01/16/2023] Open
Abstract
Acute aortic dissection (AAD) is one of the most common fatal diseases noted in vascular surgery. Human monocytes circulate in dynamic equilibrium and display a considerable heterogeneity. However, the role of monocytes in AAD remains elusive. In our recent study, we firstly obtained blood samples from 22 patients with Stanford type B AAD and 44 age-, sex-, and comorbidity-matched control subjects. And the monocyte proportions were evaluated by flow cytometry. Results showed that the percentage of total CD14+ monocytes in the blood samples of Stanford AAD patients was increased significantly compared with that of normal volunteers (P < 0.0005), and the absolute numbers of CD14brightCD16+ and CD14brightCD16− monocytes both increased significantly regardless of the percentage of PBMC or CD14+ cells, while CD14dimCD16+ monocytes displayed the opposite tendency. However, the percentage of CD14+ cells and its three subsets demonstrated no correlation with D-dimer (DD) and C-reactive protein (CRP). Then, blood mononuclear cell (PBMC) samples were collected by Ficoll density gradient centrifugation, followed with CD14+ magnetic bead sorting. After the purity of CD14+ cells was validated over 90%, AAD-related genes were concentrated in CD14+ monocytes. There were no significant differences observed with regard to the mRNA expression levels of MMP1 (P = 0.0946), MMP2 (P = 0.3941), MMP9 (P = 0.2919), IL-6 (P = 0.4223), and IL-10 (P = 0.3375) of the CD14+ monocytes in Stanford type B AAD patients compared with those of normal volunteers. The expression levels of IL-17 (P < 0.05) was higher in Stanford type B AAD patients, while the expression levels of TIMP1(P<0.05), TIMP2(P<0.01), TGF-β1 (P < 0.01), SMAD3 (P < 0.01), ACTA2 (P < 0.001), and ADAMTS-1 (P < 0.001) decreased. The data suggested that monocytes might play an important role in the development of Stanford type B AAD. Understanding of the production, differentiation, and function of monocyte subsets might dictate future therapeutic avenues for Stanford type B AAD treatment and can aid the identification of novel biomarkers or potential therapeutic targets for decreasing inflammation in AAD.
Collapse
|
11
|
Jones GT, Marsman J, Bhat B, Phillips VL, Chatterjee A, Rodger EJ, Williams MJA, van Rij AM, McCormick SPA. DNA methylation profiling identifies a high effect genetic variant for lipoprotein(a) levels. Epigenetics 2020; 15:949-958. [PMID: 32237968 DOI: 10.1080/15592294.2020.1739797] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Changes in whole blood DNA methylation levels at several CpG sites have been associated with circulating blood lipids, specifically high-density lipoprotein and triglycerides. This study performs a discovery and validation epigenome-wide association study (EWAS) for circulating lipoprotein(a) [Lp(a)], an independent risk factor for cardiovascular diseases. Whole-blood DNA methylation profiles were assessed in a cohort of 1020 elderly individuals using the Illumina EPIC array and independent validation in 359 elderly males using the Illumina 450 k array. Plasma Lp(a) was measured using an apolipoprotein(a)-size-independent ELISA. Epigenome-wide rank regression analysis identified and validated a single CpG site, cg17028067 located in intron 1 of the LPA gene, that was significantly associated with plasma Lp(a) levels after correction for multiple testing. Genotyping of the site identified a relatively uncommon SNP (rs76735376, MAF <0.02) at the CpG site that largely explained the observed methylation effect. Rs76735376 is an expression quantitative trait loci for the LPA gene and could affect expression by altering enhancer activity. This EWAS for plasma Lp(a) identified a single CpG site within LPA. This association is due to an uncommon, but highly effective genetic variant, which was not in significant linkage disequilibrium with other variants known to influence Lp(a) levels or apo(a) isoform size. This study highlights the utility of CpG site methylation to identify potentially important genetic associations that would not be readily apparent in a comparable size genetic association study.
