1
|
Liu Z, Wang Y, Li L, Liu L, Li Y, Li Z, Xie Y, Yu F. SNAI2, a potential crossing point between cancer and cardiovascular disease. FASEB J 2025; 39:e70459. [PMID: 40059450 DOI: 10.1096/fj.202500198r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/18/2025] [Accepted: 03/03/2025] [Indexed: 05/13/2025]
Abstract
Cancer and cardiovascular disease remain the leading causes of morbidity and mortality worldwide, and the two separate disease entities share several similarities and possible interactions. Patients with cancer may have underlying cardiovascular disease, which is often exacerbated by the stress of tumor growth or treatment. At the same time, cardiotoxicity induced by anti-cancer therapies or the malignant process itself can lead to new cardiovascular diseases. Efforts have been made to find a rational explanation for this phenomenon. As a classical tumor-promoting factor, we notice that SNAI2 simultaneously plays an important pathogenic role in cardiovascular diseases. Moreover, there are several striking parallels in the mechanisms of cancer and CVD, such as shared risk factors (e.g., smoking and diabetes), cellular phenotypic switching, and metabolic remodeling, all of which are mediated by SNAI2. This review aims to summarize SNAI2's role in the core mechanisms linking cancer and CVD, as well as explore therapeutic approaches targeting SNAI2 and also seeks to provide insights into the common mechanisms underlying both cancer and CVD.
Collapse
Affiliation(s)
- Zihao Liu
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yingzi Wang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Lei Li
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Linlu Liu
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yuhao Li
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Zhixin Li
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yucheng Xie
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Fengxu Yu
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Cardiovascular Remodeling and Dysfunction Key Laboratory of Luzhou, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| |
Collapse
|
2
|
Khan H, Abu-Raisi M, Feasson M, Shaikh F, Saposnik G, Mamdani M, Qadura M. Current Prognostic Biomarkers for Abdominal Aortic Aneurysm: A Comprehensive Scoping Review of the Literature. Biomolecules 2024; 14:661. [PMID: 38927064 PMCID: PMC11201473 DOI: 10.3390/biom14060661] [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: 05/02/2024] [Revised: 05/29/2024] [Accepted: 06/03/2024] [Indexed: 06/28/2024] Open
Abstract
Abdominal aortic aneurysm (AAA) is a progressive dilatation of the aorta that can lead to aortic rupture. The pathophysiology of the disease is not well characterized but is known to be caused by the general breakdown of the extracellular matrix within the aortic wall. In this comprehensive literature review, all current research on proteins that have been investigated for their potential prognostic capabilities in patients with AAA was included. A total of 45 proteins were found to be potential prognostic biomarkers for AAA, predicting incidence of AAA, AAA rupture, AAA growth, endoleak, and post-surgical mortality. The 45 proteins fell into the following seven general categories based on their primary function: (1) cardiovascular health, (2) hemostasis, (3) transport proteins, (4) inflammation and immunity, (5) kidney function, (6) cellular structure, (7) and hormones and growth factors. This is the most up-to-date literature review on current prognostic markers for AAA and their functions. This review outlines the wide pathophysiological processes that are implicated in AAA disease progression.
Collapse
Affiliation(s)
- Hamzah Khan
- Division of Vascular Surgery, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada
- Li Ka Shing Knowledge Institute, St Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1W8, Canada
| | - Mohamed Abu-Raisi
- Division of Vascular Surgery, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada
- Li Ka Shing Knowledge Institute, St Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1W8, Canada
| | - Manon Feasson
- Li Ka Shing Knowledge Institute, St Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1W8, Canada
| | - Farah Shaikh
- Division of Vascular Surgery, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada
- Li Ka Shing Knowledge Institute, St Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1W8, Canada
| | - Gustavo Saposnik
- Li Ka Shing Knowledge Institute, St Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1W8, Canada
- Division of Neurology, Department of Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Muhammad Mamdani
- Li Ka Shing Knowledge Institute, St Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1W8, Canada
| | - Mohammad Qadura
- Division of Vascular Surgery, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada
- Li Ka Shing Knowledge Institute, St Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1W8, Canada
- Department of Surgery, University of Toronto, Toronto, ON M5T 1P5, Canada
| |
Collapse
|
3
|
Kugo H, Moriyama T, Zaima N. Nicotine induces vasa vasorum stenosis in the aortic wall. Biotech Histochem 2024; 99:197-203. [PMID: 38780082 DOI: 10.1080/10520295.2024.2352724] [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] [Indexed: 05/25/2024] Open
Abstract
Abdominal aortic aneurysm (AAA) is a vascular disease that involves aortic wall dilation. Cigarette smoking is an established risk factor and rupture, and nicotine may be a major contributor to the onset of AAA. In humans the condition is associated with stenosis of the vasa vasorum (VV), which may be caused by nicotine. In this study, we evaluated the effects of nicotine on VV pathology. After 4 weeks of nicotine administration to rats using an osmotic pump, the VV patency rate in the nicotine administration group was significantly lower than that in the control group. The levels of Ki-67, a cell proliferation marker, were significantly increased in the regions containing VV in the nicotine group, as were hypoxia inducible factor-α levels. Collagen levels around VV were significantly lower in the nicotine group than in the controls. Our data suggest that nicotine can cause VV stenosis by inducing abnormal proliferation of smooth muscle cells in the VV. The increased risk of AAA development due to cigarette smoking may be partially explained by nicotine-induced VV denaturation and collagen fiber degradation.
Collapse
Affiliation(s)
- Hirona Kugo
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Kindai University, Nara City, Japan
| | - Tatsuya Moriyama
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Kindai University, Nara City, Japan
- Agricultural Technology and Innovation Research Institute, Kindai University, Nara City, Japan
| | - Nobuhiro Zaima
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Kindai University, Nara City, Japan
- Agricultural Technology and Innovation Research Institute, Kindai University, Nara City, Japan
| |
Collapse
|
4
|
Hadzikadunic H, Sjælland TB, Lindholt JS, Steffensen LB, Beck HC, Kavaliunaite E, Rasmussen LM, Stubbe J. Nicotine Administration Augments Abdominal Aortic Aneurysm Progression in Rats. Biomedicines 2023; 11:biomedicines11051417. [PMID: 37239088 DOI: 10.3390/biomedicines11051417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/30/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
Inflammation and elastin degradation are key hallmarks in the pathogenesis of abdominal aortic aneurysms (AAAs). It has been acknowledged that activation of alpha7 nicotinic acetylcholine receptors (α7nAChRs) attenuates inflammation, termed the cholinergic anti-inflammatory pathway (CAP). Thus, we hypothesize that low-dose nicotine impairs the progression of elastase-induced AAAs in rats by exerting anti-inflammatory and anti-oxidative stress properties. Male Sprague-Dawley rats underwent surgical AAA induction with intraluminal elastase infusion. We compared vehicle rats with rats treated with nicotine (1.25 mg/kg/day), and aneurysm progression was monitored by weekly ultrasound images for 28 days. Nicotine treatment significantly promoted AAA progression (p = 0.031). Additionally, gelatin zymography demonstrated that nicotine significantly reduced pro-matrix metalloproteinase (pro-MMP) 2 (p = 0.029) and MMP9 (p = 0.030) activity in aneurysmal tissue. No significant difference was found in the elastin content or the score of elastin degradation between the groups. Neither infiltrating neutrophils nor macrophages, nor aneurysmal messenger RNA (mRNA) levels of pro- or anti-inflammatory cytokines, differed between the vehicle and nicotine groups. Finally, no difference in mRNA levels of markers for anti-oxidative stress or the vascular smooth muscle cells' contractile phenotype was observed. However, proteomics analyses of non-aneurysmal abdominal aortas revealed that nicotine decreased myristoylated alanine-rich C-kinase substrate and proteins, in ontology terms, inflammatory response and reactive oxygen species, and in contradiction to augmented AAAs. In conclusion, nicotine at a dose of 1.25 mg/kg/day augments AAA expansion in this elastase AAA model. These results do not support the use of low-dose nicotine administration for the prevention of AAA progression.
