1
|
Zhu J, Meganathan I, MacAruthur R, Kassiri Z. Inflammation in Abdominal Aortic Aneurysm: Cause or Comorbidity? Can J Cardiol 2024:S0828-282X(24)00926-7. [PMID: 39181326 DOI: 10.1016/j.cjca.2024.08.274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/12/2024] [Accepted: 08/15/2024] [Indexed: 08/27/2024] Open
Abstract
Aortic aneurysm is a potentially deadly disease. It is chronic degeneration of the aortic wall that involves an inflammatory response and the immune system, aberrant remodelling of the extracellular matrix, and maladaptive transformation of the aortic cells. This review article focuses on the role of the inflammatory cells in abdominal aortic aneurysm. Studies in human aneurysmal specimens and animal models have identified various inflammatory cell types that could contribute to formation or expansion of aneurysms. These include the commonly studied leukocytes (neutrophils and macrophages) as well as the less commonly explored natural killer cells, dendritic cells, T cells, and B cells. Despite the well-demonstrated contribution of inflammatory cells and the related signalling pathways to development and expansion of aneurysms, anti-inflammatory therapy approaches have demonstrated limitations and may require additional considerations such as a combinational approach in targeting multiple pathways for significant beneficial outcomes.
Collapse
Affiliation(s)
- Jiechun Zhu
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Ilamaran Meganathan
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Roderick MacAruthur
- Department of Cardiac Surgery, Mazankowski Alberta Heart Institute, University of Alberta Hospital, Edmonton, Alberta, Canada
| | - Zamaneh Kassiri
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
2
|
Gong W, Tian Y, Li L. T cells in abdominal aortic aneurysm: immunomodulation and clinical application. Front Immunol 2023; 14:1240132. [PMID: 37662948 PMCID: PMC10471798 DOI: 10.3389/fimmu.2023.1240132] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 08/07/2023] [Indexed: 09/05/2023] Open
Abstract
Abdominal aortic aneurysm (AAA) is characterized by inflammatory cell infiltration, extracellular matrix (ECM) degradation, and vascular smooth muscle cell (SMC) dysfunction. The inflammatory cells involved in AAA mainly include immune cells including macrophages, neutrophils, T-lymphocytes and B lymphocytes and endothelial cells. As the blood vessel wall expands, more and more lymphocytes infiltrate into the outer membrane. It was found that more than 50% of lymphocytes in AAA tissues were CD3+ T cells, including CD4+, CD8+T cells, γδ T cells and regulatory T cells (Tregs). Due to the important role of T cells in inflammatory response, an increasing number of researchers have paid attention to the role of T cells in AAA and dug into the relevant mechanism. Therefore, this paper focuses on reviewing the immunoregulatory role of T cells in AAA and their role in immunotherapy, seeking potential targets for immunotherapy and putting forward future research directions.
Collapse
Affiliation(s)
| | | | - Lei Li
- Department of Vascular Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
3
|
Patel R, Hall S, Lanford H, Ward N, Grespin RT, Figueroa M, Mattia V, Xiong Y, Mukherjee R, Jones J, Ruddy JM. Signaling through the IL-6-STAT3 Pathway Promotes Proteolytically-Active Macrophage Accumulation Necessary for Development of Small AAA. Vasc Endovascular Surg 2023; 57:433-444. [PMID: 36639147 PMCID: PMC10238619 DOI: 10.1177/15385744231152961] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
INTRODUCTION Elevated interleukin-6 (IL-6) plasma levels have been associated with abdominal aortic aneurysm (AAA), but whether this cytokine plays a causative role in the degenerative remodeling or represents an effect from the inflammatory cascades initiated by infiltrating leukocytes remained unclear. This project aims to demonstrate that within the aortic wall, signaling from IL-6 through the STAT3 transcription factor is necessary for infiltration of proteolytically-active macrophages and development of small AAA. METHODS Following measurement of baseline infrarenal aortic diameter (AoD, digital microscopy), C57Bl/6 and IL-6 knockout (IL-6KO) mice underwent AAA induction by application of peri-adventitial CaCl2 (0.5 M) +/- implantation of an osmotic mini-pump delivering IL-6 (4.36 µg/kg/day over 21 days). At the terminal procedure, AoDs were measured by digital microscopy and aortas harvested for immunoblot (pSTAT3/STAT3), matrix metalloproteinase (MMP) quantification, or flow cytometric analysis of macrophage content. Plasma was collected for cytokine analysis. RESULTS IL-6 infusion significantly increased the plasma IL-6 levels in C57Bl/6 and IL-6KO animals. The C57Bl/6 + CaCl2 group developed AAA (AoD >50% above baseline) but IL-6KO + CaCl2 did not. In the IL-6KO + IL-6+CaCl2 group, AAA developed to match that of C57Bl/6 + CaCl2 mice. STAT3 activity was significantly increased in animals with advanced stages of dilation (>40% from baseline), compared to those with ectasia (≤25%). Although cytokine profiles did not support T-cells or neutrophils as being active contributors in this stage of aortic remodeling, changes in the profile of elaborated MMPs suggested macrophage activity with a trend toward alternatively activated pathways. Flow cytometry confirmed significantly increased macrophage abundance specifically in animals with upregulated STAT3 activity and advanced aortic dilation. CONCLUSION In this murine model of AAA, progressive dilation to development of true AAA was only accomplished when IL-6 signaling upregulated STAT3 activity to effect accumulation of proteolytically-active macrophages. This pathway warrants further investigation to identify potential therapeutic avenues to abrogate growth of small AAA.
Collapse
Affiliation(s)
- Raj Patel
- Division of Vascular Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - SarahRose Hall
- Division of Vascular Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Hayes Lanford
- Division of Vascular Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Nicholas Ward
- Division of Vascular Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - R. Tyler Grespin
- Division of Vascular Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Mario Figueroa
- Division of Vascular Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Victoria Mattia
- Division of Vascular Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Ying Xiong
- Division of Cardiothoracic Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Rupak Mukherjee
- Division of Cardiothoracic Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Jeffrey Jones
- Division of Cardiothoracic Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Jean Marie Ruddy
- Division of Vascular Surgery, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
4
|
Association of immunological parameters with aortic dilatation in giant cell arteritis: a cross-sectional study. Rheumatol Int 2023; 43:477-485. [PMID: 35996028 PMCID: PMC9968266 DOI: 10.1007/s00296-022-05186-1] [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: 05/09/2022] [Accepted: 08/08/2022] [Indexed: 10/15/2022]
Abstract
Aortic dilatation (AD) occurs in up to 30% of patients with giant cell arteritis (GCA). Reliable biomarkers for AD development, however, are still absent. The aim of this exploratory study was to evaluate whether immunological parameters are associated with the occurrence of AD in GCA. Cross-sectional study on 20 GCA patients with AD, 20 GCA patients without AD, and 20 non-GCA controls without AD measuring leukocytes, neutrophils, lymphocytes, C-reactive protein (CRP), erythrocyte sedimentation rate (ESR), serum amyloid A (SAA), interferon (IFN)-α, IFN-γ, IFN-γ-induced protein 10 (IP-10), interleukin (IL) 5, IL-8, IL-10, IL-17A, IL-18, IL-1 receptor antagonist, tumor necrosis factor (TNF)-α, platelet-derived growth factor (PDGF), L-selectin, P-selectin, and soluble intercellular adhesion molecule 1 (sICAM-1). AD was measured by aortic contrast-enhanced computed tomography and defined by enlargement of the aorta above population-based aortic diameters adjusted by age, gender, and body surface area. No significant differences were observed between GCA patients with AD and GCA patients without AD concerning levels of leukocytes, neutrophils, lymphocytes, CRP, ESR, SAA, IL-8, IL-18, PDGF, IP-10, selectins, and sICAM-1. Values of IFN-α, IFN-γ, IL-5, IL-10, IL-17A, IL-1 receptor antagonist, and TNF-α were all below the detection limits in more than 70% of subjects. Lymphocytes and CRP revealed positive correlations with the diameter of the thoracic descending aorta. Immunological parameters were not useful to conclude on the presence of AD in GCA. Further studies are required to test if CRP and lymphocytes may be useful to predict future development of AD in GCA.