Collapse
Affiliation(s)
- Gregory T Jones
- Departments of Surgical Sciences, University of Otago , Dunedin, New Zealand
| | - Judith Marsman
- Departments of Surgical Sciences, University of Otago , Dunedin, New Zealand
| | - Basharat Bhat
- Departments of Surgical Sciences, University of Otago , Dunedin, New Zealand
| | - Victoria L Phillips
- Departments of Surgical Sciences, University of Otago , Dunedin, New Zealand
| | | | - Euan J Rodger
- Pathology, University of Otago , Dunedin, New Zealand
| | | | - André M van Rij
- Departments of Surgical Sciences, University of Otago , Dunedin, New Zealand
| | | |
Collapse
|
12
|
Jabłońska A, Neumayer C, Bolliger M, Burghuber C, Klinger M, Demyanets S, Nanobachvili J, Huk I. Insight into the expression of toll-like receptors 2 and 4 in patients with abdominal aortic aneurysm. Mol Biol Rep 2020; 47:2685-2692. [PMID: 32146682 DOI: 10.1007/s11033-020-05366-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 02/29/2020] [Indexed: 12/15/2022]
Abstract
An abdominal aortic aneurysm (AAA) is a relatively common, life-threatening disease prevalent in persons over the age of 65. In recent years, an increasing number of studies have suggested that pattern-recognition receptors (PRRs), including Toll-like receptors (TLRs), may serve as important regulators in the development of AAAs. In this study, we evaluated the TLR2 and TLR4 expression in the aortic wall and blood of patients with AAA. The TLR2 and TLR4 mRNA expression were significantly higher in the blood of patients with AAA than in the blood of healthy volunteers (p = 0.009 and p = 0.010, respectively). The expression of TLR2 and TLR4 transcripts was also higher in the blood compared with the aortic wall tissue of AAA patients (p = 0.001 for both). Higher TLR2 protein expression was observed in the aortic wall of AAA patients compared with the blood (p = 0.026). A significantly higher concentration of TNF-α and IL-4 in patients with AAA than in healthy volunteers (p < 0.001 for both) was noticed. This study suggests that TLR2 may play a role in the inflammatory response in the aorta, both locally and systemically, in patients with AAA.
Collapse
Affiliation(s)
- Agnieszka Jabłońska
- Laboratory of Virology, Institute of Medical Biology, Polish Academy of Sciences, 106 St., 93-232, Lodz, Poland. .,Division of Vascular Surgery, Department of Surgery, Medical University of Vienna, Vienna General Hospital, Vienna, Austria.
| | - Christoph Neumayer
- Division of Vascular Surgery, Department of Surgery, Medical University of Vienna, Vienna General Hospital, Vienna, Austria
| | - Michael Bolliger
- Division of Vascular Surgery, Department of Surgery, Medical University of Vienna, Vienna General Hospital, Vienna, Austria
| | - Christopher Burghuber
- Division of Vascular Surgery, Department of Surgery, Medical University of Vienna, Vienna General Hospital, Vienna, Austria
| | - Markus Klinger
- Division of Vascular Surgery, Department of Surgery, Medical University of Vienna, Vienna General Hospital, Vienna, Austria
| | - Svitlana Demyanets
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Josif Nanobachvili
- Division of Vascular Surgery, Department of Surgery, Medical University of Vienna, Vienna General Hospital, Vienna, Austria
| | - Ihor Huk
- Division of Vascular Surgery, Department of Surgery, Medical University of Vienna, Vienna General Hospital, Vienna, Austria
| |
Collapse
|
13
|
Stienen S, Ferreira JP, Kobayashi M, Preud'homme G, Dobre D, Machu JL, Duarte K, Bresso E, Devignes MD, López N, Girerd N, Aakhus S, Ambrosio G, Brunner-La Rocca HP, Fontes-Carvalho R, Fraser AG, van Heerebeek L, Heymans S, de Keulenaer G, Marino P, McDonald K, Mebazaa A, Papp Z, Raddino R, Tschöpe C, Paulus WJ, Zannad F, Rossignol P. Enhanced clinical phenotyping by mechanistic bioprofiling in heart failure with preserved ejection fraction: insights from the MEDIA-DHF study (The Metabolic Road to Diastolic Heart Failure). Biomarkers 2020; 25:201-211. [PMID: 32063068 DOI: 10.1080/1354750x.2020.1727015] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Background: Heart failure with preserved ejection fraction (HFpEF) is a heterogeneous syndrome for which clear