Collapse
Affiliation(s)
- Hana Hadzikadunic
- Elitary Research Centre of Individualized Treatment for Arterial Disease (CIMA), Odense University Hospital, University of Southern Denmark, 5000 Odense, Denmark
- Cardiovascular and Renal Research Unit, Institute for Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark
| | - Tea Bøvling Sjælland
- Elitary Research Centre of Individualized Treatment for Arterial Disease (CIMA), Odense University Hospital, University of Southern Denmark, 5000 Odense, Denmark
- Cardiovascular and Renal Research Unit, Institute for Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark
| | - Jes S Lindholt
- Elitary Research Centre of Individualized Treatment for Arterial Disease (CIMA), Odense University Hospital, University of Southern Denmark, 5000 Odense, Denmark
- Department of Cardiothoracic and Vascular Surgery, Odense University Hospital, 5000 Odense, Denmark
| | - Lasse Bach Steffensen
- Elitary Research Centre of Individualized Treatment for Arterial Disease (CIMA), Odense University Hospital, University of Southern Denmark, 5000 Odense, Denmark
- Cardiovascular and Renal Research Unit, Institute for Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark
| | - Hans Christian Beck
- Elitary Research Centre of Individualized Treatment for Arterial Disease (CIMA), Odense University Hospital, University of Southern Denmark, 5000 Odense, Denmark
- Department of Clinical Biochemistry, Odense University Hospital, 5000 Odense, Denmark
| | - Egle Kavaliunaite
- Elitary Research Centre of Individualized Treatment for Arterial Disease (CIMA), Odense University Hospital, University of Southern Denmark, 5000 Odense, Denmark
- Cardiovascular and Renal Research Unit, Institute for Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark
- Department of Cardiothoracic and Vascular Surgery, Odense University Hospital, 5000 Odense, Denmark
| | - Lars Melholt Rasmussen
- Elitary Research Centre of Individualized Treatment for Arterial Disease (CIMA), Odense University Hospital, University of Southern Denmark, 5000 Odense, Denmark
- Department of Clinical Biochemistry, Odense University Hospital, 5000 Odense, Denmark
| | - Jane Stubbe
- Elitary Research Centre of Individualized Treatment for Arterial Disease (CIMA), Odense University Hospital, University of Southern Denmark, 5000 Odense, Denmark
- Cardiovascular and Renal Research Unit, Institute for Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark
| |
Collapse
|
5
|
Osipov AV, Averin AS, Shaykhutdinova ER, Dyachenko IA, Tsetlin VI, Utkin YN. Muscarinic and Nicotinic Acetylcholine Receptors in the Regulation of the Cardiovascular System. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2023; 49:1-18. [DOI: 10.1134/s1068162023010211] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/16/2022] [Accepted: 08/23/2022] [Indexed: 05/14/2025]
|
6
|
Kishi C, Higashihara M, Takemoto Y, Kamei M, Yoshioka Y, Matsumura S, Yamada K, Kobayashi T, Matahira Y, Moriyama T, Zaima N. Inhaled volatile β-caryophyllene is incorporated into the aortic wall and attenuates nicotine-induced aorta degeneration via a CB2 receptor-dependent pathway. Biomed Pharmacother 2022; 153:113423. [DOI: 10.1016/j.biopha.2022.113423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/13/2022] [Accepted: 07/14/2022] [Indexed: 11/24/2022] Open
|
7
|
Nicotine Exacerbates TAAD Formation Induced by Smooth Muscle-Specific Deletion of the TGF- β Receptor 2. J Immunol Res 2021; 2021:6880036. [PMID: 34646889 PMCID: PMC8505064 DOI: 10.1155/2021/6880036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 09/04/2021] [Indexed: 01/22/2023] Open
Abstract
Tobacco smoke is an established risk factor for thoracic aortic aneurysms and dissections (TAAD). However, little is known about its underlying mechanisms due to the lack of validated animal models. The present study developed a mouse model that may be utilized to investigate exacerbation of TAAD formation by mimetics of tobacco smoke. TAADs were created via inducible deletion of smooth muscle cell-specific Tgfbr2 receptors. Using this model, the first set of experiments evaluated the efficacy of nicotine salt (34.0 mg/kg/day), nicotine free base (NFB, 5.0 mg 90-day pellets), and cigarette smoke extract (0.1 ml/mouse/day). Compared with their respective control groups, only NFB pellets promoted TAAD dilation (23 ± 3% vs. 12 ± 2%, P = 0.014), and this efficacy was achieved at a cost of >50% acute mortality. Infusion of NFB with osmotic minipumps at extremely high, but nonlethal, doses (15.0 or 45.0 mg/kg/day) failed to accelerate TAAD dilation. Interestingly, costimulation with β-aminopropionitrile (BAPN) promoted TAAD dilation and aortic rupture at dosages of 3.0 and 45.0 mg/kg/day, respectively, indicating that BAPN sensitizes the response of TAADs to NFB. In subsequent analyses, the detrimental effects of NFB were associated with clustering of macrophages, neutrophils, and T-cells in areas with structural destruction, enhanced matrix metalloproteinase- (MMP-) 2 production, and pathological angiogenesis with attenuated fibrosis in the adventitia. In conclusion, modeling nicotine exacerbation of TAAD formation requires optimization of chemical form, route of delivery, and dosage of the drug as well as the pathologic complexity of TAADs. Under the optimized conditions of the present study, chronic inflammation and adventitial mal-remodeling serve as critical pathways through which NFB exacerbates TAAD formation.
Collapse
|
8
|
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
|
9
|
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: 31] [Impact Index Per Article: 7.8] [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
|
10
|
Boyd AJ. Intraluminal thrombus: Innocent bystander or factor in abdominal aortic aneurysm pathogenesis? JVS Vasc Sci 2021; 2:159-169. [PMID: 34617066 PMCID: PMC8489244 DOI: 10.1016/j.jvssci.2021.02.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 02/20/2021] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Abdominal aortic aneurysms (AAAs) represent a complex multifactorial hemodynamic, thrombotic, and inflammatory process that can ultimately result in aortic rupture and death. Despite improved screening and surgical management of AAAs, the mortality rates have remained high after rupture, and little progress has occurred in the development of nonoperative treatments. Intraluminal thrombus (ILT) is present in most AAAs and might be involved in AAA pathogenesis. The present review examined the latest clinical and experimental evidence for possible involvement of the ILT in AAA growth and rupture. METHODS A literature review was performed after a search of the PubMed database from 2012 to June 2020 using the terms "abdominal aortic aneurysm" and "intraluminal thrombus." RESULTS The structure, composition, and hemodynamics of ILT formation and propagation were reviewed in relation to the hemostatic and proteolytic factors favoring ILT deposition. The potential effects of the ILT on AAA wall degeneration and rupture, including a review of the current controversies regarding the position, thickness, and composition of ILT, are presented. Although initially potentially protective against increased wall stress, increasing evidence has shown that an increased volume and greater age of the ILT have direct detrimental effects on aortic wall integrity, which might predispose to an increased rupture risk. CONCLUSIONS ILT does not appear to be an innocent bystander in AAA pathophysiology. However, its exact role remains elusive and controversial. Despite computational evidence of a possible protective role of the ILT in reducing wall stress, increasing evidence has shown that the ILT promotes AAA wall degeneration in humans and in animal models. Further research, with large animal models and with more chronic ILT is crucial for a better understanding of the role of the ILT in AAAs and for the potential development of targeted therapies to slow or halt AAA progression.
Collapse
Affiliation(s)
- April J. Boyd
- Department of Vascular Surgery, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
11
|
Whitehead AK, Erwin AP, Yue X. Nicotine and vascular dysfunction. Acta Physiol (Oxf) 2021; 231:e13631. [PMID: 33595878 DOI: 10.1111/apha.13631] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 01/25/2021] [Accepted: 02/15/2021] [Indexed: 12/20/2022]
Abstract
Cigarette smoking is the single most important risk factor for the development of cardiovascular diseases (CVDs). However, the role of nicotine, the addictive component of all tobacco products, in the development of CVD is incompletely understood. Although increased public awareness of the harms of cigarette smoking has successfully led to a decline in its prevalence, the use of electronic cigarettes (e-cig) or electronic nicotine delivery system has increased dramatically in recent years because of the perception that these products are safe. This review summarizes our current knowledge of the expression and function of the nicotinic acetylcholine receptors in the cardiovascular system and the impact of nicotine exposure on cardiovascular health, with a focus on nicotine-induced vascular dysfunction. Nicotine alters vasoreactivity through endothelium-dependent and/or endothelium-independent mechanisms, leading to clinical manifestations in both cigarette smokers and e-cig users. In addition, nicotine induces vascular remodelling through its effects on proliferation, migration and matrix production of both vascular endothelial and vascular smooth muscle cells. The purpose of this review is to identify critical knowledge gaps regarding the effects of nicotine on the vasculature and to stimulate continued nicotine research.