Collapse
|
5
|
Márquez-Sánchez AC, Koltsova EK. Immune and inflammatory mechanisms of abdominal aortic aneurysm. Front Immunol 2022; 13:989933. [PMID: 36275758 PMCID: PMC9583679 DOI: 10.3389/fimmu.2022.989933] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 08/23/2022] [Indexed: 11/13/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) is a life-threatening cardiovascular disease. Immune-mediated infiltration and a destruction of the aortic wall during AAA development plays significant role in the pathogenesis of this disease. While various immune cells had been found in AAA, the mechanisms of their activation and function are still far from being understood. A better understanding of mechanisms regulating the development of aberrant immune cell activation in AAA is essential for the development of novel preventive and therapeutic approaches. In this review we summarize current knowledge about the role of immune cells in AAA and discuss how pathogenic immune cell activation is regulated in this disease.
Collapse
|
6
|
Griepke S, Grupe E, Lindholt JS, Fuglsang EH, Steffensen LB, Beck HC, Larsen MD, Bang-Møller SK, Overgaard M, Rasmussen LM, Lambertsen KL, Stubbe J. Selective inhibition of soluble tumor necrosis factor signaling reduces abdominal aortic aneurysm progression. Front Cardiovasc Med 2022; 9:942342. [PMID: 36186984 PMCID: PMC9523116 DOI: 10.3389/fcvm.2022.942342] [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: 05/12/2022] [Accepted: 08/08/2022] [Indexed: 11/21/2022] Open
Abstract
Background Tumor necrosis factor (TNF) is pathologically elevated in human abdominal aortic aneurysms (AAA). Non-selective TNF inhibition-based therapeutics are approved for human use but have been linked to several side effects. Compounds that target the proinflammatory soluble form of TNF (solTNF) but preserve the immunomodulatory capabilities of the transmembrane form of TNF (tmTNF) may prevent these side effects. We hypothesize that inhibition of solTNF signaling prevents AAA expansion. Methods The effect of the selective solTNF inhibitor, XPro1595, and the non-selective TNF inhibitor, Etanercept (ETN) was examined in porcine pancreatic elastase (PPE) induced AAA mice, and findings with XPro1595 was confirmed in angiotensin II (ANGII) induced AAA in hyperlipidemic apolipoprotein E (Apoe) -/- mice. Results XPro1595 treatment significantly reduced AAA expansion in both models, and a similar trend (p = 0.06) was observed in PPE-induced AAA in ETN-treated mice. In the PPE aneurysm wall, XPro1595 improved elastin integrity scores. In aneurysms, mean TNFR1 levels reduced non-significantly (p = 0.07) by 50% after TNF inhibition, but the histological location in murine AAAs was unaffected and similar to that in human AAAs. Semi-quantification of infiltrating leucocytes, macrophages, T-cells, and neutrophils in the aneurysm wall were unaffected by TNF inhibition. XPro1595 increased systemic TNF levels, while ETN increased systemic IL-10 levels. In ANGII-induced AAA mice, XPro1595 increased systemic TNF and IL-5 levels. In early AAA development, proteomic analyses revealed that XPro1595 significantly upregulated ontology terms including "platelet aggregation" and "coagulation" related to the fibrinogen complex, from which several proteins were among the top regulated proteins. Downregulated ontology terms were associated with metabolic processes. Conclusion In conclusion, selective inhibition of solTNF signaling reduced aneurysm expansion in mice, supporting its potential as an attractive treatment option for AAA patients.
Collapse
Affiliation(s)
- Silke Griepke
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Emilie Grupe
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Jes Sanddal Lindholt
- Elite Research Centre for Individualized Medicine in Arterial Diseases (CIMA), Odense University Hospital, Odense, Denmark
- Department of Cardiothoracic and Vascular Surgery, Odense University Hospital, Odense, Denmark
| | - Elizabeth Hvitfeldt Fuglsang
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Lasse Bach Steffensen
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Hans Christian Beck
- Elite Research Centre for Individualized Medicine in Arterial Diseases (CIMA), Odense University Hospital, Odense, Denmark
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark
| | - Mia Dupont Larsen
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Sissel Karoline Bang-Møller
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Martin Overgaard
- Elite Research Centre for Individualized Medicine in Arterial Diseases (CIMA), Odense University Hospital, Odense, Denmark
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark
| | - Lars Melholt Rasmussen
- Elite Research Centre for Individualized Medicine in Arterial Diseases (CIMA), Odense University Hospital, Odense, Denmark
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark
| | - Kate Lykke Lambertsen
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Department of Neurology, Odense University Hospital, Odense, Denmark
- BRIDGE—Brain Research—Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Jane Stubbe
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Elite Research Centre for Individualized Medicine in Arterial Diseases (CIMA), Odense University Hospital, Odense, Denmark
| |
Collapse
|
7
|
Rombouts KB, van Merrienboer TAR, Ket JCF, Bogunovic N, van der Velden J, Yeung KK. The role of vascular smooth muscle cells in the development of aortic aneurysms and dissections. Eur J Clin Invest 2022; 52:e13697. [PMID: 34698377 PMCID: PMC9285394 DOI: 10.1111/eci.13697] [Citation(s) in RCA: 68] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 09/12/2021] [Accepted: 10/11/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND Aortic aneurysms (AA) are pathological dilations of the aorta, associated with an overall mortality rate up to 90% in case of rupture. In addition to dilation, the aortic layers can separate by a tear within the layers, defined as aortic dissections (AD). Vascular smooth muscle cells (vSMC) are the predominant cell type within the aortic wall and dysregulation of vSMC functions contributes to AA and AD development and progression. However, since the exact underlying mechanism is poorly understood, finding potential therapeutic targets for AA and AD is challenging and surgery remains the only treatment option. METHODS In this review, we summarize current knowledge about vSMC functions within the aortic wall and give an overview of how vSMC functions are altered in AA and AD pathogenesis, organized per anatomical location (abdominal or thoracic aorta). RESULTS Important functions of vSMC in healthy or diseased conditions are apoptosis, phenotypic switch, extracellular matrix regeneration and degradation, proliferation and contractility. Stressors within the aortic wall, including inflammatory cell infiltration and (epi)genetic changes, modulate vSMC functions and cause disturbance of processes within vSMC, such as changes in TGF-β signalling and regulatory RNA expression. CONCLUSION This review underscores a central role of vSMC dysfunction in abdominal and thoracic AA and AD development and progression. Further research focused on vSMC dysfunction in the aortic wall is necessary to find potential targets for noninvasive AA and AD treatment options.