evidence of effective therapies is lacking. Understanding which factors determine this heterogeneity may be helped by better phenotyping. An unsupervised statistical approach applied to a large set of biomarkers may identify distinct HFpEF phenotypes.Methods: Relevant proteomic biomarkers were analyzed in 392 HFpEF patients included in Metabolic Road to Diastolic HF (MEDIA-DHF). We performed an unsupervised cluster analysis to define distinct phenotypes. Cluster characteristics were explored with logistic regression. The association between clusters and 1-year cardiovascular (CV) death and/or CV hospitalization was studied using Cox regression.Results: Based on 415 biomarkers, we identified 2 distinct clusters. Clinical variables associated with cluster 2 were diabetes, impaired renal function, loop diuretics and/or betablockers. In addition, 17 biomarkers were higher expressed in cluster 2 vs. 1. Patients in cluster 2 vs. those in 1 experienced higher rates of CV death/CV hospitalization (adj. HR 1.93, 95% CI 1.12-3.32, p = 0.017). Complex-network analyses linked these biomarkers to immune system activation, signal transduction cascades, cell interactions and metabolism.Conclusion: Unsupervised machine-learning algorithms applied to a wide range of biomarkers identified 2 HFpEF clusters with different CV phenotypes and outcomes. The identified pathways may provide a basis for future research.Clinical significanceMore insight is obtained in the mechanisms related to poor outcome in HFpEF patients since it was demonstrated that biomarkers associated with the high-risk cluster were related to the immune system, signal transduction cascades, cell interactions and metabolismBiomarkers (and pathways) identified in this study may help select high-risk HFpEF patients which could be helpful for the inclusion/exclusion of patients in future trials.Our findings may be the basis of investigating therapies specifically targeting these pathways and the potential use of corresponding markers potentially identifying patients with distinct mechanistic bioprofiles most likely to respond to the selected mechanistically targeted therapies.
Collapse
Affiliation(s)
- Susan Stienen
- CHRU de Nancy, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), INSERM U1116, Centre d'Investigation Clinique et Plurithématique 1433, INSERM, Université de Lorraine, Nancy, France
| | - João Pedro Ferreira
- CHRU de Nancy, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), INSERM U1116, Centre d'Investigation Clinique et Plurithématique 1433, INSERM, Université de Lorraine, Nancy, France.,Department of Physiology and Cardiothoracic Surgery, Cardiovascular Research and Development Unit, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Masatake Kobayashi
- CHRU de Nancy, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), INSERM U1116, Centre d'Investigation Clinique et Plurithématique 1433, INSERM, Université de Lorraine, Nancy, France
| | - Gregoire Preud'homme
- CHRU de Nancy, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), INSERM U1116, Centre d'Investigation Clinique et Plurithématique 1433, INSERM, Université de Lorraine, Nancy, France
| | - Daniela Dobre
- CHRU de Nancy, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), INSERM U1116, Centre d'Investigation Clinique et Plurithématique 1433, INSERM, Université de Lorraine, Nancy, France.,Clinical research and Investigation Unit, Psychotherapeutic Center of Nancy, Laxou, France
| | - Jean-Loup Machu
- CHRU de Nancy, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), INSERM U1116, Centre d'Investigation Clinique et Plurithématique 1433, INSERM, Université de Lorraine, Nancy, France
| | - Kevin Duarte
- CHRU de Nancy, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), INSERM U1116, Centre d'Investigation Clinique et Plurithématique 1433, INSERM, Université de Lorraine, Nancy, France
| | - Emmanuel Bresso
- Equipe CAPSID, LORIA (CNRS, Inria NGE, Université de Lorraine), Vandoeuvre-lès-Nancy, France
| | | | - Natalia López
- Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain
| | - Nicolas Girerd
