1
|
Yang HQ, Li ZW, Dong XX, Zhang JX, Shan J, Wang MJ, Yang J, Li MH, Wang J, Zhao HM. Vinpocetine alleviates the abdominal aortic aneurysm progression via VSMCs SIRT1-p21 signaling pathway. Acta Pharmacol Sin 2025; 46:96-106. [PMID: 39179867 PMCID: PMC11696035 DOI: 10.1038/s41401-024-01358-w] [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: 03/28/2024] [Accepted: 07/10/2024] [Indexed: 08/26/2024] Open
Abstract
Abdominal aortic aneurysm (AAA) is a degenerative disease that caused mortality in people aged >65. Senescence plays a critical role in AAA pathogenesis. Advances in AAA repair techniques have occurred, but a remaining priority is therapies to limit AAA growth and rupture. Our Previous study found cyclic nucleotide phosphodiesterase 1C (PDE1C) exacerbate AAA through aggravate vascular smooth muscle cells (VSMCs) senescence by downregulating Sirtuin1 (SIRT1) expression and activity. Vinpocetine as a selective inhibitor of PDE1 and a clinical medication for cerebral vasodilation, it is unclear whether vinpocetine can rely on SIRT1 to alleviate AAA. This study showed that pre-treatment with vinpocetine remarkably prevented aneurysmal dilation and reduced aortic rupture in elastase-induced AAA mice. In addition, the elastin degradation, MMP (matrix metalloproteinase) activity, macrophage infiltration, ROS production, collagen fibers remodeling, and VSMCs senescence were decreased in AAA treated with vinpocetine. While these effects were unable to exert in VSMCs-specific SIRT1 knockout AAA mice. Accordingly, we revealed that vinpocetine suppressed migration, proliferation, and senescence in VSMCs. Moreover, vinpocetine reduced SIRT1 degradation by inhibiting lysosome-mediated autophagy. In conclusion, this study indicated that vinpocetine may be as a potential drug for therapy AAA through alleviate VSMCs senescence via the SIRT1-dependent pathway.
Collapse
MESH Headings
- Animals
- Sirtuin 1/metabolism
- Vinca Alkaloids/pharmacology
- Vinca Alkaloids/therapeutic use
- Aortic Aneurysm, Abdominal/drug therapy
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/pathology
- Signal Transduction/drug effects
- Mice, Inbred C57BL
- Male
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Mice
- Mice, Knockout
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Autophagy/drug effects
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Cellular Senescence/drug effects
- Cells, Cultured
Collapse
Affiliation(s)
- Hong-Qin Yang
- Baotou Medical College, Baotou, 014040, Inner Mongolia Autonomous Region, China
- State Key Laboratory of Complex, Severe, and Rare Diseases, Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Peking Union Medical College Hospital, Beijing, 100005, China
| | - Zhi-Wei Li
- State Key Laboratory of Complex, Severe, and Rare Diseases, Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Peking Union Medical College Hospital, Beijing, 100005, China
| | - Xi-Xi Dong
- State Key Laboratory of Complex, Severe, and Rare Diseases, Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Peking Union Medical College Hospital, Beijing, 100005, China
| | - Jia-Xin Zhang
- State Key Laboratory of Complex, Severe, and Rare Diseases, Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Peking Union Medical College Hospital, Beijing, 100005, China
| | - Jin Shan
- State Key Laboratory of Complex, Severe, and Rare Diseases, Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Peking Union Medical College Hospital, Beijing, 100005, China
| | - Min-Jie Wang
- Medical Experimental Center, School of Basic Medical Sciences, Inner Mongolia Medical University, Chilechuan dairy economic development zone, Hohhot, Inner Mongolia Autonomous Region, Hohhot, 010110, China
| | - Jing Yang
- Baotou Medical College, Baotou, 014040, Inner Mongolia Autonomous Region, China.
| | - Min-Hui Li
- Baotou Medical College, Baotou, 014040, Inner Mongolia Autonomous Region, China.
| | - Jing Wang
- State Key laboratory of Respiratory Health and Multimorbidity, Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China.
| | - Hong-Mei Zhao
- State Key Laboratory of Complex, Severe, and Rare Diseases, Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Peking Union Medical College Hospital, Beijing, 100005, China.
| |
Collapse
|
2
|
Qi X, Gao L, Qi L. Genetic determinants of telomere length and risk of aneurysmal subarachnoid hemorrhage: a bidirectional two-sample mendelian randomization study. Int J Neurosci 2024:1-9. [PMID: 39523870 DOI: 10.1080/00207454.2024.2414285] [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: 08/05/2024] [Revised: 09/10/2024] [Accepted: 10/04/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Our objective is to investigate the potential causal relationship between telomere length (TL) and aneurysmal subarachnoid hemorrhage (aSAH) and intracranial aneurysms (IAs) by conducting a bidirectional two-sample Mendelian Randomization (MR) study. METHODS We utilized publicly available summary data from genome-wide association studies (GWAS) for comprehensive analysis. Telomere length-associated data were sourced from the Epidemiology Unit (IEU) GWAS database (n = 472,174), while data pertaining to intracranial aneurysms were derived from a GWAS meta-analysis conducted by Bakker et al. encompassing aneurysmal subtypes including aSAH (n = 77,074), IAs (n = 79,429), and unruptured intracranial aneurysms (uIA) (n = 74,004), all sampled from European populations. The primary method for MR analysis employed was the Inverse Variance Weighted (IVW) method. Additionally, we conducted various sensitivity analyses to assess the heterogeneity and pleiotropy of study findings. Reverse MR analysis was employed to explore potential reverse causality. RESULTS In the forward MR analysis, the IVW method indicated a negative association between TL and aSAH (OR = 0.636, 95% CI: 0.459-0.883, p = 0.006) as well as IAs (OR = 0.670, 95% CI: 0.499-0.900, p = 0.0079). There was no evidence of heterogeneity or horizontal pleiotropy in the forward MR analysis. Reverse MR analysis did not reveal any causal relationship between aSAH, IAs, uIA and TL. CONCLUSIONS In European populations, there exists a causal relationship between longer TL and reduced risks of aSAH and IAs Further research is warranted to elucidate the underlying mechanisms and the potential of TL as an intervention target for lowering the incidence of aSAH and IAs.
Collapse
Affiliation(s)
- Xiangjia Qi
- Department of Neurology, Liaocheng People's Hospital, Liaocheng, China
| | - Liqian Gao
- Department of Neurology, Liaocheng People's Hospital, Liaocheng, China
| | - Lifeng Qi
- Department of Neurology, Liaocheng People's Hospital, Liaocheng, China
| |
Collapse
|
3
|
Sani A, Abdullahi IL, Khan MI, Cao C. Analyses of oxidative DNA damage among coal vendors via single cell gel electrophoresis and quantification of 8-hydroxy-2'-deoxyguanosine. Mol Cell Biochem 2024; 479:2291-2306. [PMID: 37594629 DOI: 10.1007/s11010-023-04826-9] [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: 06/11/2023] [Accepted: 08/06/2023] [Indexed: 08/19/2023]
Abstract
Looking at the development status of Nigeria and other developing nations, most low-income and rural households often use coal as a source of energy which necessitates its trade very close to the communities. Moreover, the effects of exposure to coal mining activities are rarely explored or yet to be studied, not to mention the numerous street coal vendors in Nigeria. This study investigated the oxidative stress levels in serum and urine through the biomarker 8-OHdG and DNA damage via single cell gel electrophoresis (alkaline comet assay). Blood and urine levels of 8-OHdG from 130 coal vendors and 130 population-based controls were determined by ELISA. Alkaline comet assay was also performed on white blood cells for DNA damage. The average values of 8-OHdG in serum and urine of coal vendors were 22.82 and 16.03 ng/ml respectively, which were significantly greater than those detected in controls (p < 0.001; 15.46 and 10.40 ng/ml of 8-OHdG in serum and urine respectively). The average tail length, % DNA in tail and olive tail moment were 25.06 μm, 18.71% and 4.42 respectively for coal vendors. However, for controls, the average values were 4.72 μm, 3.63% and 1.50 for tail length, % DNA in tail and olive tail moment respectively which were much lower than coal vendors (p < 0.001). Therefore, prolonged exposure to coal dusts could lead to higher serum and urinary 8-OHdG and significant DNA damage in coal vendors observed in tail length, % DNA in tail, and olive tail moment by single cell gel electrophoresis. It is therefore established that coal vendors exhibit a huge risk from oxidative stress and assessment of 8-OHdG with single cell gel electrophoresis has proven to be a feasible tool as biomarkers of DNA damage.
Collapse
Affiliation(s)
- Ali Sani
- Department of Instrument Science and Engineering, School of Electronic, Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, People's Republic of China.
- Department of Biological Sciences, Faculty of Life Sciences, Bayero University, Kano, 3011, Nigeria.
| | - Ibrahim Lawal Abdullahi
- Department of Biological Sciences, Faculty of Life Sciences, Bayero University, Kano, 3011, Nigeria
| | - Muhammad Idrees Khan
- Department of Instrument Science and Engineering, School of Electronic, Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, People's Republic of China
| | - ChengXi Cao
- Department of Instrument Science and Engineering, School of Electronic, Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, People's Republic of China
| |
Collapse
|
4
|
Campagna R, Mazzanti L, Pompei V, Alia S, Vignini A, Emanuelli M. The Multifaceted Role of Endothelial Sirt1 in Vascular Aging: An Update. Cells 2024; 13:1469. [PMID: 39273039 PMCID: PMC11394039 DOI: 10.3390/cells13171469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/21/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024] Open
Abstract
NAD+-dependent deacetylase sirtuin-1 (Sirt1) belongs to the sirtuins family, known to be longevity regulators, and exerts a key role in the prevention of vascular aging. By aging, the expression levels of Sirt1 decline with a severe impact on vascular function, such as the rise of endothelial dysfunction, which in turn promotes the development of cardiovascular diseases. In this context, the impact of Sirt1 activity in preventing endothelial senescence is particularly important. Given the key role of Sirt1 in counteracting endothelial senescence, great efforts have been made to deepen the knowledge about the intricate cross-talks and interactions of Sirt1 with other molecules, in order to set up possible strategies to boost Sirt1 activity to prevent or treat vascular aging. The aim of this review is to provide a proper background on the regulation and function of Sirt1 in the vascular endothelium and to discuss the recent advances regarding the therapeutic strategies of targeting Sirt1 to counteract vascular aging.
Collapse
Affiliation(s)
- Roberto Campagna
- Department of Clinical Sciences, Polytechnic University of Marche, 60100 Ancona, Italy; (V.P.); (S.A.); (A.V.); (M.E.)
| | - Laura Mazzanti
- Department of Clinical Sciences, Polytechnic University of Marche, 60100 Ancona, Italy; (V.P.); (S.A.); (A.V.); (M.E.)
- Fondazione Salesi, Ospedale G. Salesi, 60100 Ancona, Italy
| | - Veronica Pompei
- Department of Clinical Sciences, Polytechnic University of Marche, 60100 Ancona, Italy; (V.P.); (S.A.); (A.V.); (M.E.)
| | - Sonila Alia
- Department of Clinical Sciences, Polytechnic University of Marche, 60100 Ancona, Italy; (V.P.); (S.A.); (A.V.); (M.E.)
| | - Arianna Vignini
- Department of Clinical Sciences, Polytechnic University of Marche, 60100 Ancona, Italy; (V.P.); (S.A.); (A.V.); (M.E.)
- Research Center of Health Education and Health Promotion, Università Politecnica delle Marche, 60100 Ancona, Italy
| | - Monica Emanuelli
- Department of Clinical Sciences, Polytechnic University of Marche, 60100 Ancona, Italy; (V.P.); (S.A.); (A.V.); (M.E.)
| |
Collapse
|
5
|
Zhang J, Xia X, He S. Deciphering the causal association and underlying transcriptional mechanisms between telomere length and abdominal aortic aneurysm. Front Immunol 2024; 15:1438838. [PMID: 39234237 PMCID: PMC11371612 DOI: 10.3389/fimmu.2024.1438838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 08/01/2024] [Indexed: 09/06/2024] Open
Abstract
Background The purpose of this study is to investigate the causal effect and potential mechanisms between telomere length and abdominal aortic aneurysm (AAA). Methods Summary statistics of telomere length and AAA were derived from IEU open genome-wide association studies and FinnGen R9, respectively. Bi-directional Mendelian randomization (MR) analysis was conducted to reveal the causal relationship between AAA and telomere length. Three transcriptome datasets were retrieved from the Gene Expression Omnibus database and telomere related genes was down-loaded from TelNet. The overlapping genes of AAA related differentially expressed genes (DEGs), module genes, and telomere related genes were used for further investigation. Telomere related diagnostic biomarkers of AAA were selected with machine learning algorisms and validated in datasets and murine AAA model. The correlation between biomarkers and immune infiltration landscape was established. Results Telomere length was found to have a suggestive negative associations with AAA [IVW, OR 95%CI = 0.558 (0.317-0.701), P < 0.0001], while AAA showed no suggestive effect on telomere length [IVW, OR 95%CI = 0.997 (0.990-1.004), P = 0.4061]. A total of 40 genes was considered as telomere related DEGs of AAA. PLCH2, PRKCQ, and SMG1 were selected as biomarkers after multiple algorithms and validation. Immune infiltration analysis and single cell mRNA analysis revealed that PLCH2 and PRKCQ were mainly expressed on T cells, while SMG1 predominantly expressed on T cells, B cells, and monocytes. Murine AAA model experiments further validated the elevated expression of biomarkers. Conclusion We found a suggestive effect of telomere length on AAA and revealed the potential biomarkers and immune mechanism of telomere length on AAA. This may shed new light for diagnosis and therapeutics on AAA.
Collapse
Affiliation(s)
- Jiyu Zhang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinyi Xia
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shujie He
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
6
|
Yau J, Chukwu P, Jedlicka SS, Ramamurthi A. Assessing trans-endothelial transport of nanoparticles for delivery to abdominal aortic aneurysms. J Biomed Mater Res A 2024; 112:881-894. [PMID: 38192169 DOI: 10.1002/jbm.a.37667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 12/19/2023] [Accepted: 12/27/2023] [Indexed: 01/10/2024]
Abstract
Abdominal aortic aneurysms (AAAs) are localized, rupture-prone expansions of the abdominal aorta wall. In this condition, structural extracellular matrix (ECM) proteins of the aorta wall, elastic fibers and collagen fibers, that impart elasticity and stiffness respectively, are slowly degraded by overexpressed matrix metalloproteinases (MMPs) following an injury stimulus. We are seeking to deliver therapeutics to the AAA wall using polymer nanoparticles (NPs) that are capable of stimulating on-site matrix regeneration and repair. This study aimed to determine how NP shape and size impacts endocytosis and transmigration past the endothelial cell (EC) layer from circulation into the medial layer of the AAA wall. First, rod-shaped NPs were shown to be created based mechanical stretching of PLGA NPs while embedded in a PVA film with longer rod-shaped NPs created based of the degree in which the PVA films are stretched. Live/dead assay reveals that our PLGA NPs are safe and do not cause cell death. Immunofluorescence staining reveal cytokine activation causes endothelial dysfunction in ECs by increasing expression of inflammatory marker Integrin αVβ3 and decreasing expression of adhesion protein vascular endothelial (VE)-cadherin. We showed this disruption enable greater EC uptake and translocation of NPs. Fluorescence studies demonstrate high endothelial transmigration and endocytosis with rod-shaped NPs in cytokine activated ECs compared to healthy control cells, arguing for the benefits of using higher aspect ratio (AR) NPs for accumulation at the aneurysm site. We also demonstrated that the mechanisms of NP transmigration across an activated EC layer depend on NP AR. These results show the potential of using shape as a modality for enhancing permeation of NPs into the aneurysm wall. These studies are also significance to understanding the mechanisms that are likely engaged by NPs for penetrating the endothelial lining of aneurysmal wall segments.