Collapse
Affiliation(s)
- Karlijn B Rombouts
- Department of Surgery, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center and AMC, Amsterdam, The Netherlands.,Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center, Amsterdam, The Netherlands
| | - Tara A R van Merrienboer
- Department of Surgery, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center and AMC, Amsterdam, The Netherlands.,Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center, Amsterdam, The Netherlands
| | | | - Natalija Bogunovic
- Department of Surgery, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center and AMC, Amsterdam, The Netherlands.,Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center, Amsterdam, The Netherlands.,Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jolanda van der Velden
- Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center, Amsterdam, The Netherlands
| | - Kak Khee Yeung
- Department of Surgery, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center and AMC, Amsterdam, The Netherlands.,Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
8
|
Xu JY, Xiong YY, Tang RJ, Jiang WY, Ning Y, Gong ZT, Huang PS, Chen GH, Xu J, Wu CX, Hu MJ, Xu J, Xu Y, Huang CR, Jin C, Lu XT, Qian HY, Li XD, Yang YJ. Interleukin-5-induced eosinophil population improves cardiac function after myocardial infarction. Cardiovasc Res 2021; 118:2165-2178. [PMID: 34259869 DOI: 10.1093/cvr/cvab237] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 07/08/2021] [Indexed: 12/16/2022] Open
Abstract
AIMS Interleukin (IL)-5 mediates the development of eosinophils (EOS) that are essential for tissue post-injury repair. It remains unknown whether IL-5 plays a role in heart repair after myocardial infarction (MI). This study aims to test whether IL-5-induced EOS population promotes the healing and repair process post-MI and to reveal the underlying mechanisms. METHOD AND RESULTS MI was induced by permanent ligation of the left anterior descending coronary artery in wild-type C57BL/6 mice. Western blot and real-time polymerase chain reaction revealed elevated expression of IL-5 in the heart at 5 days post-MI. Immunohistostaining indicated that IL-5 was secreted mainly from macrophages and type 2 innate lymphoid cells in the setting of experimental MI. External supply of recombinant mouse IL-5 (20 min, 1 day, and 2 days after MI surgery) reduced the infarct size and increased ejection fraction and angiogenesis in the border zone. A significant expansion of EOS was detected in both the peripheral blood and infarcted myocardium after IL-5 administration. Pharmacological depletion of EOS by TRFK5 pretreatment muted the beneficial effects of IL-5 in MI mice. Mechanistic studies demonstrated that IL-5 increased the accumulation of CD206+ macrophages in infarcted myocardium at 7 days post-MI. In vitro co-culture experiments showed that EOS shifted bone marrow-derived macrophage polarization towards the CD206+ phenotypes. This activity of EOS was abolished by IL-4 neutralizing antibody, but not IL-10 or IL-13 neutralization. Western blot analyses demonstrated that EOS promoted the macrophage downstream signal transducer and activator of transcription 6 (STAT6) phosphorylation. CONCLUSION IL-5 facilitates the recovery of cardiac dysfunction post-MI by promoting EOS accumulation and subsequent CD206+ macrophage polarization via the IL-4/STAT6 axis. TRANSLATIONAL PERSPECTIVE Accumulating evidence suggests that modulation of innate and adaptive immune responses is a promising therapeutic strategy for myocardial infarction. In this study, we demonstrate that IL-5 exerts cardioprotective effects on infarcted myocardium by promoting eosinophil accumulation and subsequent CD206+ macrophage polarization via the IL-4/STAT6 axis. Hence, regulation of cardiac IL-5 level or eosinophil count may become a therapeutic approach for post-myocardial infarction cardiac repair in humans.
Collapse
Affiliation(s)
- Jun-Yan Xu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No.167 Bei Li Shi Road, Xicheng District, Beijing, 100037, People's Republic of China.,Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yu-Yan Xiong
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No.167 Bei Li Shi Road, Xicheng District, Beijing, 100037, People's Republic of China
| | - Rui-Jie Tang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No.167 Bei Li Shi Road, Xicheng District, Beijing, 100037, People's Republic of China
| | - Wen-Yang Jiang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No.167 Bei Li Shi Road, Xicheng District, Beijing, 100037, People's Republic of China
| | - Yu Ning
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No.167 Bei Li Shi Road, Xicheng District, Beijing, 100037, People's Republic of China
| | - Zhao-Ting Gong
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No.167 Bei Li Shi Road, Xicheng District, Beijing, 100037, People's Republic of China
| | - Pei-Sen Huang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No.167 Bei Li Shi Road, Xicheng District, Beijing, 100037, People's Republic of China.,Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Key Laboratory on Assisted Circulation, Ministry of Health, Guangzhou, China
| | - Gui-Hao Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No.167 Bei Li Shi Road, Xicheng District, Beijing, 100037, People's Republic of China
| | - Jun Xu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No.167 Bei Li Shi Road, Xicheng District, Beijing, 100037, People's Republic of China
| | - Chun-Xiao Wu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No.167 Bei Li Shi Road, Xicheng District, Beijing, 100037, People's Republic of China
| | - Meng-Jin Hu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No.167 Bei Li Shi Road, Xicheng District, Beijing, 100037, People's Republic of China
| | - Jing Xu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No.167 Bei Li Shi Road, Xicheng District, Beijing, 100037, People's Republic of China
| | - Yi Xu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No.167 Bei Li Shi Road, Xicheng District, Beijing, 100037, People's Republic of China
| | - Cun-Rong Huang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No.167 Bei Li Shi Road, Xicheng District, Beijing, 100037, People's Republic of China
| | - Chen Jin
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No.167 Bei Li Shi Road, Xicheng District, Beijing, 100037, People's Republic of China
| | - Xiao-Tong Lu
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, People's Republic of China