Collapse
Affiliation(s)
- Anna K. Whitehead
- Department of Physiology Louisiana State University Health Sciences Center New Orleans LA USA
| | - Abigail P. Erwin
- Department of Physiology Louisiana State University Health Sciences Center New Orleans LA USA
| | - Xinping Yue
- Department of Physiology Louisiana State University Health Sciences Center New Orleans LA USA
| |
Collapse
|
12
|
Xie S, Ma L, Guan H, Guan S, Wen L, Han C. Daphnetin suppresses experimental abdominal aortic aneurysms in mice via inhibition of aortic mural inflammation. Exp Ther Med 2020; 20:221. [PMID: 33193836 PMCID: PMC7646695 DOI: 10.3892/etm.2020.9351] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 07/21/2020] [Indexed: 12/21/2022] Open
Abstract
Rupture of abdominal aortic aneurysm (AAA) is a devastating event that can be prevented by inhibiting the growth of small aneurysms. Therapeutic strategies targeting certain events that promote the development of AAA must be developed, in order to alter the course of AAA. Chronic inflammation of the aortic mural is a major characteristic of AAA and is related to AAA formation, development and rupture. Daphnetin (DAP) is a coumarin derivative with anti-inflammatory properties that is extracted from Daphne odora var. However, the effect of DAP on AAA development remains unclear. The present study investigated the effect of DAP on the formation and development of experimental AAAs and its potential underlying mechanisms. A mice AAA model was established by intra-aortic infusion of porcine pancreatic elastase (PPE), and mice were intraperitoneally injected with DAP immediately after PPE infusion. The maximum diameter of the abdominal aorta was measured by ultrasound system, and aortic mural changes were investigated by Elastica van Gieson (EVG) staining and immunohistochemical staining. The results demonstrated that DAP significantly suppressed PPE-induced AAA formation and attenuated the depletion of aortic medial elastin and smooth muscle cells in the media of the aorta. Furthermore, the density of mural macrophages, T cells and B cells were significantly attenuated in DAP-treated AAA mice. In addition, treatment with DAP resulted in a significant reduction in mural neovessels. These findings indicated that DAP may limit the formation and progression of experimental aneurysms by inhibiting mural inflammation and angiogenesis. These data confirmed the translational potential of DAP inclinical AAA inhibition strategies.
Collapse
Affiliation(s)
- Shiyun Xie
- Department of Vascular Surgery, Shandong Shanxian Central Hospital, Shanxian, Shandong 274300, P.R. China
| | - Li Ma
- Department of Vascular Surgery, Shandong Shanxian Central Hospital, Shanxian, Shandong 274300, P.R. China
| | - Hongliang Guan
- Department of Vascular Surgery, Shandong Shanxian Central Hospital, Shanxian, Shandong 274300, P.R. China
| | - Su Guan
- Department of Vascular Surgery, Shandong Shanxian Central Hospital, Shanxian, Shandong 274300, P.R. China
| | - Lijuan Wen
- Department of Vascular Surgery, Shandong Shanxian Central Hospital, Shanxian, Shandong 274300, P.R. China
| | - Chanchan Han
- Department of Ultrasound, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| |
Collapse
|
13
|
Xiao J, Liu J, Lio I, Yang C, Chen X, Zhang H, Wang S, Wei Z. All-trans retinoic acid attenuates the progression of Ang II-induced abdominal aortic aneurysms in ApoE -/-mice. J Cardiothorac Surg 2020; 15:160. [PMID: 32615991 PMCID: PMC7331218 DOI: 10.1186/s13019-020-01208-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 06/23/2020] [Indexed: 12/20/2022] Open
Abstract
Background To determine whether all-trans retinoic acid (ATRA) can influence the development of Angiotensin II (Ang II) induced experimental abdominal aortic aneurysms (AAAs). Methods Apolipoprotein E knock-out (ApoE−/−) mice were randomly assigned to 4 groups. Mice in the AAA and ATRA groups underwent continuous subcutaneous Ang II infusion for 28 days to induce AAA, while the Sham and Control groups were infused with saline. Systolic blood pressure was measured by the tail-cuff technique. The Control and ATRA groups received ATRA treatment. Aortic tissue samples were obtained at 28 days after surgery and evaluated by aortic diameter measurement, Western blotting, immunohistochemistry, and hematoxylin-eosin (H&E) and Verhoeff-Van Gieson (EVG) staining. Results The abdominal aortic diameter was significantly reduced in the ATRA group compared with the AAA group (3 of 12 (25%) vs 9 of 12 (75%), P < 0.05), and the ATRA group exhibited reduced blood pressure on days 7, 14, and 28. Low expression of angiotensin II receptor type 1 (AT1), matrix metalloproteinase 2 (MMP2), and matrix metalloproteinase 9 (MMP9) and EVG staining revealed a significant reduction in the disruption of elastic fibers in the abdominal aortic tissue of the ATRA group compared to the AAA group. Western blot analysis indicated that protein levels of retinoic acid receptor α (RARα), MMP2, MMP9, and AT1 were dramatically affected by ATRA treatment. Conclusions In conclusion, ATRA attenuates the progression of Ang II-induced AAAs, possibly by downregulating MMP2, MMP9, and AT-1 expression.
Collapse
Affiliation(s)
- Jie Xiao
- Department of Cardiovascular Surgery, Central Hospital of Wuhan, Huazhong University of Science and Technology, Wuhan, 430014, China
| | - Jinping Liu
- Department of Cardiovascular Surgery, Zhongnan Hospital, Wuhan University, Wuhan, 430071, Hubei, China
| | - Iohang Lio
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chuanlei Yang
- Department of Cardiovascular Surgery, Central Hospital of Wuhan, Huazhong University of Science and Technology, Wuhan, 430014, China
| | - Xing Chen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hua Zhang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Shuxia Wang
- Department of Radiology, Central Hospital of Wuhan, Huazhong University of Science and Technology, Wuhan, 430014, China.
| | - Zhanjie Wei
- Department of General Surgery, Central Hospital of Wuhan, Huazhong University of Science and Technology, Wuhan, 430014, China.
| |
Collapse
|
14
|
Xiao J, Wei Z, Chen X, Chen W, Zhang H, Yang C, Shang Y, Liu J. Experimental abdominal aortic aneurysm growth is inhibited by blocking the JAK2/STAT3 pathway. Int J Cardiol 2020; 312:100-106. [PMID: 32334849 DOI: 10.1016/j.ijcard.2020.03.072] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/05/2020] [Accepted: 03/27/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND The JAK/STAT pathway is a vital transcription signaling pathway that regulates gene expression and cellular activity. Our recently published study highlighted the role of IL-17A in abdominal aortic aneurysm (AAA) formation and rupture. IL-17A has been proven to upregulate vascular endothelial growth factor (VEGF) expression in some diseases. However, no study has demonstrated the relationships among JAK2/STAT3, IL-17A and VEGF. Therefore, we hypothesized that IL-17A may up-regulate VEGF expression via the JAK2/STAT3 signaling pathway to amplify the inflammatory response, exacerbate neovascularization, and accelerate AAA progression. METHODS To fully verify our hypothesis, two separate studies were performed: i) a study investigating the influence of JAK2/STAT3 on AAA formation and progression. ii) a study evaluating the relationship among IL-17A, JAK2/STAT3 and VEGF. Human tissues were collected from 7 AAA patients who underwent open surgery and 7 liver transplantation donors. All human aortic tissues were examined by histological and immunohistochemical staining, and Western blotting. Furthermore, mouse aortic tissues were also examined by histological and immunohistochemical staining and Western blotting, and the mouse aortic diameters were assessed by high-resolution Vevo 2100 microimaging system. RESULTS Among human aortic tissues, JAK2/STAT3, IL-17A and VEGF expression levels were higher in AAA tissues than in control tissues. Group treated with WP1066 (a selective JAK2/STAT3 pathway inhibitor), IL-17A, and VEGF groups had AAA incidences of 25%, 40%, and 65%, respectively, while the control group had an incidence of 75%. Histopathological analysis revealed that the IL-17A- and VEGF-related inflammatory responses were attenuated by WP1066. Thus, blocking the JAK2/STAT3 pathway with WP1066 attenuated experimental AAA progression. In addition, in study ii, we found that IL-17A siRNA seemed to attenuate the expression of IL-17A and VEGF in vivo study; treatment with VEGF siRNA decreased the expression of VEGF, while IL-17A expression remained high. In an in vitro study, rhIL-17A treatment increased JAK2/STAT3 and VEGF expression in macrophages in a dose-dependent manner. CONCLUSION Blocking the JAK2/STAT3 pathway with WP1066 (a JAK2/STAT3 specific inhibitor) attenuates experimental AAA progression. During AAA progression, IL-17A may influence the expression of VEGF via the JAK2/STAT3 signaling pathway. This potential mechanism may suggest a novel strategy for nonsurgical AAA treatment.