- CHRU de Nancy, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), INSERM U1116, Centre d'Investigation Clinique et Plurithématique 1433, INSERM, Université de Lorraine, Nancy, France
| | - Svend Aakhus
- Department of Cardiology and Institute for Surgical Research, Oslo University Hospital, Oslo, Norway.,ISB, Norwegian University of Science and Technology, Trondheim, Norway
| | - Giuseppe Ambrosio
- Division of Cardiology, University of Perugia School of Medicine, Perugia, Italy
| | | | - Ricardo Fontes-Carvalho
- Department of Surgery and Physiology, Cardiovascular Research Unit (UnIC), Faculty of Medicine, University of Porto, Porto, Portugal
| | - Alan G Fraser
- Wales Heart Research Institute, Cardiff University, Cardiff, UK
| | - Loek van Heerebeek
- Department of Cardiology, Onze Lieve Vrouwe Gasthuis, Amsterdam, the Netherlands
| | - Stephane Heymans
- Department of Cardiology, CARIM School for Cardiovascular Diseases Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands.,Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology, Leuven, Belgium.,William Harvey Research Institute, Barts Heart Centre, Queen Mary University of London, London, UK
| | - Gilles de Keulenaer
- Laboratory of Physiopharmacology, Antwerp University, and ZNA Hartcentrum, Antwerp, Belgium
| | - Paolo Marino
- Clinical Cardiology, Università del Piemonte Orientale, Department of Translational Medicine, Azienda Ospedaliero Universitaria "Maggiore della Carità", Novara, Italy
| | - Kenneth McDonald
- School of Medicine and Medical Sciences, St Michael's Hospital Dun Laoghaire Co. Dublin, Dublin, Ireland
| | - Alexandre Mebazaa
- Department of Anaesthesiology and Critical Care Medicine, Saint Louis and Lariboisière University Hospitals and INSERM UMR-S 942, Paris, France
| | - Zoltàn Papp
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Riccardo Raddino
- Department of Cardiology, Spedali Civili di Brescia, Brescia, Italy
| | - Carsten Tschöpe
- Department of Cardiology, Campus Virchow-Klinikum, C, Harite Universitaetsmedizin Berlin, Berlin Institute of Health - Center for Regenerative Therapies (BIH-BCRT), and the German Center for Cardiovascular Research (DZHK; Berlin partner site), Berlin, Germany
| | - Walter J Paulus
- Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Faiez Zannad
- CHRU de Nancy, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), INSERM U1116, Centre d'Investigation Clinique et Plurithématique 1433, INSERM, Université de Lorraine, Nancy, France
| | - Patrick Rossignol
- CHRU de Nancy, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), INSERM U1116, Centre d'Investigation Clinique et Plurithématique 1433, INSERM, Université de Lorraine, Nancy, France
| |
Collapse
|
14
|
Abstract
Dissections or ruptures of aortic aneurysms remain a leading cause of death in the developed world, with the majority of deaths being preventable if individuals at risk are identified and properly managed. Genetic variants predispose individuals to these aortic diseases. In the case of thoracic aortic aneurysm and dissections (thoracic aortic disease), genetic data can be used to identify some at-risk individuals and dictate management of the associated vascular disease. For abdominal aortic aneurysms, genetic associations have been identified, which provide insight on the molecular pathogenesis but cannot be used clinically yet to identify individuals at risk for abdominal aortic aneurysms. This compendium will discuss our current understanding of the genetic basis of thoracic aortic disease and abdominal aortic aneurysm disease. Although both diseases share several pathogenic similarities, including proteolytic elastic tissue degeneration and smooth muscle dysfunction, they also have several distinct differences, including population prevalence and modes of inheritance.
Collapse
Affiliation(s)
- Amélie Pinard
- From the Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School; University of Texas Health Science Center at Houston (A.P., D.M.M.)
| | - Gregory T Jones
- Department of Surgical Sciences, Dunedin School of Medicine, University of Otago, New Zealand (G.T.J.)
| | - Dianna M Milewicz
- From the Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School; University of Texas Health Science Center at Houston (A.P., D.M.M.)