| | - Hai-Yan Qian
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No.167 Bei Li Shi Road, Xicheng District, Beijing, 100037, People's Republic of China
| | - Xiang-Dong Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No.167 Bei Li Shi Road, Xicheng District, Beijing, 100037, People's Republic of China
| | - Yue-Jin Yang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No.167 Bei Li Shi Road, Xicheng District, Beijing, 100037, People's Republic of China
| |
Collapse
|
9
|
Ye D, Wang Z, Ye J, Wang M, Liu J, Xu Y, Jiang H, Chen J, Wan J. Los niveles de interleucina-5 disminuyen en plasma de pacientes con enfermedad coronaria e inhiben la diferenciación in vitro de Th1 y Th17. Rev Esp Cardiol 2020. [PMID: 31495774 DOI: 10.1016/j.recesp.2019.07.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
10
|
Aria H, Kalani M, Hodjati H, Doroudchi M. Different cytokine patterns induced by Helicobacter pylori and Lactobacillus acidophilus extracts in PBMCs of patients with abdominal aortic aneurysm. Comp Immunol Microbiol Infect Dis 2020; 70:101449. [PMID: 32126431 DOI: 10.1016/j.cimid.2020.101449] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 01/22/2020] [Accepted: 02/11/2020] [Indexed: 12/28/2022]
Abstract
Abdominal aortic aneurysm (AAA) is a degenerative inflammatory disease with unknown etiology. AAA is characterized by abdominal aortic dilatation more than 3 cm and is often asymptomatic, but the rupture of aneurysm can lead to death. Age, smoking and male sex are major predisposing factors of AAA. This study compares the effect of Helicobacter (H.) pylori and Lactobacillus (L.) acidophilus on the cytokine profile of PBMCs of 5 men with abdominal aortic aneurysm (AAA) and 5 men with normal/insignificant angiography, CT-Scan and ultrasonography results in the single-culture and in the co-culture with HUVECs. IL-2, IL-4, IL-5, IL-6, IL-9, IL-10, IL-13, IL-17A, IL-17 F, IL-21, IL-22, IFN-γ and TNF-α were measured in culture supernatants using a commercial fluorescent-labeled-bead assay. In general, CagA+ H. pylori-extract induced higher production of IFN-γ, IL-13 and IL-21 by PBMCs. Treatment of patients' PBMCs with CagA+H. pylori-extract induced Th2 cytokines while treatment of controls' PBMCs with CagA+H. pylori-extract increased Th1 cytokines. In the co-culture, however, patients' PBMCs produced Th1 cytokines irrespective of extract treatment, while controls' PBMCs produced Th2 cytokines and decreased IL-10. CagA+ H. pylori- as well as L. acidophilus-extract induced higher levels of IL-9 by controls' PBMCs in co-culture with HUVECs than patients (P = 0.05 and P = 0.01). The cytokine pattern of PBMCs induced by CagA+ H. pylori- and L. acidophilus-extracts in the co-culture with HUVECs shows differences in AAA patients and in comparison to controls. Decreased secretion of IL-9, IL-21 and IL-22 by PBMCs of patients treated with CagA+ H. pylori extract in co-culture, as opposed to non-AAA controls may indicate the active role ECs play in AAA. Simultaneous production of IL-10 and Th1 cytokines in patients and pronounced Th2 cytokines in controls in response to both bacteria may point to the inherent differences between patients and controls, which need further investigation.
Collapse
Affiliation(s)
- Hamid Aria
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehdi Kalani
- Prof. Alborzi Clinical Microbiology Research Center, Nemazee Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hossein Hodjati
- Department of Vascular Surgery, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehrnoosh Doroudchi
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
11
|
Peshkova IO, Aghayev T, Fatkhullina AR, Makhov P, Titerina EK, Eguchi S, Tan YF, Kossenkov AV, Khoreva MV, Gankovskaya LV, Sykes SM, Koltsova EK. IL-27 receptor-regulated stress myelopoiesis drives abdominal aortic aneurysm development. Nat Commun 2019; 10:5046. [PMID: 31695038 PMCID: PMC6834661 DOI: 10.1038/s41467-019-13017-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 10/15/2019] [Indexed: 02/07/2023] Open
Abstract
Abdominal aortic aneurysm (AAA) is a prevalent life-threatening disease, where aortic wall degradation is mediated by accumulated immune cells. Although cytokines regulate inflammation within the aorta, their contribution to AAA via distant alterations, particularly in the control of hematopoietic stem cell (HSC) differentiation, remains poorly defined. Here we report a pathogenic role for the interleukin-27 receptor (IL-27R) in AAA, as genetic ablation of IL-27R protects mice from the disease development. Mitigation of AAA is associated with a blunted accumulation of myeloid cells in the aorta due to the attenuation of Angiotensin II (Ang II)-induced HSC expansion. IL-27R signaling is required to induce transcriptional programming to overcome HSC quiescence and increase differentiation and output of mature myeloid cells in response to stress stimuli to promote their accumulation in the diseased aorta. Overall, our studies illuminate how a prominent vascular disease can be distantly driven by a cytokine-dependent regulation of bone marrow precursors.
Collapse
Affiliation(s)
- Iuliia O Peshkova
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania, 19111, USA
- Pirogov Russian National Research Medical University, Moscow, 117997, Russia
| | - Turan Aghayev
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania, 19111, USA
- Pirogov Russian National Research Medical University, Moscow, 117997, Russia
| | - Aliia R Fatkhullina
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania, 19111, USA
| | - Petr Makhov
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania, 19111, USA
| | - Elizaveta K Titerina
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania, 19111, USA
- Pirogov Russian National Research Medical University, Moscow, 117997, Russia
| | - Satoru Eguchi
- Lewis Katz School of Medicine, Temple University Cardiovascular Research Center, Philadelphia, Pennsylvania, 19140, USA
| | - Yin Fei Tan
- Genomics Facility, Fox Chase Cancer Center, Philadelphia, Pennsylvania, 19111, USA
| | - Andrew V Kossenkov
- Bioinformatics Facility, The Wistar Institute, Philadelphia, Pennsylvania, 19104, USA
| | - Marina V Khoreva
- Pirogov Russian National Research Medical University, Moscow, 117997, Russia
| | | | - Stephen M Sykes
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania, 19111, USA
| | - Ekaterina K Koltsova
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania, 19111, USA.