Collapse
Affiliation(s)
- Jie Xiao
- Department of Cardiovascular Surgery, Central Hospital of Wuhan, Tongji Medical college, Huazhong University of Science and Technology, Wuhan, 430014, Hubei, China
| | - Zhanjie Wei
- Department of Thyroid and Breast Surgery, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, Hubei, China
| | - Xing Chen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical college, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Weiqiang Chen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical college, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Hua Zhang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical college, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Chuanlei Yang
- Department of Cardiovascular Surgery, Central Hospital of Wuhan, Tongji Medical college, Huazhong University of Science and Technology, Wuhan, 430014, Hubei, China
| | - Yuqiang Shang
- Department of Cardiovascular Surgery, Central Hospital of Wuhan, Tongji Medical college, Huazhong University of Science and Technology, Wuhan, 430014, Hubei, China
| | - Jinping Liu
- Department of Cardiovascular Surgery, Zhongnan Hospital, Wuhan University, Wuhan, 430071, Hubei, China.
| |
Collapse
|
15
|
Wang Z, Guo J, Han X, Xue M, Wang W, Mi L, Sheng Y, Ma C, Wu J, Wu X. Metformin represses the pathophysiology of AAA by suppressing the activation of PI3K/AKT/mTOR/autophagy pathway in ApoE -/- mice. Cell Biosci 2019; 9:68. [PMID: 31467666 PMCID: PMC6712653 DOI: 10.1186/s13578-019-0332-9] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 08/16/2019] [Indexed: 02/07/2023] Open
Abstract
Background The protective effect of metformin (MET) on abdominal aortic aneurysm (AAA) has been reported. However, the related mechanism is still poor understood. In this study, we deeply investigated the role of metformin in AAA pathophysiology. Methods Angiotensin II (Ang-II) was used to construct the AAA model in ApoE−/− mice. The related mechanism was explored using Western blot and quantitative real time PCR (qRT-PCR). We also observed the morphological changes in the abdominal aorta and the influence of metformin on biological behaviors of rat abdominal aortic VSMCs. Results The PI3K/AKT/mTOR pathway was activated in aneurysmal wall tissues of AAA patients and rat model. Treatment with metformin inhibited the breakage and preserved the elastin structure of the aorta, the loss of collagen, and the apoptosis of aortic cells. In addition, metformin significantly suppressed the activation of the PI3K/AKT/mToR pathway and decreased the mRNA and protein levels of LC3B and Beclin1, which were induced by Ang-II. Moreover, PI3K inhibitors enhanced the effect of metformin while PI3K agonists largely reversed this effect. Interestingly, the cell proliferation, apoptosis, migration and autophagy of vascular smooth muscle cells (VSMCs) induced by Ang-II were also decreased following metformin treatment. PI3K inhibitors and agonists strengthened and weakened the effects of metformin in VSMCs, respectively. Conclusions Metformin represses the pathophysiology of AAA by inhibiting the activation of PI3K/AKT/mTOR/autophagy pathway. This repression may be useful as a new therapeutic strategy for AAA.
Collapse
Affiliation(s)
- Zhu Wang
- 1Department of Vascular Surgery, Shandong Provincial Hospital Affiliated to Shandong University, 324 Jing Wu Wei Qi Road, Jinan, 250021 Shandong China.,2Department of Interventional Medicine and Vascular Surgery, Binzhou Medical University Hospital, Binzhou, 256603 Shandong China
| | - Jingjing Guo
- 3Department of Obstetrics and Gynecology, Binzhou Medical University Hospital, Binzhou, 256603 Shandong China
| | - Xinqiang Han
- 2Department of Interventional Medicine and Vascular Surgery, Binzhou Medical University Hospital, Binzhou, 256603 Shandong China
| | - Ming Xue
- 4Department of Interventional Radiology, Weihai Municipal Hospital, Weihai, 264200 Shandong China
| | - Wenming Wang
- 2Department of Interventional Medicine and Vascular Surgery, Binzhou Medical University Hospital, Binzhou, 256603 Shandong China
| | - Lei Mi
- Department of General Surgery, Taian City Central Hospital, Taian, 271000 Shandong China
| | - Yuguo Sheng
- 2Department of Interventional Medicine and Vascular Surgery, Binzhou Medical University Hospital, Binzhou, 256603 Shandong China
| | - Chao Ma
- 2Department of Interventional Medicine and Vascular Surgery, Binzhou Medical University Hospital, Binzhou, 256603 Shandong China
| | - Jian Wu
- 2Department of Interventional Medicine and Vascular Surgery, Binzhou Medical University Hospital, Binzhou, 256603 Shandong China
| | - Xuejun Wu
- 1Department of Vascular Surgery, Shandong Provincial Hospital Affiliated to Shandong University, 324 Jing Wu Wei Qi Road, Jinan, 250021 Shandong China
| |
Collapse
|
16
|
Bals R, Boyd J, Esposito S, Foronjy R, Hiemstra PS, Jiménez-Ruiz CA, Katsaounou P, Lindberg A, Metz C, Schober W, Spira A, Blasi F. Electronic cigarettes: a task force report from the European Respiratory Society. Eur Respir J 2019; 53:13993003.01151-2018. [PMID: 30464018 DOI: 10.1183/13993003.01151-2018] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 08/09/2018] [Indexed: 01/10/2023]
Abstract
There is a marked increase in the development and use of electronic nicotine delivery systems or electronic cigarettes (ECIGs). This statement covers electronic cigarettes (ECIGs), defined as "electrical devices that generate an aerosol from a liquid" and thus excludes devices that contain tobacco. Database searches identified published articles that were used to summarise the current knowledge on the epidemiology of ECIG use; their ingredients and accompanied health effects; second-hand exposure; use of ECIGs for smoking cessation; behavioural aspects of ECIGs and social impact; in vitro and animal studies; and user perspectives.ECIG aerosol contains potentially toxic chemicals. As compared to conventional cigarettes, these are fewer and generally in lower concentrations. Second-hand exposures to ECIG chemicals may represent a potential risk, especially to vulnerable populations. There is not enough scientific evidence to support ECIGs as an aid to smoking cessation due to a lack of controlled trials, including those that compare ECIGs with licenced stop-smoking treatments. So far, there are conflicting data that use of ECIGs results in a renormalisation of smoking behaviour or for the gateway hypothesis. Experiments in cell cultures and animal studies show that ECIGs can have multiple negative effects. The long-term effects of ECIG use are unknown, and there is therefore no evidence that ECIGs are safer than tobacco in the long term. Based on current knowledge, negative health effects cannot be ruled out.
Collapse
Affiliation(s)
- Robert Bals
- Dept of Internal Medicine V - Pulmonology, Allergology and Critical Care Medicine, Saarland University, Homburg, Germany
| | | | - Susanna Esposito
- Pediatric Clinic, Dept of Surgical and Biomedical Sciences, Università degli Studi di Perugia, Perugia, Italy
| | - Robert Foronjy
- Pulmonary and Critical Care Medicine, SUNY Downstate Medical Center, New York, NY, USA
| | - Pieter S Hiemstra
- Dept of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Paraskevi Katsaounou
- 1st ICU Evangelismos Hospital, National Kapodistrian University of Athens, Athens, Greece
| | - Anne Lindberg
- Dept of Public Health and Clinical Medicine, Division of Medicine, Umeå University, Umeå, Sweden
| | - Carlos Metz
- Dept of Internal Medicine V - Pulmonology, Allergology and Critical Care Medicine, Saarland University, Homburg, Germany
| | - Wolfgang Schober
- Bavarian Health and Food Safety Authority, Dept of Chemical Safety and Toxicology, Munich, Germany
| | - Avrum Spira
- Boston University School of Medicine, Boston, MA, USA
| | - Francesco Blasi
- Dept of Pathophysiology and Transplantation, Università degli Studi di Milano, Internal Medicine Department, Respiratory Unit and Regional Adult Cystic Fibrosis Center, IRCCS Fondazione Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
17
|
Kugo H, Miyamoto C, Sawaragi A, Hoshino K, Hamatani Y, Matsumura S, Yoshioka Y, Moriyama T, Zaima N. Sesame Extract Attenuates the Degradation of Collagen and Elastin Fibers in the Vascular Walls of Nicotine-administered Mice. J Oleo Sci 2019; 68:79-85. [DOI: 10.5650/jos.ess18200] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Hirona Kugo
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Kindai University
| | - Chie Miyamoto
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Kindai University
| | - Ayaka Sawaragi
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Kindai University
| | - Kiyoto Hoshino
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Kindai University
| | - Yuka Hamatani
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Kindai University
| | | | | | - Tatsuya Moriyama
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Kindai University
- Agricultural Technology and Innovation Research Institute, Kindai University
| | - Nobuhiro Zaima
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Kindai University
- Agricultural Technology and Innovation Research Institute, Kindai University
| |
Collapse
|
18
|
Hou X, Yang S, Zheng Y. Licochalcone A attenuates abdominal aortic aneurysm induced by angiotensin II via regulating the miR-181b/SIRT1/HO-1 signaling. J Cell Physiol 2018; 234:7560-7568. [PMID: 30417353 DOI: 10.1002/jcp.27517] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 09/10/2018] [Indexed: 01/15/2023]
Abstract
Licochalcone A (LA), a chalcone derived from liquorice, exhibits multiple biological activities, including anti-oxidation and anti-inflammation. This study aimed to investigate the role and underlying mechanism of LA in the abdominal aortic aneurysm (AAA). AAA model was established by continuous infusion of 1000 ng/kg/min of angiotensin II (AngII) in ApoE -/- mice for 4 weeks. At 7 days before AngII administration, 5 mg/kg/day or 10 mg/kg/day of LA was intraperitoneally administered to mice and continued for 4 weeks. The characteristics and quantification of AAAs were determined in situ. Real-time PCR or western blot was used to measure mRNA or protein levels of matrix metalloproteinase 2 and matrix metalloproteinase 9; pro-inflammatory cytokines tumor necrosis factor-α, interleukin-1β, and interleukin-6; apoptosis-related proteins Bax, Bcl-2, and active caspase-3; miR-181b; Sirtuin 1 (SIRT1); and heme oxygenase-1 (HO-1). Mouse-aorta-origin vascular smooth muscle (MOVAS) cells were used to confirm the involved pathways in vitro. We found LA administration dose-dependently reduced the incidence of AngII-induced AAA, aneurysm diameter enlargement, elastin degradation, matrix metalloproteinase production, pro-inflammatory cytokines and miR-181b expression, and vascular smooth muscle cell apoptosis. It elevated SIRT1 and HO-1 expression that was suppressed by AngII. AngII enhanced miR-181b but reduced SIRT1 and HO-1 expression in MOVAS cells. In AngII-stimulated MOVAS cells, downregulation of miR-181b significantly upregulated the expression of SIRT1 and HO-1, the effect of which was abrogated by SIRT1 siRNA. Collectively, LA could attenuate AngII-induced AAA by modulating the miR-181b/SIRT1/HO-1 signaling. LA might be a potential medical therapy for small AAA.