| |
Collapse
|
15
|
Sagan A, Mikolajczyk TP, Mrowiecki W, MacRitchie N, Daly K, Meldrum A, Migliarino S, Delles C, Urbanski K, Filip G, Kapelak B, Maffia P, Touyz R, Guzik TJ. T Cells Are Dominant Population in Human Abdominal Aortic Aneurysms and Their Infiltration in the Perivascular Tissue Correlates With Disease Severity. Front Immunol 2019; 10:1979. [PMID: 31552015 PMCID: PMC6736986 DOI: 10.3389/fimmu.2019.01979] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 08/05/2019] [Indexed: 12/22/2022] Open
Abstract
Abdominal Aortic Aneurysm (AAA) is a major cause of cardiovascular mortality. Adverse changes in vascular phenotype act in concert with chronic inflammation to promote AAA progression. Perivascular adipose tissue (PVAT) helps maintain vascular homeostasis but when inflamed and dysfunctional, can also promote vascular pathology. Previous studies suggested that PVAT may be an important site of vascular inflammation in AAA; however, a detailed assessment of leukocyte populations in human AAA, their anatomic location in the vessel wall and correlation to AAA size remain undefined. Accordingly, we performed in depth immunophenotyping of cells infiltrating the pathologically altered perivascular tissue (PVT) and vessel wall in AAA samples at the site of maximal dilatation (n = 51 patients). Flow cytometry revealed that T cells, rather than macrophages, are the major leukocyte subset in AAA and that their greatest accumulations occur in PVT. Both CD4+ and CD8+ T cell populations are highly activated in both compartments, with CD4+ T cells displaying the highest activation status within the AAA wall. Finally, we observed a positive relationship between T cell infiltration in PVT and AAA wall. Interestingly, only PVT T cell infiltration was strongly related to tertiles of AAA size. In summary, this study highlights an important role for PVT as a reservoir of T lymphocytes and potentially as a key site in modulating the underlying inflammation in AAA.
Collapse
Affiliation(s)
- Agnieszka Sagan
- BHF Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom.,Department of Internal and Agricultural Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Tomasz P Mikolajczyk
- Department of Internal and Agricultural Medicine, Jagiellonian University Medical College, Kraków, Poland.,Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Wojciech Mrowiecki
- Department of Vascular Surgery, CUMRiK, University Hospital, Kraków, Poland
| | - Neil MacRitchie
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Kevin Daly
- Department of Vascular Surgery, Queen Elizabeth University Hospital, Glasgow, United Kingdom
| | - Alan Meldrum
- Department of Vascular Surgery, Queen Elizabeth University Hospital, Glasgow, United Kingdom
| | - Serena Migliarino
- BHF Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Christian Delles
- BHF Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Karol Urbanski
- Department of Internal and Agricultural Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Grzegorz Filip
- Department of Cardiovascular Surgery and Transplantology, John Paul II Hospital, Kraków, Poland
| | - Boguslaw Kapelak
- Department of Cardiovascular Surgery and Transplantology, John Paul II Hospital, Kraków, Poland.,Institute of Cardiology, Jagiellonian University Medical College, Kraków, Poland
| | - Pasquale Maffia
- BHF Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom.,Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom.,Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Rhian Touyz
- BHF Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Tomasz J Guzik
- BHF Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom.,Department of Internal and Agricultural Medicine, Jagiellonian University Medical College, Kraków, Poland
| |
Collapse
|
16
|
Establishment of Novel Murine Model showing Vascular Inflammation-derived Cognitive Dysfunction. Sci Rep 2019; 9:4023. [PMID: 30858535 PMCID: PMC6411753 DOI: 10.1038/s41598-019-40726-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 02/21/2019] [Indexed: 12/14/2022] Open
Abstract
Inflammation is a critical feature of aging and its related diseases, including cardiovascular diseases. Recent epidemiological studies demonstrated that abdominal aortic aneurysm (AAA), an aging-related vascular pathological condition, is associated with cognitive decline. However, the underlying mechanism, especially the role of vascular inflammation, is largely unknown because of lack of an available animal model. In this study, we examined whether vascular inflammation affects synaptic and cognitive dysfunction, using an AAA mouse model. In young (3 months) and middle-aged (12 months) C57BL/6J mice, AAA was induced by angiotensin II infusion with calcium chloride application. After 4 weeks of induction, aortic diameter was significantly increased and excessive Mac3-positive inflammatory cells infiltrated the destroyed aorta in middle-aged mice. AAA-induced middle-aged mice further exhibited cognitive impairment. Neuronal loss was observed in the CA3 region of the hippocampus. IBA1/MHCII-double-positive microglia activation was also seen in the hippocampus, suggesting that vascular inflammation drives neuroinflammation and subsequent cognitive dysfunction. Furthermore, we found that senescence-accelerated mice prone 8 exhibited robust AAA formation and a marked decrease of cognitive and synaptic function in the hippocampus mediated by inflammation. In conclusion, this novel murine model convincingly suggested the occurrence of vascular inflammation-derived cognitive dysfunction.