| |
Collapse
|
12
|
Ye D, Wang Z, Ye J, Wang M, Liu J, Xu Y, Jiang H, Chen J, Wan J. Interleukin-5 levels are decreased in the plasma of coronary artery disease patients and inhibit Th1 and Th17 differentiation in vitro. ACTA ACUST UNITED AC 2019; 73:393-402. [PMID: 31495774 DOI: 10.1016/j.rec.2019.07.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Accepted: 07/12/2019] [Indexed: 01/22/2023]
Abstract
INTRODUCTION AND OBJECTIVES Interleukin (IL)-5 is an anti-inflammatory cytokine that has been demonstrated to be involved in cardiovascular diseases, including aortic aneurysm and heart failure. This study aimed to investigate the involvement of IL-5 in coronary artery disease (CAD) and the possible mechanisms. METHODS We analyzed IL-5 expression in human coronary artery specimens collected from CAD patients and deceased donors. Plasma IL-5, IL-17, and interferon-γ levels in CAD patients were detected using ELISA kits, with samples from chest pain patients (non-CAD) as controls. Mouse CD4+T helper (Th) cells were separated, and the effect of IL-5 on Th1, regulatory T cell and Th17 differentiation and mRNA levels of their characteristic cytokines were detected using flow cytometry and reverse transcription-quantitative polymerase chain reaction, respectively. RESULTS IL-5 was significantly decreased in the coronary plaque of CAD patients compared with the deceased donors group, and IL-5 was mainly derived from macrophages in the coronary artery plaque. Compared with the non-CAD group, plasma IL-5 levels in the CAD groups were significantly lower, and the sequence from high to low was stable angina pectoris, unstable angina pectoris, and acute myocardial infarction. Binary linear regression analysis showed that IL-5 was independently correlated with the occurrence of CAD. Recombinant mouse IL-5 treatment decreased Th1 and Th17 levels and mRNA expression of their characteristic cytokines in oxidized low-density lipoprotein-treated CD4+Th cells. CONCLUSION IL-5 levels were decreased in CAD patients and inhibited oxidized low-density lipoprotein Th1 and Th17 differentiation in vitro.
Collapse
Affiliation(s)
- Di Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Zhen Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jing Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jianfang Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yao Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Huimin Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jiangbin Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China.
| |
Collapse
|
13
|
Wang WD, Sun R, Chen YX. PPARγ agonist rosiglitazone alters the temporal and spatial distribution of inflammation during abdominal aortic aneurysm formation. Mol Med Rep 2018; 18:3421-3428. [PMID: 30066924 DOI: 10.3892/mmr.2018.9311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 06/19/2018] [Indexed: 11/06/2022] Open
Abstract
Research into inflammation during abdominal aortic aneurysm (AAA) formation remains inconclusive. The present study aimed to demonstrate the temporal and spatial distribution of inflammatory cytokines, and to confirm the effect of peroxisome proliferator‑activated receptor γ (PPARγ) on the incidence of AAA formation and the distribution of inflammation in the disease process. Male apolipoprotein E‑/‑ mice were randomly divided into eight groups: Angiotensin II (Ang‑II)‑only 7, 14, 21, 28 and 42 days groups, Ang‑II with rosiglitazone (RGZ) 28 and 42 days groups, and the saline control 42 days group. The early stage was defined as between 7 and 21 days, and the late stage as between 28 and 42 days. Incidences of early rupture and late rupture, aneurysm formation and the maximum diameters of the aorta were recorded. Suprarenal abdominal aortic tissues were collected for histological analysis, and western blotting was performed to reveal the distribution of inflammation. Treatment with Ang‑II caused a significant dilation of the aorta in the late stage; however, this was not observed in the early stage. RGZ reduced the maximum diameters in the late stage. With the pathological process alterations, the inflammatory type shifted. Regarding temporal distribution, the tumor necrosis factor (TNF)‑α expression level was increased over time, and the interleukin (IL)‑10 expression level significantly decreased. When considering the spatial distribution, TNF‑α was expressed dominantly in the aneurysmal body and IL‑10 was dominant in the aneurysmal neck in the late stage. The PPARγ agonist RGZ may reduce the expression of TNF‑α in the late stage and increase the expression level of IL‑10, maintaining the TNF‑α or IL‑10 expression levels at the same levels as in the early stage. Aortic inflammation during AAA formation is dynamic. Protective anti‑inflammatory cytokines are upregulated in the early 'compensatory stage'; however, pro‑inflammatory cytokines are dominant in the late 'decompensatory stage'. PPARγ is likely to continue to upregulate the expression of anti‑inflammatory cytokines, extend the 'compensatory stage', and decelerate the process of AAA development and rupture.
Collapse
Affiliation(s)
- Wen-Da Wang
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Rui Sun
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Yue-Xin Chen
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| |
Collapse
|
14
|
van Puijvelde GHM, Foks AC, van Bochove RE, Bot I, Habets KLL, de Jager SC, ter Borg MND, van Osch P, Boon L, Vos M, de Waard V, Kuiper J. CD1d deficiency inhibits the development of abdominal aortic aneurysms in LDL receptor deficient mice. PLoS One 2018; 13:e0190962. [PMID: 29346401 PMCID: PMC5773169 DOI: 10.1371/journal.pone.0190962] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 12/22/2017] [Indexed: 11/19/2022] Open
Abstract
An abdominal aortic aneurysm (AAA) is a dilatation of the abdominal aorta leading to serious complications and mostly to death. AAA development is associated with an accumulation of inflammatory cells in the aorta including NKT cells. An important factor in promoting the recruitment of these inflammatory cells into tissues and thereby contributing to the development of AAA is angiotensin II (Ang II). We demonstrate that a deficiency in CD1d dependent NKT cells under hyperlipidemic conditions (LDLr-/-CD1d-/- mice) results in a strong decline in the severity of angiotensin II induced aneurysm formation when compared with LDLr-/- mice. In addition, we show that Ang II amplifies the activation of NKT cells both in vivo and in vitro. We also provide evidence that type I NKT cells contribute to AAA development by inducing the expression of matrix degrading enzymes in vSMCs and macrophages, and by cytokine dependently decreasing vSMC viability. Altogether, these data prove that CD1d-dependent NKT cells contribute to AAA development in the Ang II-mediated aneurysm model by enhancing aortic degradation, establishing that therapeutic applications which target NKT cells can be a successful way to prevent AAA development.
Collapse
Affiliation(s)
- Gijs H. M. van Puijvelde
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
- * E-mail:
| | - Amanda C. Foks
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Rosemarie E. van Bochove
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Ilze Bot
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Kim L. L. Habets
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Saskia C. de Jager
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Mariëtte N. D. ter Borg
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Puck van Osch
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | | | - Mariska Vos
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Vivian de Waard
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Johan Kuiper
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| |
Collapse
|
15
|
Ohno T, Aoki H, Ohno S, Nishihara M, Furusho A, Hiromatsu S, Akashi H, Fukumoto Y, Tanaka H. Cytokine Profile of Human Abdominal Aortic Aneurysm: Involvement of JAK/STAT Pathway. Ann Vasc Dis 2018; 11:84-90. [PMID: 29682112 PMCID: PMC5882349 DOI: 10.3400/avd.oa.17-00086] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Objective: Abdominal aortic aneurysm (AAA) is characterized by inflammation and destruction of normal tissue architecture. The present study aimed to evaluate the inflammatory signaling cascade by analyzing the cytokines of AAA tissue. Materials and Methods: We analyzed the comprehensive cytokine secretion profiles of 52 cytokines from human AAA in four patients with AAA using fluorescent beads-based multiplex assay. Further, the effect of janus kinase (JAK) inhibition by pyridone 6 on cytokine profiles was also evaluated. Results: Cytokine secretion profiles were found to be similar among the four patients. A high level of JAK/signal transducers and activator of transcription (STAT) pathway activity in AAA tissue in culture was maintained, which may be attributed to the secretion of endogenous JAK-activating cytokines. Inhibition of JAK by pyridone 6 resulted in the suppression of STAT3 phosphorylation and secretion of a subset of chemokines and JAK-activating cytokines. However, the inhibition of JAK had no effect on the secretion of matrix metalloproteinase (MMP)-2, MMP-9, or TGF-β family that is responsible for the metabolism of extracellular matrix. Conclusion: The findings of the present study suggested that AAA tissue exhibits a stereotypical profile of cytokine secretion, where JAK/STAT pathway may play a role in regulating a subset of cytokines. Identification of such a cytokine profile may reveal potential diagnostic markers and therapeutic targets for AAA.