Collapse
Affiliation(s)
- Jimmy Yau
- Department of Bioengineering, Lehigh University, Bethlehem, Pennsylvania, USA
| | - Patience Chukwu
- Department of Bioengineering, Lehigh University, Bethlehem, Pennsylvania, USA
| | - Sabrina S Jedlicka
- Department of Bioengineering, Lehigh University, Bethlehem, Pennsylvania, USA
| | - Anand Ramamurthi
- Department of Bioengineering, Lehigh University, Bethlehem, Pennsylvania, USA
| |
Collapse
|
7
|
Sun J, Wang M, Jia F, Song J, Ren J, Hu B. FTO Stabilizes MIS12 to Inhibit Vascular Smooth Muscle Cell Senescence in Atherosclerotic Plaque. J Inflamm Res 2024; 17:1857-1871. [PMID: 38523689 PMCID: PMC10961024 DOI: 10.2147/jir.s447379] [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: 10/30/2023] [Accepted: 02/28/2024] [Indexed: 03/26/2024] Open
Abstract
Purpose Atherosclerosis is the main cause of atherosclerotic cardiovascular disease (CVD). Here, we aimed to uncover the role and mechanisms of fat mass and obesity-associated genes (FTO) in the regulation of vascular smooth muscle cell (VSMC) senescence in atherosclerotic plaques. Methods ApoE-/- mice fed a high-fat diet (HFD) were used to establish an atherosclerotic animal model. Immunohistochemistry, and the staining of hematoxylin-eosin, Oil Red O, Sirius red, and Masson were performed to confirm the role of FTO in atherosclerosis in vivo. Subsequently, FTO expression in primary VSMCs is either upregulated or downregulated. Oxidized low-density lipoprotein (ox-LDL) was used to treat VSMCs, followed by EdU staining, flow cytometry, senescence-associated β-galactosidase (SA-β-gal) staining, immunofluorescence, telomere detection, RT-qPCR, and Western blotting to determine the molecular mechanisms by which FTO inhibits VSMC senescence. Results Decreased FTO expression was observed in progressive atherosclerotic plaques of ApoE-/- mice fed with HFD. FTO upregulation inhibits atherosclerotic lesions in mice. FTO inhibits VSMC aging in atherosclerotic plaques by helping VSMC withstand ox-LDL-induced cell cycle arrest and senescence. This process is achieved by stabilizing the MIS12 protein in VSMC through a proteasome-mediated pathway. Conclusion FTO inhibits VSMC senescence and subsequently slows the progression of atherosclerotic plaques by stabilizing the MIS12 protein.
Collapse
Affiliation(s)
- Jingzhao Sun
- Department of Emergency, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, People’s Republic of China
| | - Mengqi Wang
- Department of Emergency, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, People’s Republic of China
| | - Fengming Jia
- Department of Emergency, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, People’s Republic of China
| | - Jiantao Song
- Department of Emergency, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, People’s Republic of China
| | - Jinlin Ren
- Department of Emergency, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, People’s Republic of China
| | - Bo Hu
- Department of Emergency, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, People’s Republic of China
| |
Collapse
|
8
|
Kalies K, Knöpp K, Wurmbrand L, Korte L, Dutzmann J, Pilowski C, Koch S, Sedding D. Isolation of circulating endothelial cells provides tool to determine endothelial cell senescence in blood samples. Sci Rep 2024; 14:4271. [PMID: 38383692 PMCID: PMC10882010 DOI: 10.1038/s41598-024-54455-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 02/13/2024] [Indexed: 02/23/2024] Open
Abstract
Circulating endothelial cells (CEC) are arising as biomarkers for vascular diseases. However, whether they can be utilized as markers of endothelial cell (EC) senescence in vivo remains unknown. Here, we present a protocol to isolate circulating endothelial cells for a characterization of their senescent signature. Further, we characterize different models of EC senescence induction in vitro and show similar patterns of senescence being upregulated in CECs of aged patients as compared to young volunteers. Replication-(ageing), etoposide-(DNA damage) and angiotensin II-(ROS) induced senescence models showed the expected cell morphology and proliferation-reduction effects. Expression of senescence-associated secretory phenotype markers was specifically upregulated in replication-induced EC senescence. All models showed reduced telomere lengths and induction of the INK4a/ARF locus. Additional p14ARF-p21 pathway activation was observed in replication- and etoposide-induced EC senescence. Next, we established a combined magnetic activated- and fluorescence activated cell sorting (MACS-FACS) based protocol for CEC isolation. Interestingly, CECs isolated from aged volunteers showed similar senescence marker patterns as replication- and etoposide-induced senescence models. Here, we provide first proof of senescence in human blood derived circulating endothelial cells. These results hint towards an exciting future of using CECs as mirror cells for in vivo endothelial cell senescence, of particular interest in the context of endothelial dysfunction and cardiovascular diseases.
Collapse
Affiliation(s)
- Katrin Kalies
- Mid-German Heart Center, Department of Internal Medicine III, Division of Cardiology, Angiology and Intensive Medical Care, University Hospital Halle, Martin-Luther-University Halle-Wittenberg, Ernst-Grube-Strasse 40, 06120, Halle (Saale), Germany.
| | - Kai Knöpp
- Mid-German Heart Center, Department of Internal Medicine III, Division of Cardiology, Angiology and Intensive Medical Care, University Hospital Halle, Martin-Luther-University Halle-Wittenberg, Ernst-Grube-Strasse 40, 06120, Halle (Saale), Germany
| | - Leonie Wurmbrand
- Mid-German Heart Center, Department of Internal Medicine III, Division of Cardiology, Angiology and Intensive Medical Care, University Hospital Halle, Martin-Luther-University Halle-Wittenberg, Ernst-Grube-Strasse 40, 06120, Halle (Saale), Germany
| | - Laura Korte
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg Straße 1, 30625, Hannover, Germany
| | - Jochen Dutzmann
- Mid-German Heart Center, Department of Internal Medicine III, Division of Cardiology, Angiology and Intensive Medical Care, University Hospital Halle, Martin-Luther-University Halle-Wittenberg, Ernst-Grube-Strasse 40, 06120, Halle (Saale), Germany
| | - Claudia Pilowski
- Mid-German Heart Center, Department of Internal Medicine III, Division of Cardiology, Angiology and Intensive Medical Care, University Hospital Halle, Martin-Luther-University Halle-Wittenberg, Ernst-Grube-Strasse 40, 06120, Halle (Saale), Germany
| | - Susanne Koch
- Mid-German Heart Center, Department of Internal Medicine III, Division of Cardiology, Angiology and Intensive Medical Care, University Hospital Halle, Martin-Luther-University Halle-Wittenberg, Ernst-Grube-Strasse 40, 06120, Halle (Saale), Germany
| | - Daniel Sedding
- Mid-German Heart Center, Department of Internal Medicine III, Division of Cardiology, Angiology and Intensive Medical Care, University Hospital Halle, Martin-Luther-University Halle-Wittenberg, Ernst-Grube-Strasse 40, 06120, Halle (Saale), Germany
| |
Collapse
|
9
|
Wang D, Jia L, Zhao C, Wang H, Dai Z, Jing Y, Jiang B, Xin S. Mitochondrial quality control in abdominal aortic aneurysm: From molecular mechanisms to therapeutic strategies. FASEB J 2023; 37:e22969. [PMID: 37184038 DOI: 10.1096/fj.202202158rr] [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: 12/28/2022] [Revised: 03/20/2023] [Accepted: 05/01/2023] [Indexed: 05/16/2023]
Abstract
Mitochondria are the energy supply sites of cells and are crucial for eukaryotic life. Mitochondrial dysfunction is involved in the pathogenesis of abdominal aortic aneurysm (AAA). Multiple mitochondrial quality control (MQC) mechanisms, including mitochondrial DNA repair, biogenesis, antioxidant defense, dynamics, and autophagy, play vital roles in maintaining mitochondrial homeostasis under physiological and pathological conditions. Abnormalities in these mechanisms may induce mitochondrial damage and dysfunction leading to cell death and tissue remodeling. Recently, many clues suggest that dysregulation of MQC is closely related to the pathogenesis of AAA. Therefore, specific interventions targeting MQC mechanisms to maintain and restore mitochondrial function have become promising therapeutic methods for the prevention and treatment of AAA.
Collapse
Affiliation(s)
- Ding Wang
- Department of Vascular Surgery, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
- Key Laboratory of Pathogenesis, Prevention and Therapeutics of aortic aneurysm, Shenyang, Liaoning Province, China
| | - Longyuan Jia
- Department of Vascular Surgery, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
- Key Laboratory of Pathogenesis, Prevention and Therapeutics of aortic aneurysm, Shenyang, Liaoning Province, China
| | - Chengdong Zhao
- Department of Vascular Surgery, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
- Key Laboratory of Pathogenesis, Prevention and Therapeutics of aortic aneurysm, Shenyang, Liaoning Province, China
| | - Huitao Wang
- Department of Vascular Surgery, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
- Key Laboratory of Pathogenesis, Prevention and Therapeutics of aortic aneurysm, Shenyang, Liaoning Province, China
| | - Zhengnan Dai
- Department of Vascular Surgery, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
- Key Laboratory of Pathogenesis, Prevention and Therapeutics of aortic aneurysm, Shenyang, Liaoning Province, China
| | - Yuchen Jing
- Department of Vascular Surgery, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
- Key Laboratory of Pathogenesis, Prevention and Therapeutics of aortic aneurysm, Shenyang, Liaoning Province, China
| | - Bo Jiang
- Department of Vascular Surgery, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
- Key Laboratory of Pathogenesis, Prevention and Therapeutics of aortic aneurysm, Shenyang, Liaoning Province, China
| | - Shijie Xin
- Department of Vascular Surgery, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
- Key Laboratory of Pathogenesis, Prevention and Therapeutics of aortic aneurysm, Shenyang, Liaoning Province, China
| |
Collapse
|
10
|
Mitochondrial Dysfunction and Increased DNA Damage in Vascular Smooth Muscle Cells of Abdominal Aortic Aneurysm (AAA-SMC). OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:6237960. [PMID: 36743698 PMCID: PMC9891816 DOI: 10.1155/2023/6237960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/30/2022] [Accepted: 11/24/2022] [Indexed: 01/27/2023]
Abstract
There is increasing evidence for enhanced oxidative stress in the vascular wall of abdominal aortic aneurysms (AAA). Mitochondrial damage and dysfunction are hypothesized to be actors in altered production of reactive oxygen species (ROS) and oxidative stress. However, the role of mitochondria and oxidative stress in vascular remodelling and progression of AAA remains uncertain. We here addressed whether mitochondrial dysfunction is persistently increased in vascular smooth muscle cells (VSMCs) isolated from AAA compared to healthy VSMC. AAA-derived VSMC cultures (AAA-SMC, n = 10) and normal VSMC cultures derived from healthy donors (n = 7) were grown in vitro and analysed for four parameters, indicating mitochondrial dysfunction: (i) mitochondrial content and morphology, (ii) ROS production and antioxidative response, (iii) NADP+/NADPH content and ratio, and (iv) DNA damage, in the presence or absence of angiotensin II (AngII). AAA-SMC displayed increased mitochondrial circularity (rounded shape), reduced mitochondrial area, and reduced perimeter, indicating increased fragmentation and dysfunction compared to healthy controls. This was accompanied by significantly increased O2 - production, reduced NADP+/NADPH levels, a lower antioxidative response (indicated by antioxidative response element- (ARE-) driven luciferase reporter assays), more DNA damage (determined by percentage of γ-H2A.X-positive nuclei), and earlier growth arrest in AAA-SMC. Our data suggest that mitochondrial dysfunction and oxidative stress are persistently increased in AAA-SMC, emphasizing their implication in the pathophysiology of AAA.
Collapse
|
11
|
Senescence-Associated Secretory Phenotype of Cardiovascular System Cells and Inflammaging: Perspectives of Peptide Regulation. Cells 2022; 12:cells12010106. [PMID: 36611900 PMCID: PMC9818427 DOI: 10.3390/cells12010106] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 12/23/2022] [Accepted: 12/24/2022] [Indexed: 12/28/2022] Open
Abstract
A senescence-associated secretory phenotype (SASP) and a mild inflammatory response characteristic of senescent cells (inflammaging) form the conditions for the development of cardiovascular diseases: atherosclerosis, coronary heart disease, and myocardial infarction. The purpose of the review is to analyze the pool of signaling molecules that form SASP and inflammaging in cells of the cardiovascular system and to search for targets for the action of vasoprotective peptides. The SASP of cells of the cardiovascular system is characterized by a change in the synthesis of anti-proliferative proteins (p16, p19, p21, p38, p53), cytokines characteristic of inflammaging (IL-1α,β, IL-4, IL-6, IL-8, IL-18, TNFα, TGFβ1, NF-κB, MCP), matrix metalloproteinases, adhesion molecules, and sirtuins. It has been established that peptides are physiological regulators of body functions. Vasoprotective polypeptides (liraglutide, atrial natriuretic peptide, mimetics of relaxin, Ucn1, and adropin), KED tripeptide, and AEDR tetrapeptide regulate the synthesis of molecules involved in inflammaging and SASP-forming cells of the cardiovascular system. This indicates the prospects for the development of drugs based on peptides for the treatment of age-associated cardiovascular pathology.
Collapse
|
12
|
Aschacher T, Geisler D, Lenz V, Aschacher O, Winkler B, Schaefer AK, Mitterbauer A, Wolf B, Enzmann FK, Messner B, Laufer G, Ehrlich MP, Grabenwöger M, Bergmann M. Impacts of Telomeric Length, Chronic Hypoxia, Senescence, and Senescence-Associated Secretory Phenotype on the Development of Thoracic Aortic Aneurysm. Int J Mol Sci 2022; 23:ijms232415498. [PMID: 36555139 PMCID: PMC9779024 DOI: 10.3390/ijms232415498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 11/18/2022] [Accepted: 11/30/2022] [Indexed: 12/13/2022] Open
Abstract
Thoracic aortic aneurysm (TAA) is an age-related and life-threatening vascular disease. Telomere shortening is a predictor of age-related diseases, and its progression is associated with premature vascular disease. The aim of the present work was to investigate the impacts of chronic hypoxia and telomeric DNA damage on cellular homeostasis and vascular degeneration of TAA. We analyzed healthy and aortic aneurysm specimens (215 samples) for telomere length (TL), chronic DNA damage, and resulting changes in cellular homeostasis, focusing on senescence and apoptosis. Compared with healthy thoracic aorta (HTA), patients with tricuspid aortic valve (TAV) showed telomere shortening with increasing TAA size, in contrast to genetically predisposed bicuspid aortic valve (BAV). In addition, TL was associated with chronic hypoxia and telomeric DNA damage and with the induction of senescence-associated secretory phenotype (SASP). TAA-TAV specimens showed a significant difference in SASP-marker expression of IL-6, NF-κB, mTOR, and cell-cycle regulators (γH2AX, Rb, p53, p21), compared to HTA and TAA-BAV. Furthermore, we observed an increase in CD163+ macrophages and a correlation between hypoxic DNA damage and the number of aortic telocytes. We conclude that chronic hypoxia is associated with telomeric DNA damage and the induction of SASP in a diseased aortic wall, promising a new therapeutic target.