Collapse
Affiliation(s)
- Xuhui Hou
- Department of Vascular Surgery, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Songbai Yang
- Department of Vascular Surgery, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Yan Zheng
- Department of Anesthesiology, China-Japan Union Hospital, Jilin University, Changchun, China
| |
Collapse
|
19
|
Oakes JM, Fuchs RM, Gardner JD, Lazartigues E, Yue X. Nicotine and the renin-angiotensin system. Am J Physiol Regul Integr Comp Physiol 2018; 315:R895-R906. [PMID: 30088946 DOI: 10.1152/ajpregu.00099.2018] [Citation(s) in RCA: 205] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cigarette smoking is the single most important risk factor for the development of cardiovascular and pulmonary diseases (CVPD). Although cigarette smoking has been in constant decline since the 1950s, the introduction of e-cigarettes or electronic nicotine delivery systems 10 yr ago has attracted former smokers as well as a new generation of consumers. Nicotine is a highly addictive substance, and it is currently unclear whether e-cigarettes are "safer" than regular cigarettes or whether they have the potential to reverse the health benefits, notably on the cardiopulmonary system, acquired with the decline of tobacco smoking. Of great concern, nicotine inhalation devices are becoming popular among young adults and youths, emphasizing the need for awareness and further study of the potential cardiopulmonary risks of nicotine and associated products. This review focuses on the interaction between nicotine and the renin-angiotensin system (RAS), one of the most important regulatory systems on autonomic, cardiovascular, and pulmonary functions in both health and disease. The literature presented in this review strongly suggests that nicotine alters the homeostasis of the RAS by upregulating the detrimental angiotensin-converting enzyme (ACE)/angiotensin (ANG)-II/ANG II type 1 receptor axis and downregulating the compensatory ACE2/ANG-(1-7)/Mas receptor axis, contributing to the development of CVPD.
Collapse
Affiliation(s)
- Joshua M Oakes
- Department of Physiology, Louisiana State University Health Sciences Center , New Orleans, Louisiana
| | - Robert M Fuchs
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center , New Orleans, Louisiana
| | - Jason D Gardner
- Department of Physiology, Louisiana State University Health Sciences Center , New Orleans, Louisiana
| | - Eric Lazartigues
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center , New Orleans, Louisiana
| | - Xinping Yue
- Department of Physiology, Louisiana State University Health Sciences Center , New Orleans, Louisiana
| |
Collapse
|
20
|
Tang W, Yao L, Hoogeveen RC, Alonso A, Couper DJ, Lutsey PL, Steenson CC, Guan W, Hunter DW, Lederle FA, Folsom AR. The Association of Biomarkers of Inflammation and Extracellular Matrix Degradation With the Risk of Abdominal Aortic Aneurysm: The ARIC Study. Angiology 2018; 70:130-140. [PMID: 29945457 DOI: 10.1177/0003319718785278] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Animal and human laboratory studies suggest that the pathogenesis of abdominal aortic aneurysms (AAAs) involves inflammation and degradation and remodeling of the extracellular matrix. This study prospectively assessed the association between biomarkers for these mechanisms and the presence of AAA during 24 years of follow-up in the Atherosclerosis Risk in Communities (ARIC) study. The ARIC prospectively identified clinically diagnosed AAAs in 15 792 men and women from baseline in 1987 to 1989 to 2011 using hospital discharge codes and death records. Additional asymptomatic AAAs were detected by an abdominal ultrasound scan in 2011 to 2013. Matrix metalloproteinase (MMP)-3, MMP-9, interleukin 6 (IL-6), N-terminal propeptide of Type III procollagen (PIIINP), and osteopontin were measured in blood samples collected between 1987 and 1992 in participants with AAA (544 clinically diagnosed AAAs and 72 ultrasound-detected AAAs) and a random sample of 723 participants selected from baseline and matched with AAAs by age, race and sex. Higher concentrations of MMP-9 and IL-6 were associated with future risk of clinically diagnosed AAA (hazard ratios [95% confidence intervals]: 1.55 [1.22-1.97] and 1.87 [1.48-2.35], respectively, comparing highest versus lowest tertiles) after multivariable adjustment ( P for trend < .001). Matrix metalloproteinase-9 was also associated with ultrasound-detected AAA. In conclusion, blood concentrations of MMP-9 and IL-6 measured in middle age predicted the risk of AAA during 24 years of follow-up.
Collapse
Affiliation(s)
- Weihong Tang
- 1 Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Lu Yao
- 1 Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Ron C Hoogeveen
- 2 Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Alvaro Alonso
- 3 Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - David J Couper
- 4 Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, NC, USA
| | - Pamela L Lutsey
- 1 Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Carol C Steenson
- 5 Department of Imaging, Minneapolis VA Health Care System, Minneapolis, MN, USA
| | - Weihua Guan
- 6 Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - David W Hunter
- 7 Department of Medicine, School of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Frank A Lederle
- 8 Minneapolis VA Health Care System and Department of Medicine, School of Medicine, University of Minnesota, Minneapolis, MN, USA.,Frank A. Lederle, Deceased January 2018
| | - Aaron R Folsom
- 1 Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
21
|
The Society for Vascular Surgery practice guidelines on the care of patients with an abdominal aortic aneurysm. J Vasc Surg 2018; 67:2-77.e2. [DOI: 10.1016/j.jvs.2017.10.044] [Citation(s) in RCA: 1150] [Impact Index Per Article: 164.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
22
|
HASHIMOTO K, ZAIMA N, SEKIGUCHI H, KUGO H, MIYAMOTO C, HOSHINO K, KAWASAKI N, SUTOH K, USUMI K, MORIYAMA T. Dietary DNA Attenuates the Degradation of Elastin Fibers in the Aortic Wall in Nicotine-Administrated Mice. J Nutr Sci Vitaminol (Tokyo) 2018; 64:271-276. [DOI: 10.3177/jnsv.64.271] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Keisuke HASHIMOTO
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Kindai University
| | - Nobuhiro ZAIMA
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Kindai University
- Agricultural Technology and Innovation Research Institute
| | | | - Hirona KUGO
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Kindai University
| | - Chie MIYAMOTO
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Kindai University
| | - Kiyoto HOSHINO
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Kindai University
| | - Natsumi KAWASAKI
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Kindai University
| | | | | | - Tatsuya MORIYAMA
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Kindai University
- Agricultural Technology and Innovation Research Institute
| |
Collapse
|
23
|
Han SA, Joh JH, Park HC. Risk Factors for Abdominal Aortic Aneurysm in the Korean Population. Ann Vasc Surg 2017; 41:135-140. [DOI: 10.1016/j.avsg.2016.08.044] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 08/04/2016] [Accepted: 08/13/2016] [Indexed: 01/31/2023]
|
24
|
Javed F, Kellesarian SV, Sundar IK, Romanos GE, Rahman I. Recent updates on electronic cigarette aerosol and inhaled nicotine effects on periodontal and pulmonary tissues. Oral Dis 2017; 23:1052-1057. [PMID: 28168771 DOI: 10.1111/odi.12652] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 01/30/2017] [Accepted: 02/01/2017] [Indexed: 01/06/2023]
Abstract
E-cigarette-derived inhaled nicotine may contribute to the pathogenesis of periodontal and pulmonary diseases in particular via lung inflammation, injurious, and dysregulated repair responses. Nicotine is shown to have antiproliferative properties and affects fibroblasts in vitro, which may interfere in tissue myofibroblast differentiation in e-cig users. This will affect the ability to heal wounds by decreasing wound contraction. In periodontics, direct exposure to e-vapor has been shown to produce harmful effects in periodontal ligament and gingival fibroblasts in culture. This is due to the generation of reactive oxygen species/aldehydes/carbonyls from e-cig aerosol, leading to protein carbonylation of extracellular matrix and DNA adducts/damage. A limited number of studies regarding the effects of e-cig in oral and lung health are available. However, no reports are available to directly link the deleterious effects on e-cigs, inhaled nicotine, and flavorings aerosol on periodontal and pulmonary health in particular to identify the risk of oral diseases by e-cigarettes and nicotine aerosols. This mini-review summarizes the recent perspectives on e-cigarettes including inhaled nicotine effects on several pathophysiological events, such as oxidative stress, DNA damage, innate host response, inflammation, cellular senescence, profibrogenic and dysregulated repair, leading to lung remodeling, oral submucous fibrosis, and periodontal diseases.