Collapse
Affiliation(s)
- Tomokazu Ohno
- Division of Cardiovascular Surgery, Department of Surgery, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Hiroki Aoki
- Cardiovascular Research Institute, Kurume University, Kurume, Fukuoka, Japan
| | - Satoko Ohno
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Michihide Nishihara
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Aya Furusho
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Shinichi Hiromatsu
- Division of Cardiovascular Surgery, Department of Surgery, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Hidetoshi Akashi
- Division of Cardiovascular Surgery, Department of Surgery, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Yoshihiro Fukumoto
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Hiroyuki Tanaka
- Division of Cardiovascular Surgery, Department of Surgery, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| |
Collapse
|
16
|
Differentially expressed genes and canonical pathways in the ascending thoracic aortic aneurysm - The Tampere Vascular Study. Sci Rep 2017; 7:12127. [PMID: 28935963 PMCID: PMC5608723 DOI: 10.1038/s41598-017-12421-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 09/07/2017] [Indexed: 12/14/2022] Open
Abstract
Ascending thoracic aortic aneurysm (ATAA) is a multifactorial disease with a strong inflammatory component. Surgery is often required to prevent aortic rupture and dissection. We performed gene expression analysis (Illumina HumanHT-12 version 3 Expression BeadChip) for 32 samples from ATAA (26 without/6 with dissection), and 28 left internal thoracic arteries (controls) collected in Tampere Vascular study. We compared expression profiles and conducted pathway analysis using Ingenuity Pathway Analysis (IPA) to reveal differences between ATAA and a healthy artery wall. Almost 5000 genes were differentially expressed in ATAA samples compared to controls. The most downregulated gene was homeobox (HOX) A5 (fold change, FC = -25.3) and upregulated cadherin-2 (FC = 12.6). Several other HOX genes were also found downregulated (FCs between -25.3 and -1.5, FDR < 0.05). 43, mostly inflammatory, canonical pathways in ATAA were found to be significantly (p < 0.05, FDR < 0.05) differentially expressed. The results remained essentially the same when the 6 dissected ATAA samples were excluded from the analysis. We show for the first time on genome level that ATAA is an inflammatory process, revealing a more detailed molecular pathway level pathogenesis. We propose HOX genes as potentially important players in maintaining aortic integrity, altered expression of which might be important in the pathobiology of ATAA.
Collapse
|
17
|
Shen YH, LeMaire SA. Molecular pathogenesis of genetic and sporadic aortic aneurysms and dissections. Curr Probl Surg 2017; 54:95-155. [PMID: 28521856 DOI: 10.1067/j.cpsurg.2017.01.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 01/16/2017] [Indexed: 12/20/2022]
Affiliation(s)
- Ying H Shen
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX; Department of Cardiovascular Surgery, Texas Heart Institute, Houston, TX; Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX.
| | - Scott A LeMaire
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX; Department of Cardiovascular Surgery, Texas Heart Institute, Houston, TX; Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX; Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX.
| |
Collapse
|
18
|
Systemic Administration of Proteoglycan Protects BALB/c Retired Breeder Mice from Experimental Arthritis. J Immunol Res 2016; 2016:6765134. [PMID: 27294161 PMCID: PMC4887641 DOI: 10.1155/2016/6765134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 04/17/2016] [Accepted: 04/20/2016] [Indexed: 11/27/2022] Open
Abstract
This study was undertaken to evaluate the prophylactic potential of proteoglycan (PG) administration in experimental arthritis. Female BALB/c retired breeder mice received two (2xPG50 and 2xPG100 groups) or three (3xPG50 group) intraperitoneal doses of bovine PG (50 μg or 100 μg) every three days. A week later the animals were submitted to arthritis induction by immunization with three i.p. doses of bovine PG associated with dimethyldioctadecylammonium bromide adjuvant at intervals of 21 days. Disease severity was daily assessed after the third dose by score evaluation. The 3xPG50 group showed significant reduction in prevalence and clinical scores. This protective effect was associated with lower production of IFN-γ and IL-17 and increased production of IL-5 and IL-10 by spleen cells restimulated in vitro with PG. Even though previous PG administration restrained dendritic cells maturation this procedure did not alter the frequency of regulatory Foxp3+ T cells. Lower TNF-α and IL-6 levels and higher expression of ROR-γ and GATA-3 were detected in the paws of protected animals. A delayed-type hypersensitivity reaction confirmed specific tolerance induction. Taken together, these results indicate that previous PG inoculation determines a specific tolerogenic effect that is able to decrease severity of subsequently induced arthritis.
Collapse
|
19
|
Li X, Cao X, Zhang X, Kang Y, Zhang W, Yu M, Ma C, Han J, Duan Y, Chen Y. MEK1/2 inhibitors induce interleukin-5 expression in mouse macrophages and lymphocytes. Biochem Biophys Res Commun 2016; 473:939-946. [PMID: 27045084 DOI: 10.1016/j.bbrc.2016.03.156] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 03/31/2016] [Indexed: 01/27/2023]
Abstract
Uptake of oxidized low-density lipoprotein (oxLDL) by macrophages facilitates the formation of foam cells, the prominent part of atherosclerotic lesions. Interleukin-5 (IL-5) is a cytokine regulating interactions between immune cells. It also activates the production of T15/EO6 IgM antibodies in B-1 cells, which can bind oxLDL thereby demonstrating anti-atherogenic properties. We previously reported that inhibition of extracellular signal-regulated kinases 1 and 2 (ERK1/2) by mitogen-activated protein kinase kinases 1/2 (MEK1/2) inhibitors can reduce atherosclerosis. In this study, we determined the effects of MEK1/2 inhibitors on IL-5 production both in vitro and in vivo. In vitro, MEK1/2 inhibitors (PD98059 and U0126) substantially inhibited phosphorylation of MEK1/2 and ERK1/2. Associated with inhibition of ERK1/2 phosphorylation both in vitro and in vivo, MEK1/2 inhibitors induced IL-5 protein expression in macrophages (RAW macrophages and peritoneal macrophages) and lymphocytes (EL-4 cells). In vivo, administration of mice with MEK1/2 inhibitors increased serum IL-5 levels, and IL-5 protein expression in mouse spleen and liver. At the mechanistic level, we determined that MEK1/2 inhibitors activated IL-5 mRNA expression and IL-5 promoter activity in the liver X receptor (LXR) dependent manner indicating the induction of IL-5 transcription. In addition, we determined that MEK1/2 inhibitors enhanced IL-5 protein stability. Taken together, our study demonstrates that MEK1/2 inhibitors induce IL-5 production which suggests another anti-atherogenic mechanism of MEK1/2 inhibitors.