Collapse
Affiliation(s)
- Thomas Aschacher
- Department of Cardiovascular Surgery, Clinic Floridsdorf and Karl Landsteiner Institute for Cardio-Vascular Research, 1210 Vienna, Austria
- Department of Cardiac Surgery, Medical University of Vienna, 1090 Vienna, Austria
- Correspondence: ; Tel.: +43-1-277-00-74316
| | - Daniela Geisler
- Department of Cardiovascular Surgery, Clinic Floridsdorf and Karl Landsteiner Institute for Cardio-Vascular Research, 1210 Vienna, Austria
| | - Verena Lenz
- Department of Cardiovascular Surgery, Clinic Floridsdorf and Karl Landsteiner Institute for Cardio-Vascular Research, 1210 Vienna, Austria
| | - Olivia Aschacher
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Bernhard Winkler
- Department of Cardiovascular Surgery, Clinic Floridsdorf and Karl Landsteiner Institute for Cardio-Vascular Research, 1210 Vienna, Austria
- Faculty of Medicine, Sigmund Freud Private University of Vienna, 1030 Vienna, Austria
| | | | - Andreas Mitterbauer
- Department of General Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Brigitte Wolf
- Department of General Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Florian K. Enzmann
- Department of Vascular Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Barbara Messner
- Department of Cardiac Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Günther Laufer
- Department of Cardiac Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Marek P. Ehrlich
- Department of Cardiac Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Martin Grabenwöger
- Department of Cardiovascular Surgery, Clinic Floridsdorf and Karl Landsteiner Institute for Cardio-Vascular Research, 1210 Vienna, Austria
- Faculty of Medicine, Sigmund Freud Private University of Vienna, 1030 Vienna, Austria
| | - Michael Bergmann
- Department of General Surgery, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
13
|
Sanchez M, Kannengiesser C, Hoang S, Potier L, Fumeron F, Venteclef N, Scheen A, Gautier JF, Hadjadj S, Marre M, Roussel R, Mohammedi K, Velho G. Leukocyte telomere length, allelic variations in related genes and risk of coronary heart disease in people with long-standing type 1 diabetes. Cardiovasc Diabetol 2022; 21:206. [PMID: 36221106 PMCID: PMC9554968 DOI: 10.1186/s12933-022-01635-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 09/01/2022] [Indexed: 11/20/2022] Open
Abstract
Background Type 1 diabetes is associated with accelerated vascular aging and advanced atherosclerosis resulting in increased rates of cardiovascular disease and premature death. We evaluated associations between Leukocyte telomere length (LTL), allelic variations (SNPs) in LTL-related genes and the incidence of coronary heart disease (CHD) in adults with long-standing type 1 diabetes. Methods We assessed associations of LTL, measured at baseline by RT–PCR, and of SNPs in 11 LTL-related genes with the risk of coronary heart disease (CHD: myocardial infarction or coronary revascularization) and all-cause death during follow-up in two multicenter French-Belgian prospective cohorts of people with long-standing type 1 diabetes. Results In logistic and Cox analyses, the lowest tertile of LTL distribution (short telomeres) at baseline was associated with the prevalence of myocardial infarction at baseline and with increased risk of CHD (Hazard ratio 3.14 (1.39–7.70), p = 0.005, for shorter vs longer tertile of LTL) and all-cause death (Hazard ratio 1.63 (95% CI 1.04–2.55), p = 0.03, for shorter vs combined intermediate and longer tertiles of LTL) during follow-up. Allelic variations in six genes related to telomere biology (TERC, NAF1, TERT, TNKS, MEN1 and BICD1) were also associated with the incidence of CHD during follow-up. The associations were independent of sex, age, duration of diabetes, and a range of relevant confounding factors at baseline. Conclusions Our results suggest that short LTL is an independent risk factor for CHD in people with type 1 diabetes. Supplementary Information The online version contains supplementary material available at 10.1186/s12933-022-01635-0.
Collapse
Affiliation(s)
- Manuel Sanchez
- INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker-Enfants Malades, Université Paris Cité, Paris, France. .,UFR de Médecine, Université Paris Cité, Paris, France. .,Department of Geriatrics, Assistance Publique - Hôpitaux de Paris, Bichat University Hospital, 46 rue Henri Huchard, 75018, Paris, France.
| | - Caroline Kannengiesser
- UFR de Médecine, Université Paris Cité, Paris, France.,Department of Genetics, Assistance Publique - Hôpitaux de Paris, DHU FIRE, Bichat Hospital, Paris, France
| | - Sophie Hoang
- Department of Geriatrics, Charles-Foix University Hospital, Vitry sur Seine, France
| | - Louis Potier
- INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker-Enfants Malades, Université Paris Cité, Paris, France.,UFR de Médecine, Université Paris Cité, Paris, France.,Department of Diabetology, Endocrinology and Nutrition, Assistance Publique - Hôpitaux de Paris, DHU FIRE, Bichat Hospital, Paris, France
| | - Frédéric Fumeron
- INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker-Enfants Malades, Université Paris Cité, Paris, France
| | - Nicolas Venteclef
- INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker-Enfants Malades, Université Paris Cité, Paris, France
| | - André Scheen
- Department of Diabetology, Endocrinology and Nutrition, Sart Tilman University Hospital, Liège, Belgium
| | - Jean-François Gautier
- INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker-Enfants Malades, Université Paris Cité, Paris, France.,UFR de Médecine, Université Paris Cité, Paris, France.,Department of Diabetology, Endocrinology and Nutrition, Assistance Publique - Hôpitaux de Paris, Lariboisière University Hospital, Paris, France
| | - Samy Hadjadj
- Institut du Thorax, INSERM, CNRS, CHU Nantes, Université de Nantes, Nantes, France
| | - Michel Marre
- Clinique Ambroise Paré, Neuilly-sur-Seine, France
| | - Ronan Roussel
- INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker-Enfants Malades, Université Paris Cité, Paris, France.,UFR de Médecine, Université Paris Cité, Paris, France.,Department of Diabetology, Endocrinology and Nutrition, Assistance Publique - Hôpitaux de Paris, DHU FIRE, Bichat Hospital, Paris, France
| | - Kamel Mohammedi
- INSERM U1034, Bordeaux University and Hospital, Bordeaux, France
| | - Gilberto Velho
- INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker-Enfants Malades, Université Paris Cité, Paris, France
| |
Collapse
|
14
|
Phillips AR, Andraska EA, Reitz KM, Habib S, Martinez-Meehan D, Dai Y, Johnson AE, Liang NL. Association between neighborhood deprivation and presenting with a ruptured abdominal aortic aneurysm before screening age. J Vasc Surg 2022; 76:932-941.e2. [PMID: 35314299 PMCID: PMC9482667 DOI: 10.1016/j.jvs.2022.03.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 03/04/2022] [Indexed: 10/18/2022]
Abstract
OBJECTIVE Recent data indicate social determinants of health (SDOH) have a great impact on prevention and treatment outcomes across a broad variety of disease states, especially cardiovascular diseases. The area deprivation index (ADI) is a validated measure of neighborhood level disadvantage capturing key social determinate factors. Abdominal aortic aneurysm rupture (rAAA) is highly morbid, but also preventable through evidence-based screening. However, the association between rAAA and SDOH is poorly characterized. Our objective is to study the association of SDOH with rAAA and screening age. METHODS This retrospective study included patients who underwent operative repair of a rAAA at a multihospital healthcare system (2003-2019). Deprivation was measured by the ADI (scale 1-100), grouped into quintiles for simplicity, with higher quintiles indicating greater deprivation. Patients with the highest quintile ADI (89-100) were categorized as the most deprived. We investigated the association between neighborhood deprivation with the odds of (i) undergoing repair for rAAA before screening age 65 and (ii) undergoing endovascular aortic repair (EVAR) using logistic regression, sequentially modeling nonmodifiable then both nonmodifiable and modifiable confounding variables. RESULTS There were 632 patients who met the inclusion criteria (aged 74.2 ± 9.4 years; 174 women [27.6%]; 564 White [89.2%]; ADI 66.8 ± 22.3). Those from the most deprived neighborhoods (n = 118) were younger (71.7 ± 10.0 years vs 74.8 ± 9.2 years; P = .002), more likely to be female (36% vs 26%; P = .031), more likely to be Black (5.9% vs 0.4%; P = .007), and fewer underwent EVAR (28% vs 39.5%; P = .020) compared with those from other neighborhoods. On sequential modeling, residing in the most deprived neighborhoods was associated with undergoing rAAA repair before age 65 after adjusting for nonmodifiable factors (odds ratio [OR], 2.02; 95% confidence interval [CI], 1.39-2.95; P < .001), and nonmodifiable as well as modifiable factors (OR, 2.22; 95% CI, 1.56-3.16; P < .001). Those in the most deprived neighborhoods had a lower odds of undergoing EVAR compared with open repair after adjusting for nonmodifiable factors (OR, 0.64; 95% CI, 0.41-0.98; P = .042), and nonmodifiable as well as modifiable factors (OR, 0.61; 95% CI, 0.37-0.99; P = .047). CONCLUSIONS Among patients who underwent rAAA, residing in the most deprived neighborhoods was associated with greater adjusted odds of presenting under age 65 and undergoing an open repair. These neighborhoods represent tangible geographic targets that may benefit from a younger screening age, enhanced education, and access to care. These findings stress the importance of developing strategies for early prevention and diagnosis of cardiovascular diseases among patients with disadvantageous SDOH.
Collapse
Affiliation(s)
- Amanda R Phillips
- Department of Surgery, Division of Vascular Surgery, UPMC, Pittsburgh, PA.
| | | | - Katherine M Reitz
- Department of Surgery, Division of Vascular Surgery, UPMC, Pittsburgh, PA
| | - Salim Habib
- Department of Surgery, Division of Vascular Surgery, UPMC, Pittsburgh, PA
| | | | - Yancheng Dai
- University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Amber E Johnson
- Department of Medicine, Division of Cardiology, UPMC, Pittsburgh, PA
| | - Nathan L Liang
- Department of Surgery, Division of Vascular Surgery, UPMC, Pittsburgh, PA; University of Pittsburgh School of Medicine, Pittsburgh, PA
| |
Collapse
|
15
|
Ya J, Kadir RRA, Bayraktutan U. Delay of endothelial cell senescence protects cerebral barrier against age-related dysfunction: role of senolytics and senomorphics. Tissue Barriers 2022:2103353. [PMID: 35880392 PMCID: PMC10364655 DOI: 10.1080/21688370.2022.2103353] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022] Open
Abstract
Accumulation of senescent cells in cerebrovasculature is thought to play an important role in age-related disruption of blood-brain barrier (BBB). Using an in vitro model of human BBB, composed of brain microvascular endothelial cells (BMECs), astrocytes and pericytes, this study explored the so-called correlative link between BMEC senescence and the BBB dysfunction in the absence or presence of functionally distinct senotherapeutics. Replicative senescence was deemed present at passage ≥19 where BMECs displayed shortened telomere length, reduced proliferative and tubulogenic potentials and increased NADPH oxidase activity, superoxide anion production (markers of oxidative stress), S-β-galactosidase activity and γ-H2AX staining. Significant impairments observed in integrity and function of a model of BBB established with senescent BMECs, ascertained successively by decreases in transendothelial electrical resistance and increases in paracellular flux, revealed a close correlation between endothelial cell senescence and BBB dysfunction. Disruptions in the localization or expression of tight junction proteins, zonula occludens-1, occludin, and claudin-5 in senescent BMECs somewhat explained this dysfunction. Indeed, treatment of relatively old BMEC (passage 16) with a cocktail of senolytics (dasatinib and quercetin) or senomorphics targeting transcription factor NF-κB (QNZ), p38MAPK signaling pathway (BIRB-796) or pro-oxidant enzyme NADPH oxidase (VAS2870) until passage 20 rendered these cells more resistant to senescence and totally preserved BBB characteristics by restoring subcellular localization and expression of tight junction proteins. In conclusion, attempts that effectively mitigate accumulation of senescent endothelial cells in cerebrovasculature may prevent age-related BBB dysfunction and may be of prophylactic or therapeutic value to extend lifelong health and wellbeing.
Collapse
Affiliation(s)
- Jingyuan Ya
- Academic Unit of Mental Health and Clinical Neuroscience, School of Medicine, University of Nottingham, Nottingham, UK
| | - Rais Reskiawan A Kadir
- Academic Unit of Mental Health and Clinical Neuroscience, School of Medicine, University of Nottingham, Nottingham, UK
| | - Ulvi Bayraktutan
- Academic Unit of Mental Health and Clinical Neuroscience, School of Medicine, University of Nottingham, Nottingham, UK
| |
Collapse
|
16
|
Jauhiainen S, Kiema M, Hedman M, Laakkonen JP. Large Vessel Cell Heterogeneity and Plasticity: Focus in Aortic Aneurysms. Arterioscler Thromb Vasc Biol 2022; 42:811-818. [PMID: 35587695 DOI: 10.1161/atvbaha.121.316237] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Smooth muscle cells and endothelial cells have a remarkable level of plasticity in vascular pathologies. In thoracic and abdominal aortic aneurysms, smooth muscle cells have been suggested to undergo phenotypic switching and to contribute to degradation of the aortic wall structure in response to, for example, inflammatory mediators, dysregulation of growth factor signaling or oxidative stress. Recently, endothelial-to-mesenchymal transition, and a clonal expansion of degradative smooth muscle cells and immune cells, as well as mesenchymal stem-like cells have been suggested to contribute to the progression of aortic aneurysms. What are the factors driving the aortic cell phenotype changes and how vascular flow, known to affect aortic wall structure and to be altered in aortic aneurysms, could affect aortic cell remodeling? In this review, we summarize the current literature on aortic cell heterogeneity and phenotypic switching in relation to changes in vascular flow and aortic wall structure in aortic aneurysms in clinical samples with special focus on smooth muscle and endothelial cells. The differences between thoracic and abdominal aortic aneurysms are discussed.
Collapse
Affiliation(s)
- Suvi Jauhiainen
- A.I. Virtanen Institute for Molecular Sciences (S.J., M.K., J.P.L.), University of Eastern Finland, Kuopio
| | - Miika Kiema
- A.I. Virtanen Institute for Molecular Sciences (S.J., M.K., J.P.L.), University of Eastern Finland, Kuopio
| | - Marja Hedman
- Institute of Clinical Medicine (M.H.), University of Eastern Finland, Kuopio
- Department of Clinical Radiology, Kuopio University Hospital, Finland (M.H.)