Collapse
Affiliation(s)
- F Javed
- Department of General Dentistry, Eastman Institute for Oral Health, University of Rochester, Rochester, NY, USA
| | - S V Kellesarian
- Department of General Dentistry, Eastman Institute for Oral Health, University of Rochester, Rochester, NY, USA
| | - I K Sundar
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - G E Romanos
- Department of Periodontology, School of Dental Medicine, Stony Brook University, Stony Brook, NY, USA.,Department of Oral Surgery and Implant Dentistry, Johann Wolfgang Goethe University, Dental School, Frankfurt, Germany
| | - I Rahman
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
25
|
Bchir S, Nasr HB, Bouchet S, Benzarti M, Garrouch A, Tabka Z, Susin S, Chahed K, Bauvois B. Concomitant elevations of MMP-9, NGAL, proMMP-9/NGAL and neutrophil elastase in serum of smokers with chronic obstructive pulmonary disease. J Cell Mol Med 2016; 21:1280-1291. [PMID: 28004483 PMCID: PMC5487915 DOI: 10.1111/jcmm.13057] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 11/10/2016] [Indexed: 12/22/2022] Open
Abstract
A growing body of evidence points towards smoking‐related phenotypic differences in chronic obstructive pulmonary disease (COPD). As COPD is associated with systemic inflammation, we determined whether smoking status is related to serum levels of matrix metalloproteinase‐9 (pro‐ and active MMP‐9), neutrophil gelatinase‐associated lipocalin (NGAL) and the proMMP‐9/NGAL complex in patients with COPD. Serum samples were collected in 100 stable‐phase COPD patients (82 smokers, 18 never‐smokers) and 28 healthy adults (21 smokers, 7 never‐smokers). Serum levels of studied factors were measured in ELISA. Our data provide the first evidence of simultaneously elevated serum levels of MMP‐9, NGAL and proMMP‐9/NGAL in COPD smokers. While the triad discriminated between smokers and non‐smokers in the COPD group, MMP‐9 and proMMP‐9/NGAL (but not NGAL) discriminated between smokers with and without COPD. Adjustment for age and smoking pack‐years did not alter the findings. Serum MMP‐9, NGAL and proMMP‐9/NGAL levels were not correlated with the GOLD stage or FEV1 decline. Furthermore, serum levels of neutrophil elastase (NE) and MMP‐3 (but not of IL‐6 and MMP‐12) were also higher in COPD smokers than in healthy smokers before and after adjustment for age and pack‐years. Among COPD smokers, levels of MMP‐9, NGAL and proMMP‐9/NGAL were positively correlated with NE (P < 0.0001) but not with the remaining factors. Gelatin zymography detected proMMP‐9 in serum samples of healthy and COPD smoking groups. Our results suggest that associated serum levels of proMMP‐9, NGAL, proMMP‐9/NGAL and NE may reflect the state of systemic inflammation in COPD related to cigarette smoking.
Collapse
Affiliation(s)
- Sarra Bchir
- Unité de recherche UR12ES06, Physiologie de l'Exercice et Physiopathologie de l'Intégré au Moléculaire, Biologie, Médecine et Santé, Faculté de Médecine de Sousse, Université de Sousse, Sousse, Tunisia.,Institut Supérieur de Biotechnologie de Monastir, Université de Monastir, Monastir, Tunisia.,Centre de Recherche des Cordeliers, INSERM UMRS1138, Sorbonne Universités UPMC Paris 06, Université Paris Descartes Sorbonne Paris Cité, Paris, France
| | - Hela Ben Nasr
- Unité de recherche UR12ES06, Physiologie de l'Exercice et Physiopathologie de l'Intégré au Moléculaire, Biologie, Médecine et Santé, Faculté de Médecine de Sousse, Université de Sousse, Sousse, Tunisia
| | - Sandrine Bouchet
- Centre de Recherche des Cordeliers, INSERM UMRS1138, Sorbonne Universités UPMC Paris 06, Université Paris Descartes Sorbonne Paris Cité, Paris, France.,Assistance Publique des Hôpitaux de Paris, Paris, France
| | - Mohamed Benzarti
- Service de Pneumo-Allergologie, CHU Farhat Hached, Sousse, Tunisia
| | | | - Zouhair Tabka
- Unité de recherche UR12ES06, Physiologie de l'Exercice et Physiopathologie de l'Intégré au Moléculaire, Biologie, Médecine et Santé, Faculté de Médecine de Sousse, Université de Sousse, Sousse, Tunisia
| | - Santos Susin
- Centre de Recherche des Cordeliers, INSERM UMRS1138, Sorbonne Universités UPMC Paris 06, Université Paris Descartes Sorbonne Paris Cité, Paris, France
| | - Karim Chahed
- Unité de recherche UR12ES06, Physiologie de l'Exercice et Physiopathologie de l'Intégré au Moléculaire, Biologie, Médecine et Santé, Faculté de Médecine de Sousse, Université de Sousse, Sousse, Tunisia.,Faculté des Sciences de Sfax, Université de Sfax, Sfax, Tunisia
| | - Brigitte Bauvois
- Centre de Recherche des Cordeliers, INSERM UMRS1138, Sorbonne Universités UPMC Paris 06, Université Paris Descartes Sorbonne Paris Cité, Paris, France
| |
Collapse
|
26
|
SP600125 Attenuates Nicotine-Related Aortic Aneurysm Formation by Inhibiting Matrix Metalloproteinase Production and CC Chemokine-Mediated Macrophage Migration. Mediators Inflamm 2016; 2016:9142425. [PMID: 27688602 PMCID: PMC5023844 DOI: 10.1155/2016/9142425] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 06/13/2016] [Indexed: 12/27/2022] Open
Abstract
Nicotine, a major chemical component of cigarettes, plays a pivotal role in the development of abdominal aortic aneurysm (AAA). c-Jun N-terminal kinase (JNK) has been demonstrated to participate in elastase-induced AAA. This study aimed to elucidate whether the JNK inhibitor SP600125 can attenuate nicotine plus angiotensin II- (AngII-) induced AAA formation and to assess the underlying molecular mechanisms. SP600125 significantly attenuated nicotine plus AngII-induced AAA formation. The expression of matrix metalloproteinase- (MMP-) 2, MMP-9, monocyte chemoattractant protein- (MCP-) 1, and regulated-on-activation, normal T-cells expressed and secreted (RANTES) was significantly upregulated in aortic aneurysm lesions but inhibited by SP600125. In vitro, nicotine induced the expression of MCP-1 and RANTES in both RAW264.7 (mouse macrophage) and MOVAS (mouse vascular smooth muscle) cells in a dose-dependent manner; expression was upregulated by 0.5 ng/mL nicotine but strongly downregulated by 500 ng/mL nicotine. SP600125 attenuated the upregulation of MCP-1 and RANTES expression and subsequent macrophage migration. In conclusion, SP600125 attenuates nicotine plus AngII-induced AAA formation likely by inhibiting MMP-2, MMP-9, MCP-1, and RANTES. The expression of chemokines in MOVAS cells induced by nicotine has an effect on RAW264.7 migration, which is likely to contribute to the development of nicotine-related AAA.