Collapse
Affiliation(s)
- Xiaoju Li
- College of Life Sciences, Nankai University, Tianjin, China
| | - Xingyue Cao
- College of Life Sciences, Nankai University, Tianjin, China
| | - Xiaomeng Zhang
- College of Life Sciences, Nankai University, Tianjin, China
| | - Yanhua Kang
- School of Medicine, Hangzhou Normal University, Hangzhou, China
| | - Wenwen Zhang
- College of Life Sciences, Nankai University, Tianjin, China
| | - Miao Yu
- College of Life Sciences, Nankai University, Tianjin, China
| | - Chuanrui Ma
- College of Life Sciences, Nankai University, Tianjin, China
| | - Jihong Han
- College of Medical Engineering, Hefei University of Technology, Hefei, China; College of Life Sciences, Nankai University, Tianjin, China
| | - Yajun Duan
- College of Medical Engineering, Hefei University of Technology, Hefei, China; College of Life Sciences, Nankai University, Tianjin, China.
| | - Yuanli Chen
- College of Medical Engineering, Hefei University of Technology, Hefei, China; School of Medicine, Nankai University, Tianjin, China.
| |
Collapse
|
20
|
Peshkova IO, Schaefer G, Koltsova EK. Atherosclerosis and aortic aneurysm – is inflammation a common denominator? FEBS J 2016; 283:1636-52. [DOI: 10.1111/febs.13634] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 11/20/2015] [Accepted: 12/18/2015] [Indexed: 12/30/2022]
Affiliation(s)
- Iuliia O. Peshkova
- Blood Cell Development and Function Program Fox Chase Cancer Center Philadephia PA USA
| | - Giulia Schaefer
- Blood Cell Development and Function Program Fox Chase Cancer Center Philadephia PA USA
| | - Ekaterina K. Koltsova
- Blood Cell Development and Function Program Fox Chase Cancer Center Philadephia PA USA
| |
Collapse
|
21
|
Liu CL, Wang Y, Liao M, Wemmelund H, Ren J, Fernandes C, Zhou Y, Sukhova GK, Lindholt JS, Johnsen SP, Zhang JY, Cheng X, Huang X, Daugherty A, Levy BD, Libby P, Shi GP. Allergic Lung Inflammation Aggravates Angiotensin II-Induced Abdominal Aortic Aneurysms in Mice. Arterioscler Thromb Vasc Biol 2015; 36:69-77. [PMID: 26543094 DOI: 10.1161/atvbaha.115.305911] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 10/14/2015] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Asthma and abdominal aortic aneurysms (AAA) both involve inflammation. Patients with asthma have an increased risk of developing AAA or experiencing aortic rupture. This study tests the development of one disease on the progression of the other. APPROACH AND RESULTS Ovalbumin sensitization and challenge in mice led to the development of allergic lung inflammation (ALI). Subcutaneous infusion of angiotensin II into mice produced AAA. Simultaneous production of ALI in AAA mice doubled abdominal aortic diameter and increased macrophage and mast cell content, arterial media smooth muscle cell loss, cell proliferation, and angiogenesis in AAA lesions. ALI also increased plasma IgE, reduced plasma interleukin-5, and increased bronchioalveolar total inflammatory cell and eosinophil accumulation. Intraperitoneal administration of an anti-IgE antibody suppressed AAA lesion formation and reduced lesion inflammation, plasma IgE, and bronchioalveolar inflammation. Pre-establishment of ALI also increased AAA lesion size, lesion accumulation of macrophages and mast cells, media smooth muscle cell loss, and plasma IgE, reduced plasma interleukin-5, interleukin-13, and transforming growth factor-β, and increased bronchioalveolar inflammation. Consequent production of ALI also doubled lesion size of pre-established AAA and increased lesion mast cell and T-cell accumulation, media smooth muscle cell loss, lesion cell proliferation and apoptosis, plasma IgE, and bronchioalveolar inflammation. In periaortic CaCl2 injury-induced AAA in mice, production of ALI also increased AAA formation, lesion inflammation, plasma IgE, and bronchioalveolar inflammatory cell accumulation. CONCLUSIONS This study suggests a pathological link between airway allergic disease and AAA. Production of one disease aggravates the progression of the other.
Collapse
Affiliation(s)
- Cong-Lin Liu
- From the Department of Cardiology, Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (C.-L.L., J.-Y.Z., G.-P.S.); Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (C.-L.L., Y.W., M.L., J.R., C.F., Y.Z., G.K.S., B.D.L., P.L., G.-P.S.); Department of Cardiology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Y.W.); Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China (M.L., X.C.); Department of Vascular Surgery, Viborg Regional Hospital, Viborg, Denmark (H.W.); Department of Hypertension, Beijing Anzhen Hospital, Capital Medical University, Beijing, China (J.R.); Department of Clinical Epidemiology, Institute of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark (H.W., S.P.J.); Department of Cardiothoracic and Vascular Surgery, Elitary Research Centre of Individualized Medicine of Arterial Disease, Odense University Hospital, Odense, Denmark (J.S.L.); Department of Medicine, University of California, San Francisco (X.H.); and Saha Cardiovascular Research Center, University of Kentucky, Lexington (A.D.)
| | - Yi Wang
- From the Department of Cardiology, Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (C.-L.L., J.-Y.Z., G.-P.S.); Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (C.-L.L., Y.W., M.L., J.R., C.F., Y.Z., G.K.S., B.D.L., P.L., G.-P.S.); Department of Cardiology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Y.W.); Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China (M.L., X.C.); Department of Vascular Surgery, Viborg Regional Hospital, Viborg, Denmark (H.W.); Department of Hypertension, Beijing Anzhen Hospital, Capital Medical University, Beijing, China (J.R.); Department of Clinical Epidemiology, Institute of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark (H.W., S.P.J.); Department of Cardiothoracic and Vascular Surgery, Elitary Research Centre of Individualized Medicine of Arterial Disease, Odense University Hospital, Odense, Denmark (J.S.L.); Department of Medicine, University of California, San Francisco (X.H.); and Saha Cardiovascular Research Center, University of Kentucky, Lexington (A.D.)