- Department of Heart and Thoracic Surgery, Kuopio University Hospital, Heart Center, Kuopio, Finland (M.H.)
| | - Johanna P Laakkonen
- A.I. Virtanen Institute for Molecular Sciences (S.J., M.K., J.P.L.), University of Eastern Finland, Kuopio
| |
Collapse
|
17
|
Inci N, Kamali D, Akyildiz EO, Tahir Turanli E, Bozaykut P. Translation of Cellular Senescence to Novel Therapeutics: Insights From Alternative Tools and Models. FRONTIERS IN AGING 2022; 3:828058. [PMID: 35821852 PMCID: PMC9261353 DOI: 10.3389/fragi.2022.828058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 04/12/2022] [Indexed: 01/10/2023]
Abstract
Increasing chronological age is the greatest risk factor for human diseases. Cellular senescence (CS), which is characterized by permanent cell-cycle arrest, has recently emerged as a fundamental mechanism in developing aging-related pathologies. During the aging process, senescent cell accumulation results in senescence-associated secretory phenotype (SASP) which plays an essential role in tissue dysfunction. Although discovered very recently, senotherapeutic drugs have been already involved in clinical studies. This review gives a summary of the molecular mechanisms of CS and its role particularly in the development of cardiovascular diseases (CVD) as the leading cause of death. In addition, it addresses alternative research tools including the nonhuman and human models as well as computational techniques for the discovery of novel therapies. Finally, senotherapeutic approaches that are mainly classified as senolytics and senomorphics are discussed.
Collapse
Affiliation(s)
- Nurcan Inci
- Graduate School of Natural and Applied Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Dilanur Kamali
- Graduate School of Natural and Applied Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Erdogan Oguzhan Akyildiz
- Graduate School of Natural and Applied Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Eda Tahir Turanli
- Graduate School of Natural and Applied Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
- Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Perinur Bozaykut
- Graduate School of Natural and Applied Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
- Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| |
Collapse
|
18
|
Phillippe M. Telomeres, oxidative stress, and timing for spontaneous term and preterm labor. Am J Obstet Gynecol 2022; 227:148-162. [PMID: 35460626 DOI: 10.1016/j.ajog.2022.04.024] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 04/03/2022] [Accepted: 04/12/2022] [Indexed: 11/26/2022]
Abstract
Telomeres are nucleoprotein complexes located at the distal ends of chromosomes. In adults, progressive telomere shortening occurs throughout the lifetime and is thought to contribute to progressive aging, physiological senescence, multiorgan dysfunction, and ultimately, death. As discussed in this review, multiple lines of evidence provide support for the biological plausibility that a telomere-based clock mechanism also determines the length of gestation, leading to the onset of labor (parturition). After telomere expansion at the beginning of pregnancy, the telomere lengths in the gestational tissues (ie, the placenta and fetal membranes) progressively shorten throughout the remainder of pregnancy. The rate of telomere shortening can be accelerated by conditions that affect the mother and result in oxidative stress. Preterm births in the United States are associated with multiple risk factors that are linked with increased oxidative stress. Antioxidant vitamins (ie, vitamins E and C) mitigate the effects of oxidative stress and delay or prevent telomere shortening. Clinical trials with vitamins E and C and with multivitamins started during the periconception period have been associated with reduced rates of preterm births. In the United States, African-American women have a 2-3-fold higher rate of preterm birth. African-American women have multiple risk factors for premature birth, all of which are distinct and potentially additive with regard to epigenetic telomere shortening. The "weathering effect" is the hypothesis to explain the increased rates of chronic illness, disabilities, and early death observed in African-Americans. With regard to pregnancy, accelerated weathering with the associated telomere shortening in the gestational tissues would not only explain the preterm birth disparity but could also explain why highly educated, affluent African-American women continue to have an increased rate of preterm birth. These studies suggest that the racial disparities in preterm birth are potentially mediated by telomere shortening produced by lifetime or even generational exposure to the effects of systemic racism and socioeconomic marginalization. In conclusion, this review presents multiple lines of evidence supporting a novel hypothesis regarding the biological clock mechanism that determines the length of pregnancy, and it opens the possibility of new approaches to prevent or reduce the rate of spontaneous preterm birth.
Collapse
|
19
|
Dookun E, Passos JF, Arthur HM, Richardson GD. Therapeutic Potential of Senolytics in Cardiovascular Disease. Cardiovasc Drugs Ther 2022; 36:187-196. [PMID: 32979174 PMCID: PMC8770386 DOI: 10.1007/s10557-020-07075-w] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/03/2020] [Indexed: 02/07/2023]
Abstract
Ageing is the biggest risk factor for impaired cardiovascular health, with cardiovascular disease being the leading cause of death in 40% of individuals over 65 years old. Ageing is associated with both an increased prevalence of cardiovascular disease including heart failure, coronary artery disease, and myocardial infarction. Furthermore, ageing is associated with a poorer prognosis to these diseases. Genetic models allowing the elimination of senescent cells revealed that an accumulation of senescence contributes to the pathophysiology of cardiovascular ageing and promotes the progression of cardiovascular disease through the expression of a proinflammatory and profibrotic senescence-associated secretory phenotype. These studies have resulted in an effort to identify pharmacological therapeutics that enable the specific elimination of senescent cells through apoptosis induction. These senescent cell apoptosis-inducing compounds are termed senolytics and their potential to ameliorate age-associated cardiovascular disease is the focus of this review.
Collapse
Affiliation(s)
- Emily Dookun
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE1 3BZ, UK
| | - João F Passos
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Helen M Arthur
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE1 3BZ, UK
| | - Gavin D Richardson
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE1 3BZ, UK.
| |
Collapse
|
20
|
Kessler V, Klopf J, Eilenberg W, Neumayer C, Brostjan C. AAA Revisited: A Comprehensive Review of Risk Factors, Management, and Hallmarks of Pathogenesis. Biomedicines 2022; 10:94. [PMID: 35052774 PMCID: PMC8773452 DOI: 10.3390/biomedicines10010094] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 12/30/2021] [Indexed: 01/27/2023] Open
Abstract
Despite declining incidence and mortality rates in many countries, the abdominal aortic aneurysm (AAA) continues to represent a life-threatening cardiovascular condition with an overall prevalence of about 2-3% in the industrialized world. While the risk of AAA development is considerably higher for men of advanced age with a history of smoking, screening programs serve to detect the often asymptomatic condition and prevent aortic rupture with an associated death rate of up to 80%. This review summarizes the current knowledge on identified risk factors, the multifactorial process of pathogenesis, as well as the latest advances in medical treatment and surgical repair to provide a perspective for AAA management.
Collapse
Affiliation(s)
| | | | | | | | - Christine Brostjan
- Department of General Surgery, Division of Vascular Surgery, Medical University of Vienna, Vienna General Hospital, 1090 Vienna, Austria; (V.K.); (J.K.); (W.E.); (C.N.)
| |
Collapse
|
21
|
Sanhueza-Olivares F, Troncoso MF, Pino-de la Fuente F, Martinez-Bilbao J, Riquelme JA, Norambuena-Soto I, Villa M, Lavandero S, Castro PF, Chiong M. A potential role of autophagy-mediated vascular senescence in the pathophysiology of HFpEF. Front Endocrinol (Lausanne) 2022; 13:1057349. [PMID: 36465616 PMCID: PMC9713703 DOI: 10.3389/fendo.2022.1057349] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 10/26/2022] [Indexed: 11/18/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is one of the most complex and most prevalent cardiometabolic diseases in aging population. Age, obesity, diabetes, and hypertension are the main comorbidities of HFpEF. Microvascular dysfunction and vascular remodeling play a major role in its development. Among the many mechanisms involved in this process, vascular stiffening has been described as one the most prevalent during HFpEF, leading to ventricular-vascular uncoupling and mismatches in aged HFpEF patients. Aged blood vessels display an increased number of senescent endothelial cells (ECs) and vascular smooth muscle cells (VSMCs). This is consistent with the fact that EC and cardiomyocyte cell senescence has been reported during HFpEF. Autophagy plays a major role in VSMCs physiology, regulating phenotypic switch between contractile and synthetic phenotypes. It has also been described that autophagy can regulate arterial stiffening and EC and VSMC senescence. Many studies now support the notion that targeting autophagy would help with the treatment of many cardiovascular and metabolic diseases. In this review, we discuss the mechanisms involved in autophagy-mediated vascular senescence and whether this could be a driver in the development and progression of HFpEF.
Collapse
Affiliation(s)
- Fernanda Sanhueza-Olivares
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Mayarling F. Troncoso
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Francisco Pino-de la Fuente
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Javiera Martinez-Bilbao
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Jaime A. Riquelme
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Ignacio Norambuena-Soto
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Monica Villa
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Sergio Lavandero
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Pablo F. Castro
- Advanced Center for Chronic Diseases, Faculty of Medicine, Pontifical University Catholic of Chile, Santiago, Chile
| | - Mario Chiong
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
- *Correspondence: Mario Chiong,
| |
Collapse
|
22
|
Begum MK, Konja D, Singh S, Chlopicki S, Wang Y. Endothelial SIRT1 as a Target for the Prevention of Arterial Aging: Promises and Challenges. J Cardiovasc Pharmacol 2021; 78:S63-S77. [PMID: 34840264 DOI: 10.1097/fjc.0000000000001154] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 09/25/2021] [Indexed: 12/15/2022]
Abstract
ABSTRACT SIRT1, a member of the sirtuin family of longevity regulators, possesses potent activities preventing vascular aging. The expression and function of SIRT1 in endothelial cells are downregulated with age, in turn causing early vascular aging and predisposing various vascular abnormalities. Overexpression of SIRT1 in the vascular endothelium prevents aging-associated endothelial dysfunction and senescence, thus the development of hypertension and atherosclerosis. Numerous efforts have been directed to increase SIRT1 signaling as a potential strategy for different aging-associated diseases. However, the complex mechanisms underlying the regulation of SIRT1 have posed a significant challenge toward the design of specific and effective therapeutics. This review aimed to provide a summary on the regulation and function of SIRT1 in the vascular endothelium and to discuss the different approaches targeting this molecule for the prevention and treatment of age-related cardiovascular and cerebrovascular diseases.
Collapse
Affiliation(s)
- Musammat Kulsuma Begum
- The State Key Laboratory of Pharmaceutical Biotechnology
- The Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Daniels Konja
- The State Key Laboratory of Pharmaceutical Biotechnology
- The Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Sandeep Singh
- The State Key Laboratory of Pharmaceutical Biotechnology
- The Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland; and
- Chair of Pharmacology, Jagiellonian University Medical College, Krakow, Poland
| | - Yu Wang
- The State Key Laboratory of Pharmaceutical Biotechnology
- The Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
23
|
Poussin C, van der Toorn M, Scheuner S, Piault R, Kondylis A, Savioz R, Dulize R, Peric D, Guedj E, Maranzano F, Merg C, Morelli M, Egesipe AL, Johne S, Majeed S, Pak C, Schneider T, Schlage WK, Ivanov NV, Peitsch MC, Hoeng J. Systems toxicology study reveals reduced impact of heated tobacco product aerosol extract relative to cigarette smoke on premature aging and exacerbation effects in aged aortic cells in vitro. Arch Toxicol 2021; 95:3341-3359. [PMID: 34313809 PMCID: PMC8448694 DOI: 10.1007/s00204-021-03123-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 07/15/2021] [Indexed: 12/17/2022]
Abstract
Aging and smoking are major risk factors for cardiovascular diseases (CVD). Our in vitro study compared, in the context of aging, the effects of the aerosol of Tobacco Heating System 2.2 (THS; an electrically heated tobacco product) and 3R4F reference cigarette smoke (CS) on processes that contribute to vascular pathomechanisms leading to CVD. Young and old human aortic smooth muscle cells (HAoSMC) were exposed to various concentrations of aqueous extracts (AE) from 3R4F CS [0.014-0.22 puffs/mL] or THS aerosol [0.11-1.76 puffs/mL] for 24 h. Key markers were measured by high-content imaging, transcriptomics profiling and multianalyte profiling. In our study, in vitro aging increased senescence, DNA damage, and inflammation and decreased proliferation in the HAoSMCs. At higher concentrations of 3R4F AE, young HAoSMCs behaved similarly to aged cells, while old HAoSMCs showed additional DNA damage and apoptosis effects. At 3R4F AE concentrations with the maximum effect, the THS AE showed no significant effect in young or old HAoSMCs. It required an approximately ten-fold higher concentration of THS AE to induce effects similar to those observed with 3R4F. These effects were independent of nicotine, which did not show a significant effect on HAoSMCs at any tested concentration. Our results show that 3R4F AE accelerates aging in young HAoSMCs and exacerbates the aging effect in old HAoSMCs in vitro, consistent with CS-related contributions to the risk of CVD. Relative to 3R4F AE, the THS AE showed a significantly reduced impact on HAoSMCs, suggesting its lower risk for vascular SMC-associated pathomechanisms leading to CVD.
Collapse
Affiliation(s)
- Carine Poussin
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Marco van der Toorn
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland
| | - Sophie Scheuner
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland
| | - Romain Piault
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland
| | - Athanasios Kondylis
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland
| | - Rebecca Savioz
- Consultants in Science Sàrl, Biopole, Route de la Corniche 4, 1066, Epalinges, Switzerland
| | - Rémi Dulize
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland
| | - Dariusz Peric
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland
| | - Emmanuel Guedj
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland
| | - Fabio Maranzano
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland
| | - Celine Merg
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland
| | - Moran Morelli
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland
| | - Anne-Laure Egesipe
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland
| | - Stéphanie Johne
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland
| | - Shoaib Majeed
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland
| | - Claudius Pak
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland
| | - Thomas Schneider
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland
| | - Walter K Schlage
- Biology Consultant, Max-Baermann-Str. 21, 51429, Bergisch Gladbach, Germany
| | - Nikolai V Ivanov
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland
| | - Manuel C Peitsch
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland
| | - Julia Hoeng
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland
| |
Collapse
|
24
|
Ramprasath T, Han YM, Zhang D, Yu CJ, Zou MH. Tryptophan Catabolism and Inflammation: A Novel Therapeutic Target For Aortic Diseases. Front Immunol 2021; 12:731701. [PMID: 34630411 PMCID: PMC8496902 DOI: 10.3389/fimmu.2021.731701] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 09/03/2021] [Indexed: 12/14/2022] Open
Abstract
Aortic diseases are the primary public health concern. As asymptomatic diseases, abdominal aortic aneurysm (AAA) and atherosclerosis are associated with high morbidity and mortality. The inflammatory process constitutes an essential part of a pathogenic cascade of aortic diseases, including atherosclerosis and aortic aneurysms. Inflammation on various vascular beds, including endothelium, smooth muscle cell proliferation and migration, and inflammatory cell infiltration (monocytes, macrophages, neutrophils, etc.), play critical roles in the initiation and progression of aortic diseases. The tryptophan (Trp) metabolism or kynurenine pathway (KP) is the primary way of degrading Trp in most mammalian cells, disturbed by cytokines under various stress. KP generates several bioactive catabolites, such as kynurenine (Kyn), kynurenic acid (KA), 3-hydroxykynurenine (3-HK), etc. Depends on the cell types, these metabolites can elicit both hyper- and anti-inflammatory effects. Accumulating evidence obtained from various animal disease models indicates that KP contributes to the inflammatory process during the development of vascular disease, notably atherosclerosis and aneurysm development. This review outlines current insights into how perturbed Trp metabolism instigates aortic inflammation and aortic disease phenotypes. We also briefly highlight how targeting Trp metabolic pathways should be considered for treating aortic diseases.