Collapse
|
17
|
Nie H, Wang HX, Tian C, Ren HL, Li FD, Wang CY, Li HH, Zheng YH. Chemokine (C-X-C motif) receptor 2 blockade by SB265610 inhibited angiotensin II-induced abdominal aortic aneurysm in Apo E -/- mice. Heart Vessels 2018; 34:875-882. [PMID: 30535755 DOI: 10.1007/s00380-018-1301-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 11/09/2018] [Indexed: 11/29/2022]
Abstract
Inflammation plays a critical role in the development of abdominal aortic aneurysm (AAA). Chemokine receptor CXCR2 mediates inflammatory cell chemotaxis in several diseases. However, the role of CXCR2 in AAA and the underlying mechanisms remain unknown. In this study, we found that the CXCR2 expressions in AAA tissues from human and angiotensin II (Ang II)-infused apolipoprotein E knockout (Apo E-/-) mice were significantly increased. The pharmacological inhibition of CXCR2 (SB265610) markedly reduced Ang II-induced AAA formation. Furthermore, SB265610 treatment significantly reduced collagen deposition, elastin degradation, the metal matrix metalloprotease expression and accumulation of macrophage cells. In conclusion, these results showed CXCR2 plays a pathogenic role in AAA formation. Inhibition of CXCR2 pathway may represent a novel therapeutic approach to treat AAA.
Collapse
Affiliation(s)
- Hao Nie
- Department of Vascular Surgery, Peking Union Medical College Hospital, Beijing, 100005, People's Republic of China
| | - Hong-Xia Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Cui Tian
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Hua-Liang Ren
- Department of Vascular Surgery, Peking Union Medical College Hospital, Beijing, 100005, People's Republic of China
| | - Fang-Da Li
- Department of Vascular Surgery, Peking Union Medical College Hospital, Beijing, 100005, People's Republic of China
| | - Chao-Yu Wang
- Department of Vascular Surgery, Peking Union Medical College Hospital, Beijing, 100005, People's Republic of China
| | - Hui-Hua Li
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, People's Republic of China.
| | - Yue-Hong Zheng
- Department of Vascular Surgery, Peking Union Medical College Hospital, Beijing, 100005, People's Republic of China.
| |
Collapse
|
18
|
Heme oxygenase-1 deficiency exacerbates angiotensin II-induced aortic aneurysm in mice. Oncotarget 2018; 7:67760-67776. [PMID: 27626316 PMCID: PMC5356517 DOI: 10.18632/oncotarget.11917] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 09/02/2016] [Indexed: 01/23/2023] Open
Abstract
Abdominal aortic aneurysm (AAA) is a chronic but often fatal disease in elderly population. Heme oxygenase-1 (HO-1) is a stress response protein with antioxidative and anti-inflammatory properties. HO-1 has been shown to protect against atherogenesis and arterial intimal thickening. Emerging evidences suggest that AAA and arterial occlusive disease have distinct pathogenic mechanisms. Thus, in this study we investigated the role of HO-1 in angiotensin II-induced AAA formation in HO-1+/+apoE−/− and HO-1−/−apoE−/− mice. We found that complete loss of HO-1 increased AAA incidence and rupture rate, and drastically increased aneurysmal area and severity, accompanied with severe elastin degradation and medial degeneration. Interestingly, we often observed not only AAA but also thoracic aortic aneurysm in HO-1−/−apoE−/− mice. Furthermore, reactive oxygen species levels, vascular smooth muscle cell (VSMC) loss, macrophage infiltration, matrix metalloproteinase (MMP) activity were markedly enhanced in the aneurysmal aortic wall in HO-1−/−apoE−/− mice. In addition, HO-1−/−apoE−/− VSMCs were more susceptible to oxidant-induced cell death and macrophages from HO-1−/−apoE−/− mice had aggravated responses to angiotensin II with substantial increases in inflammatory cytokine productions and MMP9 activity. Taken together, our results demonstrate the essential roles of HO-1 in suppressing the pathogenesis of AAA. Targeting HO-1 might be a promising therapeutic strategy for AAA.