Collapse
|
27
|
Gordon P, Flanagan P. Smoking: A risk factor for vascular disease. JOURNAL OF VASCULAR NURSING 2016; 34:79-86. [DOI: 10.1016/j.jvn.2016.04.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 04/04/2016] [Accepted: 04/04/2016] [Indexed: 01/05/2023]
|
28
|
Piechota-Polanczyk A, Jozkowicz A, Nowak W, Eilenberg W, Neumayer C, Malinski T, Huk I, Brostjan C. The Abdominal Aortic Aneurysm and Intraluminal Thrombus: Current Concepts of Development and Treatment. Front Cardiovasc Med 2015; 2:19. [PMID: 26664891 PMCID: PMC4671358 DOI: 10.3389/fcvm.2015.00019] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 04/10/2015] [Indexed: 01/09/2023] Open
Abstract
The pathogenesis of the abdominal aortic aneurysm (AAA) shows several hallmarks of atherosclerotic and atherothrombotic disease, but comprises an additional, predominant feature of proteolysis resulting in the degradation and destabilization of the aortic wall. This review aims to summarize the current knowledge on AAA development, involving the accumulation of neutrophils in the intraluminal thrombus and their central role in creating an oxidative and proteolytic environment. Particular focus is placed on the controversial role of heme oxygenase 1/carbon monoxide and nitric oxide synthase/peroxynitrite, which may exert both protective and damaging effects in the development of the aneurysm. Treatment indications as well as surgical and pharmacological options for AAA therapy are discussed in light of recent reports.
Collapse
Affiliation(s)
- Aleksandra Piechota-Polanczyk
- Department of Surgery, Medical University of Vienna , Vienna , Austria ; Department of Biochemistry, Medical University of Lodz , Lodz , Poland
| | - Alicja Jozkowicz
- Department of Medical Biotechnology, Jagiellonian University , Krakow , Poland
| | - Witold Nowak
- Department of Medical Biotechnology, Jagiellonian University , Krakow , Poland
| | - Wolf Eilenberg
- Department of Surgery, Medical University of Vienna , Vienna , Austria
| | | | - Tadeusz Malinski
- Department of Chemistry and Biochemistry, Ohio University , Athens, OH , USA
| | - Ihor Huk
- Department of Surgery, Medical University of Vienna , Vienna , Austria
| | | |
Collapse
|
29
|
Schraufnagel DE, Blasi F, Drummond MB, Lam DCL, Latif E, Rosen MJ, Sansores R, Van Zyl-Smit R. Electronic cigarettes. A position statement of the forum of international respiratory societies. Am J Respir Crit Care Med 2014; 190:611-8. [PMID: 25006874 DOI: 10.1164/rccm.201407-1198pp] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Awareness and usage of electronic cigarettes has exponentially increased during the last few years, especially among young people and women in some countries. The rapid acceptance of electronic cigarettes may be attributed in part to the perception created by marketing and the popular press that they are safer than combustible cigarettes. GOALS To alert and advise policy makers about electronic cigarettes and their potential hazards. METHODS Using The Union's position paper on electronic cigarettes as the starting template, the document was written using an iterative process. Portions of the manuscript have been taken directly from the position papers of participating societies. RESULTS Because electronic cigarettes generate less tar and carcinogens than combustible cigarettes, use of electronic cigarettes may reduce disease caused by those components. However, the health risks of electronic cigarettes have not been adequately studied. Studies looking at whether electronic cigarettes can aid smoking cessation have had inconsistent results. Moreover, the availability of electronic cigarettes may have an overall adverse health impact by increasing initiation and reducing cessation of combustible nicotine delivery products. CONCLUSIONS The health and safety claims regarding electronic nicotine delivery devices should be subject to evidentiary review. The potential benefits of electronic cigarettes to an individual smoker should be weighed against potential harm to the population of increased social acceptability of smoking and use of nicotine, the latter of which has addictive power and untoward effects. As a precaution, electronic nicotine delivery devices should be restricted or banned until more information about their safety is available. If they are allowed, they should be closely regulated as medicines or tobacco products.
Collapse
Affiliation(s)
- Dean E Schraufnagel
- 1 Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Wei Z, Wang Y, Zhang K, Liao Y, Ye P, Wu J, Wang Y, Li F, Yao Y, Zhou Y, Liu J. Inhibiting the Th17/IL-17A–Related Inflammatory Responses With Digoxin Confers Protection Against Experimental Abdominal Aortic Aneurysm. Arterioscler Thromb Vasc Biol 2014; 34:2429-38. [DOI: 10.1161/atvbaha.114.304435] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Zhanjie Wei
- From the Department of Cardiovascular Surgery, Union Hospital (Z.W., K.Z., Y.L., P.Y., J.W., Y.W., F.L., J.L.) and Department of Biochemistry and Molecular Biology (Y.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Cardiology, Central Hospital of Wuhan, Wuhan, China (P.Y.); and Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Institute, College of Life Science and Technology and Center for Human Genome Research (Y.Y
| | - Yu Wang
- From the Department of Cardiovascular Surgery, Union Hospital (Z.W., K.Z., Y.L., P.Y., J.W., Y.W., F.L., J.L.) and Department of Biochemistry and Molecular Biology (Y.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Cardiology, Central Hospital of Wuhan, Wuhan, China (P.Y.); and Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Institute, College of Life Science and Technology and Center for Human Genome Research (Y.Y
| | - Kailun Zhang
- From the Department of Cardiovascular Surgery, Union Hospital (Z.W., K.Z., Y.L., P.Y., J.W., Y.W., F.L., J.L.) and Department of Biochemistry and Molecular Biology (Y.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Cardiology, Central Hospital of Wuhan, Wuhan, China (P.Y.); and Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Institute, College of Life Science and Technology and Center for Human Genome Research (Y.Y
| | - Yaohang Liao
- From the Department of Cardiovascular Surgery, Union Hospital (Z.W., K.Z., Y.L., P.Y., J.W., Y.W., F.L., J.L.) and Department of Biochemistry and Molecular Biology (Y.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Cardiology, Central Hospital of Wuhan, Wuhan, China (P.Y.); and Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Institute, College of Life Science and Technology and Center for Human Genome Research (Y.Y
| | - Ping Ye
- From the Department of Cardiovascular Surgery, Union Hospital (Z.W., K.Z., Y.L., P.Y., J.W., Y.W., F.L., J.L.) and Department of Biochemistry and Molecular Biology (Y.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Cardiology, Central Hospital of Wuhan, Wuhan, China (P.Y.); and Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Institute, College of Life Science and Technology and Center for Human Genome Research (Y.Y
| | - Jie Wu
- From the Department of Cardiovascular Surgery, Union Hospital (Z.W., K.Z., Y.L., P.Y., J.W., Y.W., F.L., J.L.) and Department of Biochemistry and Molecular Biology (Y.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Cardiology, Central Hospital of Wuhan, Wuhan, China (P.Y.); and Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Institute, College of Life Science and Technology and Center for Human Genome Research (Y.Y
| | - Yang Wang
- From the Department of Cardiovascular Surgery, Union Hospital (Z.W., K.Z., Y.L., P.Y., J.W., Y.W., F.L., J.L.) and Department of Biochemistry and Molecular Biology (Y.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Cardiology, Central Hospital of Wuhan, Wuhan, China (P.Y.); and Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Institute, College of Life Science and Technology and Center for Human Genome Research (Y.Y
| | - Feifei Li
- From the Department of Cardiovascular Surgery, Union Hospital (Z.W., K.Z., Y.L., P.Y., J.W., Y.W., F.L., J.L.) and Department of Biochemistry and Molecular Biology (Y.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Cardiology, Central Hospital of Wuhan, Wuhan, China (P.Y.); and Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Institute, College of Life Science and Technology and Center for Human Genome Research (Y.Y
| | - Yufeng Yao
- From the Department of Cardiovascular Surgery, Union Hospital (Z.W., K.Z., Y.L., P.Y., J.W., Y.W., F.L., J.L.) and Department of Biochemistry and Molecular Biology (Y.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Cardiology, Central Hospital of Wuhan, Wuhan, China (P.Y.); and Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Institute, College of Life Science and Technology and Center for Human Genome Research (Y.Y
| | - Yanzhao Zhou
- From the Department of Cardiovascular Surgery, Union Hospital (Z.W., K.Z., Y.L., P.Y., J.W., Y.W., F.L., J.L.) and Department of Biochemistry and Molecular Biology (Y.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Cardiology, Central Hospital of Wuhan, Wuhan, China (P.Y.); and Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Institute, College of Life Science and Technology and Center for Human Genome Research (Y.Y
| | - Jinping Liu
- From the Department of Cardiovascular Surgery, Union Hospital (Z.W., K.Z., Y.L., P.Y., J.W., Y.W., F.L., J.L.) and Department of Biochemistry and Molecular Biology (Y.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Cardiology, Central Hospital of Wuhan, Wuhan, China (P.Y.); and Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Institute, College of Life Science and Technology and Center for Human Genome Research (Y.Y
| |
Collapse
|
31
|
Colombo G, Clerici M, Giustarini D, Portinaro NM, Aldini G, Rossi R, Milzani A, Dalle-Donne I. Pathophysiology of tobacco smoke exposure: recent insights from comparative and redox proteomics. MASS SPECTROMETRY REVIEWS 2014; 33:183-218. [PMID: 24272816 DOI: 10.1002/mas.21392] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Revised: 05/23/2013] [Accepted: 05/23/2013] [Indexed: 06/02/2023]
Abstract
First-hand and second-hand tobacco smoke are causally linked to a huge number of deaths and are responsible for a broad spectrum of pathologies such as cancer, cardiovascular, respiratory, and eye diseases as well as adverse effects on female reproductive function. Cigarette smoke is a complex mixture of thousands of different chemical species, which exert their negative effects on macromolecules and biochemical pathways, both directly and indirectly. Many compounds can act as oxidants, pro-inflammatory agents, carcinogens, or a combination of these. The redox behavior of cigarette smoke has many implications for smoke related diseases. Reactive oxygen and nitrogen species (both radicals and non-radicals), reactive carbonyl compounds, and other species may induce oxidative damage in almost all the biological macromolecules, compromising their structure and/or function. Different quantitative and redox proteomic approaches have been applied in vitro and in vivo to evaluate, respectively, changes in protein expression and specific oxidative protein modifications induced by exposure to cigarette smoke and are overviewed in this review. Many gel-based and gel-free proteomic techniques have already been used successfully to obtain clues about smoke effects on different proteins in cell cultures, animal models, and humans. The further implementation with other sensitive screening techniques could be useful to integrate the comprehension of cigarette smoke effects on human health. In particular, the redox proteomic approach may also help identify biomarkers of exposure to tobacco smoke useful for preventing these effects or potentially predictive of the onset and/or progression of smoking-induced diseases as well as potential targets for therapeutic strategies.