Collapse
Affiliation(s)
| | | | | | | | - Ming-Hui Zou
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA, United States
| |
Collapse
|
25
|
Cyclic nucleotide phosphodiesterase 1C contributes to abdominal aortic aneurysm. Proc Natl Acad Sci U S A 2021; 118:2107898118. [PMID: 34312235 DOI: 10.1073/pnas.2107898118] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Abdominal aortic aneurysm (AAA) is characterized by aorta dilation due to wall degeneration, which mostly occurs in elderly males. Vascular aging is implicated in degenerative vascular pathologies, including AAA. Cyclic nucleotide phosphodiesterases, by hydrolyzing cyclic nucleotides, play critical roles in regulating vascular structure remodeling and function. Cyclic nucleotide phosphodiesterase 1C (PDE1C) expression is induced in dedifferentiated and aging vascular smooth muscle cells (SMCs), while little is known about the role of PDE1C in aneurysm. We observed that PDE1C was not expressed in normal aorta but highly induced in SMC-like cells in human and murine AAA. In mouse AAA models induced by Angiotensin II or periaortic elastase, PDE1C deficiency significantly decreased AAA incidence, aortic dilation, and elastin degradation, which supported a causative role of PDE1C in AAA development in vivo. Pharmacological inhibition of PDE1C also significantly suppressed preestablished AAA. We showed that PDE1C depletion antagonized SMC senescence in vitro and/or in vivo, as assessed by multiple senescence biomarkers, including senescence-associated β-galactosidase activity, γ-H2AX foci number, and p21 protein level. Interestingly, the role of PDE1C in SMC senescence in vitro and in vivo was dependent on Sirtuin 1 (SIRT1). Mechanistic studies further showed that cAMP derived from PDE1C inhibition stimulated SIRT1 activation, likely through a direct interaction between cAMP and SIRT1, which leads to subsequent up-regulation of SIRT1 expression. Our findings provide evidence that PDE1C elevation links SMC senescence to AAA development in both experimental animal models and human AAA, suggesting therapeutical significance of PDE1C as a potential target against aortic aneurysms.
Collapse
|
26
|
Yeap BB, Hui J, Knuiman MW, Flicker L, Divitini ML, Arscott GM, Twigg SM, Almeida OP, Hankey GJ, Golledge J, Norman PE, Beilby JP. U-Shaped Relationship of Leukocyte Telomere Length With All-Cause and Cancer-Related Mortality in Older Men. J Gerontol A Biol Sci Med Sci 2021; 76:164-171. [PMID: 32761187 DOI: 10.1093/gerona/glaa190] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Telomeres are essential DNA-protein complexes whose attrition results in cellular dysfunction and senescence. Leukocyte telomere length (LTL) correlates with tissue telomere length, representing a biomarker for biological age. However, its predictive value for mortality risk, and for cardiovascular versus cancer deaths, in older adults remains uncertain. METHOD We studied 3608 community-dwelling men aged 77.0 ± 3.6 years. Leukocyte telomere length was measured using multiplex quantitative PCR, expressed as amount of telomeric DNA relative to single-copy control gene (T/S ratio). Deaths from any cause, cardiovascular disease (CVD), and cancer were ascertained using data linkage. Curve fitting used restricted cubic splines and Cox regression analyses adjusted for age, cardiometabolic risk factors, and prevalent disease. RESULTS There was a U-shaped association of LTL with all-cause mortality. Men with T/S ratio in the middle quartiles had lower mortality (quartiles, Q2 vs Q1, hazard ratio [HR] = 0.86, 95% confidence interval [CI] 0.77-0.97, p = .012; Q3 vs Q1 HR = 0.88, CI 0.79-0.99, p = .032). There was no association of LTL with CVD mortality. There was a U-shaped association of LTL with cancer mortality. Men with LTL in the middle quartiles had lower risk of cancer death (Q2 vs Q1, HR = 0.73, CI 0.59-0.90, p = .004; Q3 vs Q1, HR = 0.75, CI 0.61-0.92, p = .007). CONCLUSIONS In older men, both shorter and longer LTL are associated with all-cause mortality. A similar U-shaped association was seen with cancer deaths, with no association found for cardiovascular deaths. Further research is warranted to explore the prognostic utility of LTL in ageing.
Collapse
Affiliation(s)
- Bu B Yeap
- Medical School, University of Western Australia, Perth, Australia.,Department of Endocrinology and Diabetes, Fiona Stanley Hospital, Perth, Western Australia, Australia
| | - Jennie Hui
- PathWest Laboratory Medicine, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia
| | - Matthew W Knuiman
- School of Population and Global Health, University of Western Australia, Perth, Australia
| | - Leon Flicker
- Medical School, University of Western Australia, Perth, Australia.,Department of Endocrinology, Sydney Medical School, University of Sydney, New South Wales, Australia
| | - Mark L Divitini
- School of Population and Global Health, University of Western Australia, Perth, Australia
| | - Gillian M Arscott
- PathWest Laboratory Medicine, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia
| | - Stephen M Twigg
- Department of Endocrinology, Sydney Medical School, University of Sydney, New South Wales, Australia
| | - Osvaldo P Almeida
- Medical School, University of Western Australia, Perth, Australia.,WA Centre for Health & Ageing, University of Western Australia, Perth, Australia
| | - Graeme J Hankey
- Medical School, University of Western Australia, Perth, Australia
| | - Jonathan Golledge
- Queensland Research Centre for Peripheral Vascular Disease, James Cook University, and Department of Vascular and Endovascular Surgery, Townsville Hospital, Queensland, Australia
| | - Paul E Norman
- Medical School, University of Western Australia, Perth, Australia
| | - John P Beilby
- PathWest Laboratory Medicine, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia
| |
Collapse
|
27
|
Zhao G, Chang Z, Zhao Y, Guo Y, Lu H, Liang W, Rom O, Wang H, Sun J, Zhu T, Fan Y, Chang L, Yang B, Garcia-Barrio MT, Chen YE, Zhang J. KLF11 protects against abdominal aortic aneurysm through inhibition of endothelial cell dysfunction. JCI Insight 2021; 6:141673. [PMID: 33507881 PMCID: PMC8021107 DOI: 10.1172/jci.insight.141673] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 01/27/2021] [Indexed: 12/13/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) is a life-threatening degenerative vascular disease. Endothelial cell (EC) dysfunction is implicated in AAA. Our group recently demonstrated that Krüppel-like factor 11 (KLF11) plays an essential role in maintaining vascular homeostasis, at least partially through inhibition of EC inflammatory activation. However, the functions of endothelial KLF11 in AAA remain unknown. Here we found that endothelial KLF11 expression was reduced in the ECs from human aneurysms and was time dependently decreased in the aneurysmal endothelium from both elastase- and Pcsk9/AngII-induced AAA mouse models. KLF11 deficiency in ECs markedly aggravated AAA formation, whereas EC-selective KLF11 overexpression markedly inhibited AAA formation. Mechanistically, KLF11 not only inhibited the EC inflammatory response but also diminished MMP9 expression and activity and reduced NADPH oxidase 2-mediated production of reactive oxygen species in ECs. In addition, KLF11-deficient ECs induced smooth muscle cell dedifferentiation and apoptosis. Overall, we established endothelial KLF11 as a potentially novel factor protecting against AAA and a potential target for intervention in aortic aneurysms.
Collapse
Affiliation(s)
- Guizhen Zhao
- Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Ziyi Chang
- Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, Michigan, USA
- Department of Pathology, China-Japan Friendship Hospital, Beijing, China
| | - Yang Zhao
- Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Yanhong Guo
- Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Haocheng Lu
- Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Wenying Liang
- Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Oren Rom
- Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Huilun Wang
- Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Jinjian Sun
- Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, Michigan, USA
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Tianqing Zhu
- Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Yanbo Fan
- Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, Michigan, USA
- Department of Cancer Biology, College of Medicine, University of Cincinnati, Cincinnati, Ohio. USA
| | - Lin Chang
- Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Bo Yang
- Department of Cardiac Surgery, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Minerva T. Garcia-Barrio
- Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Y. Eugene Chen
- Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, Michigan, USA
- Department of Cardiac Surgery, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Jifeng Zhang
- Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| |
Collapse
|
28
|
Cooper HA, Cicalese S, Preston KJ, Kawai T, Okuno K, Choi ET, Kasahara S, Uchida HA, Otaka N, Scalia R, Rizzo V, Eguchi S. Targeting mitochondrial fission as a potential therapeutic for abdominal aortic aneurysm. Cardiovasc Res 2021; 117:971-982. [PMID: 32384150 PMCID: PMC7898955 DOI: 10.1093/cvr/cvaa133] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 04/15/2020] [Accepted: 04/30/2020] [Indexed: 11/12/2022] Open
Abstract
AIMS Angiotensin II (AngII) is a potential contributor to the development of abdominal aortic aneurysm (AAA). In aortic vascular smooth muscle cells (VSMCs), exposure to AngII induces mitochondrial fission via dynamin-related protein 1 (Drp1). However, pathophysiological relevance of mitochondrial morphology in AngII-associated AAA remains unexplored. Here, we tested the hypothesis that mitochondrial fission is involved in the development of AAA. METHODS AND RESULTS Immunohistochemistry was performed on human AAA samples and revealed enhanced expression of Drp1. In C57BL6 mice treated with AngII plus β-aminopropionitrile, AAA tissue also showed an increase in Drp1 expression. A mitochondrial fission inhibitor, mdivi1, attenuated AAA size, associated aortic pathology, Drp1 protein induction, and mitochondrial fission but not hypertension in these mice. Moreover, western-blot analysis showed that induction of matrix metalloproteinase-2, which precedes the development of AAA, was blocked by mdivi1. Mdivi1 also reduced the development of AAA in apolipoprotein E-deficient mice infused with AngII. As with mdivi1, Drp1+/- mice treated with AngII plus β-aminopropionitrile showed a decrease in AAA compared to control Drp1+/+ mice. In abdominal aortic VSMCs, AngII induced phosphorylation of Drp1 and mitochondrial fission, the latter of which was attenuated with Drp1 silencing as well as mdivi1. AngII also induced vascular cell adhesion molecule-1 expression and enhanced leucocyte adhesion and mitochondrial oxygen consumption in smooth muscle cells, which were attenuated with mdivi1. CONCLUSION These data indicate that Drp1 and mitochondrial fission play salient roles in AAA development, which likely involves mitochondrial dysfunction and inflammatory activation of VSMCs.
Collapse
MESH Headings
- Aminopropionitrile
- Angiotensin II
- Animals
- Anti-Inflammatory Agents/pharmacology
- Aorta, Abdominal/drug effects
- Aorta, Abdominal/metabolism
- Aorta, Abdominal/pathology
- Aortic Aneurysm, Abdominal/chemically induced
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/prevention & control
- Case-Control Studies
- Cell Adhesion/drug effects
- Cells, Cultured
- Disease Models, Animal
- Dynamins/genetics
- Dynamins/metabolism
- Humans
- Leukocytes/drug effects
- Leukocytes/metabolism
- Male
- Mice, Inbred C57BL
- Mice, Knockout, ApoE
- Mitochondria, Muscle/drug effects
- Mitochondria, Muscle/genetics
- Mitochondria, Muscle/metabolism
- Mitochondria, Muscle/pathology
- Mitochondrial Dynamics/drug effects
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Oxygen Consumption/drug effects
- Phosphorylation
- Quinazolinones/pharmacology
- Mice
Collapse
Affiliation(s)
- Hannah A Cooper
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Stephanie Cicalese
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Kyle J Preston
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Tatsuo Kawai
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Keisuke Okuno
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Eric T Choi
- Department of Surgery, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Shingo Kasahara
- Department of Cardiovascular Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Haruhito A Uchida
- Department of Chronic Kidney Disease and Cardiovascular Disease, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Nozomu Otaka
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Rosario Scalia
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Victor Rizzo
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Satoru Eguchi
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
29
|
Chen T, Zhang H, Zhang Y, Yang M, Wu J, Yang M, Lin J, Gao W, Tang L, Xu B, Jiang J, Chen X. Association of Circulating and Aortic Zinc and Copper Levels with Clinical Abdominal Aortic Aneurysm: a Meta-analysis. Biol Trace Elem Res 2021; 199:513-526. [PMID: 32557106 DOI: 10.1007/s12011-020-02187-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 05/06/2020] [Indexed: 01/26/2023]
Abstract
It remains obscure whether circulating aortic zinc (Zn) and copper (Cu) levels are associated with the progress of human abdominal aortic aneurysms (AAA). Therefore, we conducted a meta-analysis to explore this relationship. A literature search on circulating and aortic zinc and copper levels and AAA patients was conducted using online databases including PubMed, Embase, and Cochrane up to March 20, 2019. To compare Zn and Cu concentrations in AAA patients with those in aortic occlusive disease (AOD) patients or healthy aorta donors or healthy blood donors, pooled weighted mean difference (WMD) and its 95% confidence interval (CI) were calculated. Subgroup analysis, sensitivity analysis, and meta-regression analysis were applied to explain the heterogeneity and evaluate the robustness of combined results. A total of 10 cross-sectional studies, including 252 cases and 304 controls, were used for meta-analysis. We found that circulating zinc and Zn/Cu ratio in AAA patients were significantly lower [WMD (95%CI): - 2.23 (- 4.10, - 0.36); - 0.18 (- 0.31, - 0.05), respectively] than those in non-AAA patients. Similarly, aneurysmal aorta had significantly lower zinc levels and Zn/Cu ratio [WMD (95%CI): - 9.22 (- 15.37, - 3.07); - 6.46 (- 10.14, - 2.77), respectively] than those in control group. No difference in circulating or aortic copper levels was noted between AAA patients and control group [WMD (95%CI): - 0.24 (- 2.09, 1.61); 0.30 (- 0.01, 0.61) , respectively]. Our meta-analysis suggests that zinc levels and Zn-Cu ratio, but not copper levels, may influence aneurysmal progress of AAA.
Collapse
Affiliation(s)
- Tingting Chen
- Department of Cardiology, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, 317000, Zhejiang Province, China
- Laboratory of Cardiovascular Disease, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, 317000, Zhejiang Province, China
| | - Hongliang Zhang
- Department of Cardiology, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, 317000, Zhejiang Province, China
- Laboratory of Cardiovascular Disease, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, 317000, Zhejiang Province, China
| | - Yang Zhang
- Department of Cardiology, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, 317000, Zhejiang Province, China
- Laboratory of Cardiovascular Disease, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, 317000, Zhejiang Province, China
| | - Mengqi Yang
- Department of Cardiology, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, 317000, Zhejiang Province, China
- Laboratory of Cardiovascular Disease, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, 317000, Zhejiang Province, China
| | - Juntao Wu
- Department of Cardiology, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, 317000, Zhejiang Province, China
- Laboratory of Cardiovascular Disease, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, 317000, Zhejiang Province, China
| | - Minjun Yang
- Department of Cardiology, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, 317000, Zhejiang Province, China
- Laboratory of Cardiovascular Disease, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, 317000, Zhejiang Province, China
| | - Jiangbo Lin
- Department of Cardiology, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, 317000, Zhejiang Province, China
- Laboratory of Cardiovascular Disease, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, 317000, Zhejiang Province, China
| | - Weixu Gao
- Department of Endocrinology, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, 317000, Zhejiang Province, China
| | - Lijiang Tang
- Department of Cardiology, Zhejiang Hospital, Hangzhou, 310013, Zhejiang Province, China
| | - Baohui Xu
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Jianjun Jiang
- Department of Cardiology, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, 317000, Zhejiang Province, China
- Laboratory of Cardiovascular Disease, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, 317000, Zhejiang Province, China
| | - Xiaofeng Chen
- Department of Cardiology, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, 317000, Zhejiang Province, China.