Collapse
|
19
|
Wang Y, Shen G, Wang H, Yao Y, Sun Q, Jing B, Liu G, Wu J, Yuan C, Liu S, Liu X, Li S, Li H. Association of high sensitivity C-reactive protein and abdominal aortic aneurysm: a meta-analysis and systematic review. Curr Med Res Opin 2017; 33:2145-2152. [PMID: 28699805 DOI: 10.1080/03007995.2017.1354825] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
OBJECTIVE To evaluate the association of high sensitivity C-reactive protein (hsCRP) with the presence of abdominal aortic aneurysm (AAA). METHODS Medline, Cochrane, Embase, and Google Scholar databases were searched until 22 June 2016 using the keywords predictive factors, biomarkers, abdominal aortic aneurysm, prediction, high sensitivity C-reactive protein, and hsCRP. Prospective studies, retrospective studies, and cohort studies were included. RESULTS Twelve case-control studies were included in the meta-analysis with a total of 8345 patients (1977 in the AAA group and 6368 in the control group). The pooled results showed that AAA patients had higher hsCRP value than the control group (difference in means = 1.827, 95% CI = 0.010 to 3.645, p = .049). Subgroup analysis found AAA patients with medium or small aortic diameter (<50 mm) had higher hsCRP plasma levels than the control group (difference in means = 1.301, 95% CI = 0.821 to 1.781, p < .001). In patients with large aortic diameter (≥50 mm), no difference was observed in hsCRP levels between the AAA and control groups (difference in means = 1.769, 95% CI = -1.387 to 4.925, p = .272). Multi-regression analysis found the difference in means of hsCRP plasma levels between AAA and control groups decreased as aortic diameter increased (slope = -0.04, p < .001), suggesting that hsCRP levels may be inversely associated with increasing aneurysm size. CONCLUSIONS Our findings suggest that hsCRP levels may possibly be used as a diagnostic biomarker for AAA patients with medium or small aortic diameter but not for AAA patients with large aortic diameter. The correlation between serum hsCRP level and AAA aneurysm is not conclusive due to the small number of included articles and between-study heterogeneity.
Collapse
Affiliation(s)
- Yunpeng Wang
- a Department of Vascular Surgery , The First Affiliated Hospital of Harbin Medical University , Harbin , China
| | - Guanghui Shen
- a Department of Vascular Surgery , The First Affiliated Hospital of Harbin Medical University , Harbin , China
| | - Haiyang Wang
- a Department of Vascular Surgery , The First Affiliated Hospital of Harbin Medical University , Harbin , China
| | - Ye Yao
- a Department of Vascular Surgery , The First Affiliated Hospital of Harbin Medical University , Harbin , China
| | - Qingfeng Sun
- a Department of Vascular Surgery , The First Affiliated Hospital of Harbin Medical University , Harbin , China
| | - Bao Jing
- a Department of Vascular Surgery , The First Affiliated Hospital of Harbin Medical University , Harbin , China
| | - Gaoyan Liu
- a Department of Vascular Surgery , The First Affiliated Hospital of Harbin Medical University , Harbin , China
| | - Jia Wu
- a Department of Vascular Surgery , The First Affiliated Hospital of Harbin Medical University , Harbin , China
| | - Chao Yuan
- a Department of Vascular Surgery , The First Affiliated Hospital of Harbin Medical University , Harbin , China
| | - Siqi Liu
- a Department of Vascular Surgery , The First Affiliated Hospital of Harbin Medical University , Harbin , China
| | - Xinyu Liu
- a Department of Vascular Surgery , The First Affiliated Hospital of Harbin Medical University , Harbin , China
| | - Shiyong Li
- a Department of Vascular Surgery , The First Affiliated Hospital of Harbin Medical University , Harbin , China
| | - Haocheng Li
- a Department of Vascular Surgery , The First Affiliated Hospital of Harbin Medical University , Harbin , China
| |
Collapse
|