Collapse
Affiliation(s)
- Graziano Colombo
- Department of Biosciences, Università degli Studi di Milano, Milan, Italy
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Correlation of bacterial coinfection versus matrix metalloproteinase 9 and tissue inhibitor of metalloproteinase 1 expression in aortic aneurysm and atherosclerosis. Ann Vasc Surg 2014; 27:964-71. [PMID: 23993112 DOI: 10.1016/j.avsg.2013.02.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Revised: 02/15/2013] [Accepted: 02/22/2013] [Indexed: 01/27/2023]
Abstract
BACKGROUND We searched for any relationship between Chlamydophila pneumoniae, Mycoplasma pneumoniae, matrix metalloproteinase 9 (MMP-9), and tissue inhibitor of metalloproteinase 1 (TIMP-1) in aneurysmatic atherosclerotic lesions, and whether this relationship differed from that in atherosclerotic nonaneurysmatic lesions. METHODS Twenty-eight tissue samples paired by age and sex were grouped as follows: group 1 included 14 nonaneurysmal atherosclerotic fragments obtained from abdominal aortas collected from necropsies; group 2 included 14 aneurysmatic atherosclerotic aortic fragments obtained from patients during corrective surgery. Immunohistochemistry reactions were evaluated for C pneumoniae, M pneumoniae, MMP-9, and TIMP-1 antigens. Both groups were compared using the Mann-Whitney test, and the correlations among variables were obtained using the Spearman correlation test. P ≤ 0.05 was considered statistically significant. RESULTS C pneumoniae and M pneumoniae antigens were detected in 100% of cases. A higher amount of C pneumoniae (P = 0.005), M pneumoniae (P = 0.002), and MMP-9 (P = 0.021) was found in adventitia of group 2 with aneurysm. A positive correlation was found in the aneurysm group, as follows: intima C pneumoniae versus adventitia thickness (r = 0.70; P = 0.01), media C pneumoniae versus adventitia C pneumoniae (r = 0.75; P = 0.002), intima C pneumoniae versus media C pneumoniae (r = 0.8; P = 0.00), and adventitia C pneumoniae versus intima M pneumoniae (r = 0.54; P = 0.05); negative correlations were as follows: adventitia thickness and adventitia M pneumoniae (r = -0.65; P = 0.01), media MMP-9 and media thickness (r = -0.55; P = 0.04), TIMP-1 media versus adventitia C pneumoniae (r = -0.86; P = 0.00), and TIMP-1 media versus M pneumoniae intima (r = -0.67; P = 0.03). Nonaneurysmal atherosclerotic group 1 results are as follows: adventitia C pneumoniae versus TIMP-1 media (r = 0.75; P = 0.01) and media C pneumoniae and adventitia C pneumoniae (r = 0.59; P = 0.03). CONCLUSIONS The present work favors a role for coinfection of both M pneumoniae and C pneumoniae in the development of aortic atherosclerotic aneurysm, with increased adventitial inflammation, inhibition of TIMP-1 activity, and increased collagen degradation.
Collapse
|
33
|
Cathepsins: a new culprit behind abdominal aortic aneurysm. Regen Med Res 2013; 1:5. [PMID: 25984324 PMCID: PMC4431531 DOI: 10.1186/2050-490x-1-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 09/19/2013] [Indexed: 01/17/2023] Open
Abstract
Abdominal aortic aneurysm (AAA) is a fatal disease defined as an abdominal aortic diameter of 3.0 cm or more, where the abdominal aorta exceeds the normal diameter by more than 50%. Histopathological changes of AAA mainly include extracellular matrix (ECM) remodeling at the abdominal aorta wall, but there is lack of specific drugs to treat AAA. Recent studies have reported that lysosomal cathepsins could induce vascular remodeling and AAA formation by regulating vascular inflammation, medial smooth muscle cell apoptosis, neovascularization, and protease expression. Thus, cathepsins are expected to become a new therapeutic target for AAA treatment.
Collapse
|
34
|
Jaldin RG, Castardelli É, Perobelli JE, Yoshida WB, de Castro Rodrigues A, Sequeira JL, Paiva SAR. Morphologic and Biomechanical Changes of Thoracic and Abdominal Aorta in a Rat Model of Cigarette Smoke Exposure. Ann Vasc Surg 2013; 27:791-800. [DOI: 10.1016/j.avsg.2013.03.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2012] [Revised: 02/12/2013] [Accepted: 03/22/2013] [Indexed: 10/26/2022]
|
35
|
Norman PE, Curci JA. Understanding the effects of tobacco smoke on the pathogenesis of aortic aneurysm. Arterioscler Thromb Vasc Biol 2013; 33:1473-7. [PMID: 23685557 DOI: 10.1161/atvbaha.112.300158] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Aneurysmal arterial disease is a vascular degenerative condition that is distinct from atherosclerotic and other occlusive arterial diseases. There is regionalization of the predisposition to aneurysm formation within the vascular tree, and the pathological process varies with location. Infrarenal abdominal aortic aneurysm (AAA) is the most common manifestation of aneurysmal disease, and smoking is the dominant risk factor. Smoking is a much greater risk factor for AAA than for atherosclerosis. In addition to playing a role in the pathogenesis of AAA, smoking also increases the rate of expansion and risk of rupture of established AAA. The mechanistic relationship between AAA and smoking is being established by the use of enhanced animal models that are dependent on smoke or smoke components. The mechanisms seem to involve durable alterations in vascular smooth muscle cell and inflammatory cell function. This review examines the clinical, epidemiological, and mechanistic evidence implicating smoking as a cause of aneurysms, focusing on AAA.
Collapse
Affiliation(s)
- Paul E Norman
- School of Surgery, University of Western Australia, Fremantle, Australia.
| | | |
Collapse
|
36
|
Joel DL, Denlinger RL, Dermody SS, Hatsukami DK, Benowitz NL, Donny EC. Very low nicotine content cigarettes and potential consequences on cardiovascular disease. CURRENT CARDIOVASCULAR RISK REPORTS 2012; 6:534-541. [PMID: 23264843 DOI: 10.1007/s12170-012-0266-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cigarette smoking remains highly prevalent in the U.S. and contributes significantly to cardiovascular disease (CVD). Tobacco control policies, including product regulation, can reduce smoking-related harm. One approach being considered in the U.S. is for the FDA to set a low nicotine standard for cigarettes. Such a standard could result in multiple beneficial outcomes including reduced cardiovascular toxicity related to nicotine, reduced smoking intensity in current smokers, increased cessation rates, decreased development of smoking dependence in youth, and decreased passive smoke exposure. Consequently, CVD risk in the U.S. could be dramatically improved by nicotine reduction in cigarettes. Possible pathways linking nicotine reduction in cigarettes to decreased CVD risk are discussed, while potential unintended consequences that could offset expected gains are also presented. Gaps in the literature, including limited data on CVD biomarkers and long-term CVD outcomes following the use of very low nicotine cigarettes, are discussed to highlight areas for new research.
Collapse
Affiliation(s)
- Danielle L Joel
- Department of Psychology University of Pittsburgh 4311 Sennott Square 201 S. Bouquet Street Pittsburgh, PA 15260
| | | | | | | | | | | |
Collapse
|