- Laboratory of Cardiovascular Disease, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, 317000, Zhejiang Province, China.
| |
Collapse
|
30
|
Ting KK, Coleman P, Zhao Y, Vadas MA, Gamble JR. The aging endothelium. VASCULAR BIOLOGY 2021; 3:R35-R47. [PMID: 33880430 PMCID: PMC8052565 DOI: 10.1530/vb-20-0013] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 01/12/2021] [Indexed: 01/10/2023]
Abstract
Cellular senescence is now recognized as one of the hallmarks of aging. Herein, we examine current findings on senescence of the vascular endothelium and its impacts on age-related vascular diseases. Endothelial senescence can result in systemic metabolic changes, implicating senescence in chronic diseases such as diabetes, obesity and atherosclerosis. Senolytics, drugs that eliminate senescent cells, afford new therapeutic strategies for control of these chronic diseases.
Collapse
Affiliation(s)
- Ka Ka Ting
- Centre for the Endothelium Vascular Biology Program Centenary Institute, The University of Sydney, Sydney, New South Wales, Australia
| | - Paul Coleman
- Centre for the Endothelium Vascular Biology Program Centenary Institute, The University of Sydney, Sydney, New South Wales, Australia
| | - Yang Zhao
- Centre for the Endothelium Vascular Biology Program Centenary Institute, The University of Sydney, Sydney, New South Wales, Australia
| | - Mathew A Vadas
- Centre for the Endothelium Vascular Biology Program Centenary Institute, The University of Sydney, Sydney, New South Wales, Australia
| | - Jennifer R Gamble
- Centre for the Endothelium Vascular Biology Program Centenary Institute, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
31
|
MicroRNA-34a: the bad guy in age-related vascular diseases. Cell Mol Life Sci 2021; 78:7355-7378. [PMID: 34698884 PMCID: PMC8629897 DOI: 10.1007/s00018-021-03979-4] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/08/2021] [Accepted: 10/12/2021] [Indexed: 12/12/2022]
Abstract
The age-related vasculature alteration is the prominent risk factor for vascular diseases (VD), namely, atherosclerosis, abdominal aortic aneurysm, vascular calcification (VC) and pulmonary arterial hypertension (PAH). The chronic sterile low-grade inflammation state, alias inflammaging, characterizes elderly people and participates in VD development. MicroRNA34-a (miR-34a) is emerging as an important mediator of inflammaging and VD. miR-34a increases with aging in vessels and induces senescence and the acquisition of the senescence-associated secretory phenotype (SASP) in vascular smooth muscle (VSMCs) and endothelial (ECs) cells. Similarly, other VD risk factors, including dyslipidemia, hyperglycemia and hypertension, modify miR-34a expression to promote vascular senescence and inflammation. miR-34a upregulation causes endothelial dysfunction by affecting ECs nitric oxide bioavailability, adhesion molecules expression and inflammatory cells recruitment. miR-34a-induced senescence facilitates VSMCs osteoblastic switch and VC development in hyperphosphatemia conditions. Conversely, atherogenic and hypoxic stimuli downregulate miR-34a levels and promote VSMCs proliferation and migration during atherosclerosis and PAH. MiR34a genetic ablation or miR-34a inhibition by anti-miR-34a molecules in different experimental models of VD reduce vascular inflammation, senescence and apoptosis through sirtuin 1 Notch1, and B-cell lymphoma 2 modulation. Notably, pleiotropic drugs, like statins, liraglutide and metformin, affect miR-34a expression. Finally, human studies report that miR-34a levels associate to atherosclerosis and diabetes and correlate with inflammatory factors during aging. Herein, we comprehensively review the current knowledge about miR-34a-dependent molecular and cellular mechanisms activated by VD risk factors and highlight the diagnostic and therapeutic potential of modulating its expression in order to reduce inflammaging and VD burn and extend healthy lifespan.
Collapse
|
32
|
Zhang W, Cheng W, Parlato R, Guo X, Cui X, Dai C, Xu L, Zhu J, Zhu M, Luo K, Zhang W, Dong B, Wang J, Jiang F. Nucleolar stress induces a senescence-like phenotype in smooth muscle cells and promotes development of vascular degeneration. Aging (Albany NY) 2020; 12:22174-22198. [PMID: 33146634 PMCID: PMC7695416 DOI: 10.18632/aging.104094] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 08/31/2020] [Indexed: 12/24/2022]
Abstract
Senescence of smooth muscle cells (SMCs) has a crucial role in the pathogenesis of abdominal aortic aneurysm (AAA), a disease of vascular degeneration. Perturbation of cellular ribosomal DNA (rDNA) transcription triggers nucleolar stress response. Previously we demonstrated that induction of nucleolar stress in SMCs elicited cell cycle arrest via the ataxia-telangiectasia mutated (ATM)/ATM- and Rad3-related (ATR)-p53 axis. However, the specific roles of nucleolar stress in vascular degeneration remain unexplored. In the present study, we demonstrated for the first time that in both human and animal AAA tissues, there were non-coordinated changes in the expression of RNA polymerase I machinery components, including a downregulation of transcription initiation factor-IA (TIF-IA). Genetic deletion of TIF-IA in SMCs in mice (smTIF-IA-/-) caused spontaneous aneurysm-like lesions in the aorta. In vitro, induction of nucleolar stress triggered a non-canonical DNA damage response, leading to p53 phosphorylation and a senescence-like phenotype in SMCs. In human AAA tissues, there was increased nucleolar stress in medial cells, accompanied by localized DNA damage response within the nucleolar compartment. Our data suggest that perturbed rDNA transcription and induction of nucleolar stress contribute to the pathogenesis of AAA. Moreover, smTIF-IA-/- mice may be a novel animal model for studying spontaneous AAA-like vascular degenerations.
Collapse
Affiliation(s)
- Wenjing Zhang
- Department of Physiology and Pathophysiology, School of Basic Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China.,Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
| | - Wen Cheng
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, and The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Rosanna Parlato
- Institute of Applied Physiology, University of Ulm, Ulm, Germany.,Institute of Anatomy and Cell Biology, University of Heidelberg, Heidelberg, Germany
| | - Xiaosun Guo
- Department of Physiology and Pathophysiology, School of Basic Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Xiaopei Cui
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
| | - Chaochao Dai
- Department of Physiology and Pathophysiology, School of Basic Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China.,Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
| | - Lei Xu
- Department of Vascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jiankang Zhu
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Min Zhu
- Department of Transplant Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Kun Luo
- Department of Vascular Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Wencheng Zhang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, and The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Bo Dong
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong Province, China
| | - Jianli Wang
- Department of Physiology and Pathophysiology, School of Basic Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China.,Current address: Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Fan Jiang
- Department of Physiology and Pathophysiology, School of Basic Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China.,Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
33
|
Specific miRNA and Gene Deregulation Characterize the Increased Angiogenic Remodeling of Thoracic Aneurysmatic Aortopathy in Marfan Syndrome. Int J Mol Sci 2020; 21:ijms21186886. [PMID: 32961817 PMCID: PMC7555983 DOI: 10.3390/ijms21186886] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/17/2020] [Accepted: 09/19/2020] [Indexed: 12/11/2022] Open
Abstract
Marfan syndrome (MFS) is a connective tissue disease caused by mutations in the FBN1 gene, leading to alterations in the extracellular matrix microfibril assembly and the early formation of thoracic aorta aneurysms (TAAs). Non-genetic TAAs share many clinico-pathological aspects with MFS and deregulation of some microRNAs (miRNAs) has been demonstrated to be involved in the progression of TAA. In this study, 40 patients undergoing elective ascending aorta surgery were enrolled to compare TAA histomorphological features, miRNA profile and related target genes in order to find specific alterations that may explain the earlier and more severe clinical outcomes in MFS patients. Histomorphological, ultrastructural and in vitro studies were performed in order to compare aortic wall features of MFS and non-MFS TAA. MFS displayed greater glycosaminoglycan accumulation and loss/fragmentation of elastic fibers compared to non-MFS TAA. Immunohistochemistry revealed increased CD133+ angiogenic remodeling, greater MMP-2 expression, inflammation and smooth muscle cell (SMC) turnover in MFS TAA. Cultured SMCs from MFS confirmed higher turnover and α-smooth muscle actin expression compared with non-MFS TAA. Moreover, twenty-five miRNAs, including miR-26a, miR-29, miR-143 and miR-145, were found to be downregulated and only miR-632 was upregulated in MFS TAA in vivo. Bioinformatics analysis revealed that some deregulated miRNAs in MFS TAA are implicated in cell proliferation, extracellular matrix structure/function and TGFβ signaling. Finally, gene analysis showed 28 upregulated and seven downregulated genes in MFS TAA, some of them belonging to the CDH1/APC and CCNA2/TP53 signaling pathways. Specific miRNA and gene deregulation characterized the aortopathy of MFS and this was associated with increased angiogenic remodeling, likely favoring the early and more severe clinical outcomes, compared to non-MFS TAA. Our findings provide new insights concerning the pathogenetic mechanisms of MFS TAA; further investigation is needed to confirm if these newly identified specific deregulated miRNAs may represent potential therapeutic targets to counteract the rapid progression of MFS aortopathy.
Collapse
|
34
|
Song P, Zhao Q, Zou MH. Targeting senescent cells to attenuate cardiovascular disease progression. Ageing Res Rev 2020; 60:101072. [PMID: 32298812 DOI: 10.1016/j.arr.2020.101072] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 04/07/2020] [Accepted: 04/09/2020] [Indexed: 12/19/2022]
Abstract
Cardiovascular disease (CVD) is the most common disease to increase as life expectancy increases. Most high-profile pharmacological treatments for age-related CVD have led to inefficacious results, implying that novel approaches to treating these pathologies are needed. Emerging data have demonstrated that senescent cardiovascular cells, which are characterized by irreversible cell cycle arrest and a distinct senescence-associated secretory phenotype, accumulate in aged or diseased cardiovascular systems, suggesting that they may impair cardiovascular function. This review discusses the evidence implicating senescent cells in cardiovascular ageing, the onset and progression of CVD, and the molecular mechanisms underlying cardiovascular cell senescence. We also review eradication of senescent cardiovascular cells by small-molecule-drug-mediated apoptosis and immune cell-mediated efferocytosis and toxicity as promising and precisely targeted therapeutics for CVD prevention and treatment.
Collapse
|
35
|
Serino A, Zhao Y, Hwang J, Cullen A, Deeb C, Akhavan N, Arjmandi B, Salazar G. Gender differences in the effect of blackberry supplementation in vascular senescence and atherosclerosis in ApoE -/- mice. J Nutr Biochem 2020; 80:108375. [PMID: 32248057 DOI: 10.1016/j.jnutbio.2020.108375] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 01/09/2020] [Accepted: 03/07/2020] [Indexed: 12/20/2022]
Abstract
As the cardiovascular system ages, it becomes more vulnerable to the effects of oxidative stress and inflammation. The aging process, along with external factors such as radiation exposure and lifestyle, induces vascular senescence and accelerates atherosclerotic plaque accumulation. Expression of nicotinamide adenine dinucleotide phosphate oxidase 1 (Nox1), which produces superoxide, is associated with senescence in vascular smooth muscle cells in vitro and atherosclerosis in ApoE-/- mice in vivo. However, it is unknown whether Nox1 could be down-regulated by nutritional interventions aimed to reduce atherosclerosis. Here we study the effect of blackberry supplementation in Nox1 expression and atherosclerosis. Four-month-old ApoE-/- male and female mice were fed low-fat, high-fat or high-fat supplemented with 2% freeze-dried blackberry powder diets for 5 weeks. Analysis of the aorta showed that diet supplemented with blackberry significantly decreased plaque accumulation, senescence associated-β-galactosidase and Nox1 expression in the aorta of male but not female mice. The lipid profile was unchanged by blackberry in both female and male animals. Thus, the known role of Nox1 in atherosclerosis suggests that the atheroprotective effect of blackberry is mediated by Nox1 down-regulation in male mice and that Nox1 is regulated in a gender-dependent manner in females.
Collapse
Affiliation(s)
- Alexa Serino
- Department of Nutrition, Food and Exercise Sciences, Florida State University, Tallahassee, FL, USA
| | - Yitong Zhao
- Division of Nephrology and Hypertension, Department of Medicine, University of California, Irvine, CA, USA
| | - Jingwen Hwang
- Department of Nutrition, Food and Exercise Sciences, Florida State University, Tallahassee, FL, USA
| | - Abigail Cullen
- Department of Nutrition, Food and Exercise Sciences, Florida State University, Tallahassee, FL, USA
| | - Carolyn Deeb
- Department of Nutrition, Food and Exercise Sciences, Florida State University, Tallahassee, FL, USA
| | - Neda Akhavan
- Department of Nutrition, Food and Exercise Sciences, Florida State University, Tallahassee, FL, USA
| | - Bahram Arjmandi
- Department of Nutrition, Food and Exercise Sciences, Florida State University, Tallahassee, FL, USA; Center for Advancing Exercise and Nutrition Research on Aging (CAENRA), Florida State University, Tallahassee, FL, USA
| | - Gloria Salazar
- Department of Nutrition, Food and Exercise Sciences, Florida State University, Tallahassee, FL, USA; Center for Advancing Exercise and Nutrition Research on Aging (CAENRA), Florida State University, Tallahassee, FL, USA.
| |
Collapse
|
36
|
Spartalis E, Spartalis M, Athanasiou A, Paschou SA, Patelis N, Voudris V, Iliopoulos DC. Endothelium in Aortic Aneurysm Disease: New Insights. Curr Med Chem 2020; 27:1081-1088. [PMID: 31549591 DOI: 10.2174/0929867326666190923151959] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 04/10/2019] [Accepted: 04/28/2019] [Indexed: 01/12/2023]
Abstract
Inflammation is recognized as a fundamental element in the development and growth of aortic aneurysms. Aortic aneurysm is correlated with aortic wall deformities and injury, as a result of inflammation, matrix metalloproteinases activation, oxidative stress, and apoptosis of vascular smooth muscle cells. The endothelial wall has a critical part in the inflammation of the aorta and endothelial heterogeneity has proven to be significant for modeling aneurysm formation. Endothelial shear stress and blood flow affect the aortic wall through hindrance of cytokines and adhesion molecules excreted by endothelial cells, causing reduction of the inflammation process in the media and adventitia. This pathophysiological process results in the disruption of elastic fibers, degradation of collagen fibers, and destruction of vascular smooth muscle cells. Consequently, the aortic wall is impaired due to reduced thickness, decreased mechanical function, and cannot tolerate the impact of blood flow leading to aortic expansion. Surgery is still considered the mainstay therapy for large aortic aneurysms. The prevention of aortic dilation, though, is based on the hinderance of endothelial dysregulation with drugs, the reduction of reactive oxygen and nitrogen species, and also the reduction of pro-inflammatory molecules and metalloproteinases. Further investigations are required to enlighten the emerging role of endothelial cells in aortic disease.
Collapse
Affiliation(s)
- Eleftherios Spartalis
- Laboratory of Experimental Surgery and Surgical Research, University of Athens, Medical School, Athens, Greece
| | - Michael Spartalis
- Division of Cardiology, Onassis Cardiac Surgery Center, Athens, Greece
| | - Antonios Athanasiou
- Laboratory of Experimental Surgery and Surgical Research, University of Athens, Medical School, Athens, Greece
| | - Stavroula A. Paschou
- Division of Endocrinology and Diabetes, "Aghia Sophia" Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Nikolaos Patelis
- Laboratory of Experimental Surgery and Surgical Research, University of Athens, Medical School, Athens, Greece
| | - Vassilis Voudris
- Division of Cardiology, Onassis Cardiac Surgery Center, Athens, Greece
| | - Dimitrios C. Iliopoulos
- Laboratory of Experimental Surgery and Surgical Research, University of Athens, Medical School, Athens, Greece
| |
Collapse
|
37
|
Pejenaute Á, Cortés A, Marqués J, Montero L, Beloqui Ó, Fortuño A, Martí A, Orbe J, Zalba G. NADPH Oxidase Overactivity Underlies Telomere Shortening in Human Atherosclerosis. Int J Mol Sci 2020; 21:ijms21041434. [PMID: 32093292 PMCID: PMC7073034 DOI: 10.3390/ijms21041434] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 02/10/2020] [Accepted: 02/18/2020] [Indexed: 11/16/2022] Open
Abstract
Telomere shortening and oxidative stress are involved in the pathogenesis of atherosclerosis. Different studies have shown that phagocytic NADPH oxidase is associated with this disease. This study aimed to investigate the association between phagocytic NADPH oxidase and telomere shortening in human atherosclerosis. To assess this potential association, telomere length and phagocytic NADPH oxidase activity were determined by PCR and chemiluminescence, respectively, in a population of asymptomatic subjects free of overt clinical atherosclerosis. We also measured serum 8-hydroxy-2-deoxyguanosine (8-OHdG) levels (an index of oxidative stress) and carotid intima-media thickness (IMT), a surrogate marker of atherosclerosis. After adjusting them for age and sex, telomere length inversely correlated (p < 0.05) with NADPH oxidase-mediated superoxide production, with 8-OHdG values, and with carotid IMT. Interestingly, the asymptomatic subjects with plaques have a lower telomere length (p < 0.05), and higher values of plasma 8-OHdG and superoxide production (p < 0.05). These data were confirmed in a second population in which patients with coronary artery disease showed lower telomere length and higher 8-OHdG and superoxide production than the asymptomatic subjects. In both studies, NADPH oxidase-dependent superoxide production in phagocytic cells was only due to the specific expression of the Nox2 isoform. In conclusion, these findings suggest that phagocytic NADPH oxidase may be involved in oxidative stress-mediated telomere shortening, and that this axis may be critically involved in human atherosclerosis.
Collapse
Affiliation(s)
- Álvaro Pejenaute
- Department of Biochemistry and Genetics, University of Navarra, 31008 Pamplona, Spain; (Á.P.); (A.C.); (J.M.); (L.M.)
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain; (Ó.B.); (A.M.); (J.O.)
| | - Adriana Cortés
- Department of Biochemistry and Genetics, University of Navarra, 31008 Pamplona, Spain; (Á.P.); (A.C.); (J.M.); (L.M.)
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain; (Ó.B.); (A.M.); (J.O.)
| | - Javier Marqués
- Department of Biochemistry and Genetics, University of Navarra, 31008 Pamplona, Spain; (Á.P.); (A.C.); (J.M.); (L.M.)
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain; (Ó.B.); (A.M.); (J.O.)
| | - Laura Montero
- Department of Biochemistry and Genetics, University of Navarra, 31008 Pamplona, Spain; (Á.P.); (A.C.); (J.M.); (L.M.)
| | - Óscar Beloqui
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain; (Ó.B.); (A.M.); (J.O.)
- Department of Internal Medicine, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Ana Fortuño
- Program of Cardiovascular Diseases, CIMA, University of Navarra, 31008 Pamplona, Spain;
| | - Amelia Martí
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain; (Ó.B.); (A.M.); (J.O.)
- Department of Food Sciences and Physiology, University of Navarra, 31008 Pamplona, Spain
| | - Josune Orbe
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain; (Ó.B.); (A.M.); (J.O.)
- Program of Cardiovascular Diseases, CIMA, University of Navarra, 31008 Pamplona, Spain;
| | - Guillermo Zalba
- Department of Biochemistry and Genetics, University of Navarra, 31008 Pamplona, Spain; (Á.P.); (A.C.); (J.M.); (L.M.)
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain; (Ó.B.); (A.M.); (J.O.)
- Correspondence:
| |
Collapse
|
38
|
Shimizu I, Minamino T. Cellular Senescence in Arterial Diseases. J Lipid Atheroscler 2020; 9:79-91. [PMID: 32821723 PMCID: PMC7379072 DOI: 10.12997/jla.2020.9.1.79] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 12/25/2019] [Accepted: 12/25/2019] [Indexed: 12/11/2022] Open
Abstract
Cell-proliferation potency is limited, as cells cannot proceed through the cell cycle continually. Instead, they eventually show an irreversible arrest of proliferation, commonly referred to as cellular senescence. Following the initial discovery of this phenomenon by Hayflick et al., studies have indicated that cells are also destined to undergo aging. In addition to the irreversible termination of proliferation, senescent cells are characterized by a flattened and enlarged morphology. Senescent cells become pro-inflammatory and contribute to the initiation and maintenance of sustained chronic sterile inflammation. Aging is associated with the accumulation of senescent cells in the cardiovascular system, and in general these cells are considered to be pathogenic because they mediate vascular remodeling. Recently, genetic and pharmacological approaches have enabled researchers to eliminate senescent cells both in vitro and in vivo. The term “senolysis” is now used to refer to the depletion of senescent cells, and evidence indicates that senolysis contributes to the reversal of age-related pathogenic phenotypes without the risk of tumorigenesis. The concept of senolysis has opened new avenues in research on aging, and senolysis may be a promising therapeutic approach for combating age-related disorders, including arterial diseases.
Collapse
Affiliation(s)
- Ippei Shimizu
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.,Division of Molecular Aging and Cell Biology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.,Japan Agency for Medical Research and Development-Core Research for Evolutionary Medical Science and Technology (AMED-CREST), Japan Agency for Medical Research and Development, Tokyo, Japan
| |
Collapse
|
39
|
Harky A, Fan KS, Fan KH. The genetics and biomechanics of thoracic aortic diseases. VASCULAR BIOLOGY 2019; 1:R13-R25. [PMID: 32923967 PMCID: PMC7439919 DOI: 10.1530/vb-19-0027] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 10/15/2019] [Indexed: 12/20/2022]
Abstract
Thoracic aortic aneurysms and aortic dissections (TAAD) are highly fatal emergencies within cardiothoracic surgery. With increasing age, thoracic aneurysms become more prevalent and pose an even greater threat when they develop into aortic dissections. Both diseases are multifactorial and are influenced by a multitude of physiological and biomechanical processes. Structural stability of aorta can be disrupted by genes, such as those for extracellular matrix and contractile protein, as well as telomere dysfunction, which leads to senescence of smooth muscle and endothelial cells. Biomechanical changes such as increased luminal pressure imposed by hypertension are also very prevalent and lead to structural instability. Furthermore, ageing is associated with a pro-inflammatory state that exacerbates degeneration of vessel wall, facilitating the development of both aortic aneurysms and aortic dissection. This literature review provides an overview of the aetiology and pathophysiology of both thoracic aneurysms and aortic dissections. With an improved understanding, new therapeutic targets may eventually be identified to facilitate treatment and prevention of these diseases.
Collapse
Affiliation(s)
- Amer Harky
- Department of Cardiothoracic Surgery, Liverpool Heart and Chest, Liverpool, UK
| | - Ka Siu Fan
- St. George's Medical School, University of London, London, UK
| | - Ka Hay Fan
- Faculty of Medicine, Imperial College London, London, UK
| |
Collapse
|
40
|
The pseudogene PTENP1 regulates smooth muscle cells as a competing endogenous RNA. Clin Sci (Lond) 2019; 133:1439-1455. [PMID: 31235554 DOI: 10.1042/cs20190156] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 05/30/2019] [Accepted: 06/24/2019] [Indexed: 02/06/2023]
Abstract
The long non-coding RNA (lncRNA) PTENP1 is a pseudogene of phosphatase and tensin homologue deleted on chromosome ten (PTEN), has been implicated in smooth muscle cell (SMC) proliferation and apoptosis. PTENP1 is the pseudogene of PTEN. However, it is unclear whether and how PTENP1 functions in the proliferation and apoptosis of human aortic SMCs (HASMCs). Here, we hypothesised that PTENP1 inhibits HASMC proliferation and enhances apoptosis by promoting PTEN expression. PCR analysis and Western blot assays respectively showed that both PTENP1 and PTEN were up-regulated in human aortic dissection (AD) samples. PTENP1 overexpression significantly increased the protein expression of PTEN, promoted apoptosis and inhibited the proliferation of HASMCs. PTENP1 silencing exhibited the opposite effects and mitigated H2O2-induced apoptosis of HASMCs. In an angiotensin II (Ang II)-induced mouse aortic aneurysm (AA) model, PTENP1 overexpression potentiated aortic SMC apoptosis, exacerbated aneurysm formation. Mechanistically, RNA pull-down assay and a series of luciferase reporter assays using miR-21 mimics or inhibitors identified PTENP1 as a molecular sponge for miR-21 to endogenously compete for the binding between miR-21 and the PTEN transcript, releasing PTEN expression. This finding was further supported by in vitro immunofluorescent evidence showing decreased cell apoptosis upon miR-21 mimic administration under baseline PTENP1 overexpression. Ex vivo rescue of PTEN significantly mitigated the SMC apoptosis induced by PTENP1 overexpression. Finally, Western blot assays showed substantially reduced Akt phosphorylation and cyclin D1 and cyclin E levels with up-regulated PTENP1 in HASMCs. Our study identified PTENP1 as a mediator of HASMC homeostasis and suggests that PTENP1 is a potential target in AD or AA intervention.
Collapse
|
41
|
Carpinus turczaninowii Extract May Alleviate High Glucose-Induced Arterial Damage and Inflammation. Antioxidants (Basel) 2019; 8:antiox8060172. [PMID: 31212679 PMCID: PMC6616550 DOI: 10.3390/antiox8060172] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 06/02/2019] [Accepted: 06/07/2019] [Indexed: 01/06/2023] Open
Abstract
Hyperglycemia-induced oxidative stress triggers severe vascular damage and induces an inflammatory vascular state, and is, therefore, one of the main causes of atherosclerosis. Recently, interest in the natural compound Carpinus turczaninowii has increased because of its reported antioxidant and anti-inflammatory properties. We investigated whether a C. turczaninowii extract was capable of attenuating high glucose-induced inflammation and arterial damage using human aortic vascular smooth muscle cells (hASMCs). mRNA expression levels of proinflammatory response [interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α)], endoplasmic reticulum (ER) stress [CCAAT-enhancer-binding proteins (C/EBP) homologous protein (CHOP)], and adenosine monophosphate (AMP)-protein activated kinase α2 (AMPK α2)], and DNA damage [phosphorylated H2.AX (p-H2.AX)] were measured in hASMCs treated with the C. turczaninowii extracts (1 and 10 μg/mL) after being stimulated by high glucose (25 mM) or not. The C. turczaninowii extract attenuated the increased mRNA expression of IL-6, TNF-α, and CHOP in hASMCs under high glucose conditions. The expression levels of p-H2.AX and AMPK α2 induced by high glucose were also significantly decreased in response to treatment with the C. turczaninowii extract. In addition, 15 types of phenolic compounds including quercetin, myricitrin, and ellagic acid, which exhibit antioxidant and anti-inflammatory properties, were identified in the C. turczaninowii extract through ultra-performance liquid chromatography-quadrupole-time of flight (UPLC-Q-TOF) mass spectrometry. In conclusion, C. turczaninowii may alleviate high glucose-induced inflammation and arterial damage in hASMCs, and may have potential in the treatment of hyperglycemia-induced atherosclerosis.
Collapse
|
42
|
Cold parenting is associated with cellular aging in offspring: A retrospective study. Biol Psychol 2019; 145:142-149. [PMID: 31014776 DOI: 10.1016/j.biopsycho.2019.03.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 01/30/2019] [Accepted: 03/22/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND Early life stress is a known risk factor for diseases and premature death. We tested whether parenting style impacts telomere length (TL), a cellular aging biomarker. METHODS Information on parents' style of parenting was obtained from 199 participants in the Adventist Health Study-1 (AHS-1) who 27+ years later also enrolled in the AHS-2 where blood was collected for relative TL (rTL) assessment. RESULTS Subjects describing their mothers' parenting style as cold had on average 25% smaller rTL compared to subjects not reporting a cold mother (1.89 vs 2.53). This association was greatest among those with less education, and those who stayed overweight/obese or put on weight during follow-up. CONCLUSIONS These results support previous findings that early life stress may have health implications by promoting cellular aging, and expands these stressors to include cold parenting during an individuals' formative years. Higher education and normal weight seem to provide some resilience.
Collapse
|
43
|
Debelec-Butuner B, Bostancı A, Ozcan F, Singin O, Karamil S, Aslan M, Roggenbuck D, Korkmaz KS. Oxidative DNA Damage-Mediated Genomic Heterogeneity Is Regulated by NKX3.1 in Prostate Cancer. Cancer Invest 2019; 37:113-126. [PMID: 30836777 DOI: 10.1080/07357907.2019.1576192] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The 8-hydroxy-2'-deoxyguanosine (8-OHdG) damages are base damages induced by reactive oxygen species. We aimed to investigate the role of Androgen Receptor and NKX3.1 in 8-OHdG formation and repair activation by quantitating the DNA damage using Aklides.NUK system. The data demonstrated that the loss of NKX3.1 resulted in increased oxidative DNA damage and its overexpression contributes to the removal of menadione-induced 8-OHdG damage even under oxidative stress conditions. Moreover, 8-oxoguanine DNA glycosylase-1 (OGG1) expression level positively correlates to NKX3.1 expression. Also in this study, first time a reliable cell-based quantitation method for 8-OHdG damages is reported and used for data collection.
Collapse
Affiliation(s)
- Bilge Debelec-Butuner
- a Department of Pharmaceutical Biotechnology, Faculty of Pharmacy , Ege University , Izmir , Turkey
| | - Aykut Bostancı
- b Department of Bioengineering, Cancer Biology Laboratory, Faculty of Engineering , Ege University , Izmir , Turkey
| | - Filiz Ozcan
- c Mass Spectrometry Laboratory, Department of Biochemistry, Faculty of Medicine , Akdeniz University , Antalya , Turkey
| | - Oznur Singin
- b Department of Bioengineering, Cancer Biology Laboratory, Faculty of Engineering , Ege University , Izmir , Turkey
| | - Selda Karamil
- b Department of Bioengineering, Cancer Biology Laboratory, Faculty of Engineering , Ege University , Izmir , Turkey
| | - Mutay Aslan
- c Mass Spectrometry Laboratory, Department of Biochemistry, Faculty of Medicine , Akdeniz University , Antalya , Turkey
| | - Dirk Roggenbuck
- d Medipan GmBH , Dahlewitz , Germany.,e Faculty Environment and Natural Sciences , Brandenburg University of Technology Cottbus-Senftenberg , Senftenberg , Germany
| | - Kemal Sami Korkmaz
- b Department of Bioengineering, Cancer Biology Laboratory, Faculty of Engineering , Ege University , Izmir , Turkey
| |
Collapse
|
44
|
Aurelian SV, Adrian M, Andercou O, Bruno S, Alexandru O, Catalin T, Dan B. Neutrophil-to-Lymphocyte Ratio: A Comparative Study of Rupture to Nonruptured Infrarenal Abdominal Aortic Aneurysm. Ann Vasc Surg 2019; 58:270-275. [PMID: 30769065 DOI: 10.1016/j.avsg.2018.11.026] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 10/20/2018] [Accepted: 11/09/2018] [Indexed: 11/16/2022]
Abstract
BACKGROUND Neutrophil-to-lymphocyte ratio (NLR) has recently emerged as a useful predictor of cardiovascular risk and adverse outcomes. According to previous studies, an NLR >5 has the highest sensitivity and specificity for postoperative morbidity and mortality in cardiovascular disease. This study aims to evaluate the NLR in cases of infrarenal unruptured abdominal aortic aneurysm (uAAA) and ruptured abdominal aortic aneurysm (rAAA) and to assess the role of NLR as a prognostic marker of 30-day mortality in patients with uAAA and rAAA who underwent surgical repair. METHODS This retrospective cohort study examined 255 consecutive patients with intact or ruptured infrarenal AAA who underwent elective or urgent open repair surgery within our clinic in a 10-year period. Differences in prevalence were assessed using chi-squared calculations and values greater than 5 and a P-value less than 0.05 were considered significant. The averages were compared using the ANOVA parameter test when the Bartlett P-value was greater than 0.05. RESULTS The average NLR appeared to be significantly higher in the group of patients with rAAA (9.3 vs. 3.39, respectively P < 0001). Furthermore, NLR > 5 occurred in 77.6% of patients with rAAA but only 32.5% in patients with uAAA (odds ratio 5.085; 95% confidence interval [CI]: 3.0025-8.6145; P < 0000.1). In terms of the postoperative prognosis in patients with uAAA, mortality after 30 days postoperatively was considerably higher at 16.6% in patients with NLR >5 compared with 6% for patients with NLR < 5 (RR: 2.77; 95% CI: 1.020-7.55; P < 0.045). In the case of rAAA, mortality after 30 days was higher in patients with NLR >5 (61.44%) than those with NLR < 5 (45.83%). There was no relationship between NLR and length of hospital stay or between NLR and the maximum diameter of the AAA. There was also no difference in the NLR between genders or age groups. CONCLUSIONS The main findings of this study were the poor outcomes in terms of 30-day mortality for the patients presenting NLR values greater than 5 undergoing open surgical repair in both categories: infrarenal uAAA and rAAA. We also show that NLR is significantly higher among patients with rAAA and that an NLR >5 indicates a 5 times greater possibility of AAA being ruptured. We can use this easily determinable, broadly available, and inexpensive marker to identify high-risk patients, individually, or integrated into a risk-stratification system for patients diagnosed with AAA. This would help in the therapeutic management of AAA, including the avoidance of open surgery when there are prohibitive risks, instead opting for an endovascular approach.
Collapse
Affiliation(s)
- Sasarman Vasile Aurelian
- Regional Hospital Center Metz-Thionville, Hôpital de Mercy, Metz, France; Heart Institute "Niculae Stancioiu" Cluj-Napoca, Cluj-Napoca, Roumania
| | - Molnar Adrian
- Heart Institute "Niculae Stancioiu" Cluj-Napoca, Cluj-Napoca, Roumania.
| | | | - Schjoth Bruno
- Regional Hospital Center Metz-Thionville, Hôpital de Mercy, Metz, France
| | - Oprea Alexandru
- Heart Institute "Niculae Stancioiu" Cluj-Napoca, Cluj-Napoca, Roumania
| | - Trifan Catalin
- Heart Institute "Niculae Stancioiu" Cluj-Napoca, Cluj-Napoca, Roumania
| | - Bindea Dan
- Heart Institute "Niculae Stancioiu" Cluj-Napoca, Cluj-Napoca, Roumania
| |
Collapse
|
45
|
Protective Role of Polyphenols against Vascular Inflammation, Aging and Cardiovascular Disease. Nutrients 2018; 11:nu11010053. [PMID: 30597847 PMCID: PMC6357531 DOI: 10.3390/nu11010053] [Citation(s) in RCA: 153] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 12/19/2018] [Accepted: 12/21/2018] [Indexed: 01/02/2023] Open
Abstract
Aging is a major risk factor in the development of chronic diseases affecting various tissues including the cardiovascular system, muscle and bones. Age-related diseases are a consequence of the accumulation of cellular damage and reduced activity of protective stress response pathways leading to low-grade systemic inflammation and oxidative stress. Both inflammation and oxidative stress are major contributors to cellular senescence, a process in which cells stop proliferating and become dysfunctional by secreting inflammatory molecules, reactive oxygen species (ROS) and extracellular matrix components that cause inflammation and senescence in the surrounding tissue. This process is known as the senescence associated secretory phenotype (SASP). Thus, accumulation of senescent cells over time promotes the development of age-related diseases, in part through the SASP. Polyphenols, rich in fruits and vegetables, possess antioxidant and anti-inflammatory activities associated with protective effects against major chronic diseases, such as cardiovascular disease (CVD). In this review, we discuss molecular mechanisms by which polyphenols improve anti-oxidant capacity, mitochondrial function and autophagy, while reducing oxidative stress, inflammation and cellular senescence in vascular smooth muscle cells (VSMCs) and endothelial cells (ECs). We also discuss the therapeutic potential of polyphenols in reducing the effects of the SASP and the incidence of CVD.
Collapse
|
46
|
Abstract
Advancing age promotes cardiovascular disease (CVD), the leading cause of death in the United States and many developed nations. Two major age-related arterial phenotypes, large elastic artery stiffening and endothelial dysfunction, are independent predictors of future CVD diagnosis and likely are responsible for the development of CVD in older adults. Not limited to traditional CVD, these age-related changes in the vasculature also contribute to other age-related diseases that influence mammalian health span and potential life span. This review explores mechanisms that influence age-related large elastic artery stiffening and endothelial dysfunction at the tissue level via inflammation and oxidative stress and at the cellular level via Klotho and energy-sensing pathways (AMPK [AMP-activated protein kinase], SIRT [sirtuins], and mTOR [mammalian target of rapamycin]). We also discuss how long-term calorie restriction-a health span- and life span-extending intervention-can prevent many of these age-related vascular phenotypes through the prevention of deleterious alterations in these mechanisms. Lastly, we discuss emerging novel mechanisms of vascular aging, including senescence and genomic instability within cells of the vasculature. As the population of older adults steadily expands, elucidating the cellular and molecular mechanisms of vascular dysfunction with age is critical to better direct appropriate and measured strategies that use pharmacological and lifestyle interventions to reduce risk of CVD within this population.
Collapse
Affiliation(s)
- Anthony J. Donato
- University of Utah, Department of Internal Medicine, Division of Geriatrics, Salt Lake City, Utah
- Veterans Affairs Medical Center-Salt Lake City, Geriatrics Research Education and Clinical Center, Salt Lake City, Utah
| | - Daniel R. Machin
- University of Utah, Department of Internal Medicine, Division of Geriatrics, Salt Lake City, Utah
- Veterans Affairs Medical Center-Salt Lake City, Geriatrics Research Education and Clinical Center, Salt Lake City, Utah
| | - Lisa A. Lesniewski
- University of Utah, Department of Internal Medicine, Division of Geriatrics, Salt Lake City, Utah
- Veterans Affairs Medical Center-Salt Lake City, Geriatrics Research Education and Clinical Center, Salt Lake City, Utah
| |
Collapse
|
47
|
Myrianthopoulos V, Evangelou K, Vasileiou PVS, Cooks T, Vassilakopoulos TP, Pangalis GA, Kouloukoussa M, Kittas C, Georgakilas AG, Gorgoulis VG. Senescence and senotherapeutics: a new field in cancer therapy. Pharmacol Ther 2018; 193:31-49. [PMID: 30121319 DOI: 10.1016/j.pharmthera.2018.08.006] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Cellular senescence is a stress response mechanism ensuring homeostasis. Its temporal activation during embryonic development or normal adult life is linked with beneficial properties. In contrast, persistent (chronic) senescence seems to exert detrimental effects fostering aging and age-related disorders, such as cancer. Due to the lack of a reliable marker able to detect senescence in vivo, its precise impact in age-related diseases is to a large extent still undetermined. A novel reagent termed GL13 (SenTraGorTM) that we developed, allowing senescence recognition in any type of biological material, emerges as a powerful tool to study the phenomenon of senescence in vivo. Exploiting the advantages of this novel methodological approach, scientists will be able to detect and connect senescence with aggressive behavior in human malignancies, such as tolerance to chemotherapy in classical Hodgkin Lymphoma and Langerhans Cell Histiocytosis. The latter depicts the importance of developing the new and rapidly expanding field of senotherapeutic agents targeting and driving to cell death senescent cells. We discuss in detail the current progress of this exciting area of senotherapeutics and suggest its future perspectives and applications.
Collapse
Affiliation(s)
- Vassilios Myrianthopoulos
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, Athens, Greece; Division of Pharmaceutical Chemistry, School of Pharmacy, National and Kapodistrian University of Athens, Greece; PharmaInformatics Unit, Athena Research Center, Greece
| | - Konstantinos Evangelou
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, Athens, Greece; Department of Anatomy-Histology-Embryology, Medical School, University of Ioannina, Ioannina, Greece
| | - Panagiotis V S Vasileiou
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Tomer Cooks
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Theodoros P Vassilakopoulos
- Department of Haematology and Bone Marrow Transplantation, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Myrsini Kouloukoussa
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, Athens, Greece; Museum of Anthropology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Christos Kittas
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Alexandros G Georgakilas
- DNA Damage Laboratory, Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens (NTUA), Athens, Greece.
| | - Vassilis G Gorgoulis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, Athens, Greece; Center for New Biotechnologies and Precision Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece; Faculty Institute for Cancer Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, UK; Biomedical Research Foundation, Academy of Athens, Athens, Greece.
| |
Collapse
|
48
|
The protective effect of puerarin on angiotensin II-induced aortic aneurysm formation by the inhibition of NADPH oxidase activation and oxidative stress-triggered AP-1 signaling pathways. Oncol Lett 2018; 16:3327-3332. [PMID: 30127931 DOI: 10.3892/ol.2018.9021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 04/05/2018] [Indexed: 01/02/2023] Open
Abstract
Puerarin, an active ingredient of Pueraria lobata, has a range of pharmacological effects and excellent pharmacodynamic properties. In the present study, the effect of puerarin on angiotensin II-induced aortic aneurysm formation and the potential underlying molecular mechanisms were examined. The results revealed that puerarin significantly suppressed the viability, and induced the apoptosis, of aneurysm-inducing cells in a time- and dose-dependent manner. Furthermore, treatment with puerarin significantly suppressed the production of reactive oxygen species (ROS) and the expression of matrix metalloproteinase-2 (MMP-2) protein in aneurysm cells. Puerarin treatment significantly increased caspase-9 and -3 activity, induced the protein expression of phosphorylated (p)-Jun and inhibited the protein expression of activator protein 1 (AP-1) in aneurysm cells. It was also demonstrated that Puerarin significantly suppressed the reduced nicotinamide-adenine dinucleotide phosphate (NADPH) oxidase activity in aneurysm cells. Therefore, it was demonstrated that puerarin on suppressed the cell growth of angiotensin II-induced aortic aneurysm formation by affecting the rate of apoptosis, the generation of ROS, MMP-2, AP-1 and p-Jun protein expression and NADPH oxidase.
Collapse
|
49
|
Katsuumi G, Shimizu I, Yoshida Y, Minamino T. Vascular Senescence in Cardiovascular and Metabolic Diseases. Front Cardiovasc Med 2018; 5:18. [PMID: 29556500 PMCID: PMC5845435 DOI: 10.3389/fcvm.2018.00018] [Citation(s) in RCA: 140] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 02/21/2018] [Indexed: 01/08/2023] Open
Abstract
In mammals, aging is associated with accumulation of senescent cells. Stresses such as telomere shortening and reactive oxygen species induce “cellular senescence”, which is characterized by growth arrest and alteration of the gene expression profile. Chronological aging is associated with development of age-related diseases, including heart failure, diabetes, and atherosclerotic disease, and studies have shown that accumulation of senescent cells has a causative role in the pathology of these age-related disorders. Endothelial cell senescence has been reported to develop in heart failure and promotes pathologic changes in the failing heart. Senescent endothelial cells and vascular smooth muscle cells are found in atherosclerotic plaque, and studies indicate that these cells are involved in progression of plaque. Diabetes is also linked to accumulation of senescent vascular endothelial cells, while endothelial cell senescence per se induces systemic glucose intolerance by inhibiting skeletal muscle metabolism. A close connection between derangement of systemic metabolism and cellular senescence is also well recognized. Aging is a complex phenomenon, and there is no simple approach to understanding the whole process. However, there is accumulating evidence that cellular senescence has a central role in the development and progression of various undesirable aspects of aging. Suppression of cellular senescence or elimination of senescent cells reverses phenotypic changes of aging in several models, and proof-of-concept has been established that inhibiting accumulation of senescent cells could become a next generation therapy for age-related disorders. It is clear that cellular senescence drives various pathological changes associated with aging. Accordingly, further investigation into the role of this biological process in age-related disorders and discovery of senolytic compounds are important fields for future exploration.
Collapse
Affiliation(s)
- Goro Katsuumi
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Ippei Shimizu
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.,Division of Molecular Aging and Cell Biology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Yohko Yoshida
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.,Division of Molecular Aging and Cell Biology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| |
Collapse
|
50
|
Telomere Biology and Thoracic Aortic Aneurysm. Int J Mol Sci 2017; 19:ijms19010003. [PMID: 29267201 PMCID: PMC5795955 DOI: 10.3390/ijms19010003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 12/13/2017] [Accepted: 12/19/2017] [Indexed: 12/27/2022] Open
Abstract
Ascending aortic aneurysms are mostly asymptomatic and present a great risk of aortic dissection or perforation. Consequently, ascending aortic aneurysms are a source of lethality with increased age. Biological aging results in progressive attrition of telomeres, which are the repetitive DNA sequences at the end of chromosomes. These telomeres play an important role in protection of genomic DNA from end-to-end fusions. Telomere maintenance and telomere attrition-associated senescence of endothelial and smooth muscle cells have been indicated to be part of the pathogenesis of degenerative vascular diseases. This systematic review provides an overview of telomeres, telomere-associated proteins and telomerase to the formation and progression of aneurysms of the thoracic ascending aorta. A better understanding of telomere regulation in the vascular pathology might provide new therapeutic approaches. Measurements of telomere length and telomerase activity could be potential prognostic biomarkers for increased risk of death in elderly patients suffering from an aortic aneurysm.
Collapse
|