| | - Mengyang Liao
- From the Department of Cardiology, Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (C.-L.L., J.-Y.Z., G.-P.S.); Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (C.-L.L., Y.W., M.L., J.R., C.F., Y.Z., G.K.S., B.D.L., P.L., G.-P.S.); Department of Cardiology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Y.W.); Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China (M.L., X.C.); Department of Vascular Surgery, Viborg Regional Hospital, Viborg, Denmark (H.W.); Department of Hypertension, Beijing Anzhen Hospital, Capital Medical University, Beijing, China (J.R.); Department of Clinical Epidemiology, Institute of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark (H.W., S.P.J.); Department of Cardiothoracic and Vascular Surgery, Elitary Research Centre of Individualized Medicine of Arterial Disease, Odense University Hospital, Odense, Denmark (J.S.L.); Department of Medicine, University of California, San Francisco (X.H.); and Saha Cardiovascular Research Center, University of Kentucky, Lexington (A.D.)
| | - Holger Wemmelund
- From the Department of Cardiology, Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (C.-L.L., J.-Y.Z., G.-P.S.); Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (C.-L.L., Y.W., M.L., J.R., C.F., Y.Z., G.K.S., B.D.L., P.L., G.-P.S.); Department of Cardiology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Y.W.); Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China (M.L., X.C.); Department of Vascular Surgery, Viborg Regional Hospital, Viborg, Denmark (H.W.); Department of Hypertension, Beijing Anzhen Hospital, Capital Medical University, Beijing, China (J.R.); Department of Clinical Epidemiology, Institute of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark (H.W., S.P.J.); Department of Cardiothoracic and Vascular Surgery, Elitary Research Centre of Individualized Medicine of Arterial Disease, Odense University Hospital, Odense, Denmark (J.S.L.); Department of Medicine, University of California, San Francisco (X.H.); and Saha Cardiovascular Research Center, University of Kentucky, Lexington (A.D.)
| | - Jingyuan Ren
- From the Department of Cardiology, Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (C.-L.L., J.-Y.Z., G.-P.S.); Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (C.-L.L., Y.W., M.L., J.R., C.F., Y.Z., G.K.S., B.D.L., P.L., G.-P.S.); Department of Cardiology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Y.W.); Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China (M.L., X.C.); Department of Vascular Surgery, Viborg Regional Hospital, Viborg, Denmark (H.W.); Department of Hypertension, Beijing Anzhen Hospital, Capital Medical University, Beijing, China (J.R.); Department of Clinical Epidemiology, Institute of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark (H.W., S.P.J.); Department of Cardiothoracic and Vascular Surgery, Elitary Research Centre of Individualized Medicine of Arterial Disease, Odense University Hospital, Odense, Denmark (J.S.L.); Department of Medicine, University of California, San Francisco (X.H.); and Saha Cardiovascular Research Center, University of Kentucky, Lexington (A.D.)
| | - Cleverson Fernandes
- From the Department of Cardiology, Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (C.-L.L., J.-Y.Z., G.-P.S.); Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (C.-L.L., Y.W., M.L., J.R., C.F., Y.Z., G.K.S., B.D.L., P.L., G.-P.S.); Department of Cardiology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Y.W.); Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China (M.L., X.C.); Department of Vascular Surgery, Viborg Regional Hospital, Viborg, Denmark (H.W.); Department of Hypertension, Beijing Anzhen Hospital, Capital Medical University, Beijing, China (J.R.); Department of Clinical Epidemiology, Institute of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark (H.W., S.P.J.); Department of Cardiothoracic and Vascular Surgery, Elitary Research Centre of Individualized Medicine of Arterial Disease, Odense University Hospital, Odense, Denmark (J.S.L.); Department of Medicine, University of California, San Francisco (X.H.); and Saha Cardiovascular Research Center, University of Kentucky, Lexington (A.D.)
| | - Yi Zhou
- From the Department of Cardiology, Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (C.-L.L., J.-Y.Z., G.-P.S.); Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (C.-L.L., Y.W., M.L., J.R., C.F., Y.Z., G.K.S., B.D.L., P.L., G.-P.S.); Department of Cardiology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Y.W.); Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China (M.L., X.C.); Department of Vascular Surgery, Viborg Regional Hospital, Viborg, Denmark (H.W.); Department of Hypertension, Beijing Anzhen Hospital, Capital Medical University, Beijing, China (J.R.); Department of Clinical Epidemiology, Institute of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark (H.W., S.P.J.); Department of Cardiothoracic and Vascular Surgery, Elitary Research Centre of Individualized Medicine of Arterial Disease, Odense University Hospital, Odense, Denmark (J.S.L.); Department of Medicine, University of California, San Francisco (X.H.); and Saha Cardiovascular Research Center, University of Kentucky, Lexington (A.D.)
| | - Galina K Sukhova
- From the Department of Cardiology, Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (C.-L.L., J.-Y.Z., G.-P.S.); Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (C.-L.L., Y.W., M.L., J.R., C.F., Y.Z., G.K.S., B.D.L., P.L., G.-P.S.); Department of Cardiology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Y.W.); Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China (M.L., X.C.); Department of Vascular Surgery, Viborg Regional Hospital, Viborg, Denmark (H.W.); Department of Hypertension, Beijing Anzhen Hospital, Capital Medical University, Beijing, China (J.R.); Department of Clinical Epidemiology, Institute of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark (H.W., S.P.J.); Department of Cardiothoracic and Vascular Surgery, Elitary Research Centre of Individualized Medicine of Arterial Disease, Odense University Hospital, Odense, Denmark (J.S.L.); Department of Medicine, University of California, San Francisco (X.H.); and Saha Cardiovascular Research Center, University of Kentucky, Lexington (A.D.)
| | - Jes S Lindholt
- From the Department of Cardiology, Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (C.-L.L., J.-Y.Z., G.-P.S.); Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (C.-L.L., Y.W., M.L., J.R., C.F., Y.Z., G.K.S., B.D.L., P.L., G.-P.S.); Department of Cardiology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Y.W.); Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China (M.L., X.C.); Department of Vascular Surgery, Viborg Regional Hospital, Viborg, Denmark (H.W.); Department of Hypertension, Beijing Anzhen Hospital, Capital Medical University, Beijing, China (J.R.); Department of Clinical Epidemiology, Institute of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark (H.W., S.P.J.); Department of Cardiothoracic and Vascular Surgery, Elitary Research Centre of Individualized Medicine of Arterial Disease, Odense University Hospital, Odense, Denmark (J.S.L.); Department of Medicine, University of California, San Francisco (X.H.); and Saha Cardiovascular Research Center, University of Kentucky, Lexington (A.D.)
| | - Søren P Johnsen
- From the Department of Cardiology, Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (C.-L.L., J.-Y.Z., G.-P.S.); Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (C.-L.L., Y.W., M.L., J.R., C.F., Y.Z., G.K.S., B.D.L., P.L., G.-P.S.); Department of Cardiology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Y.W.); Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China (M.L., X.C.); Department of Vascular Surgery, Viborg Regional Hospital, Viborg, Denmark (H.W.); Department of Hypertension, Beijing Anzhen Hospital, Capital Medical University, Beijing, China (J.R.); Department of Clinical Epidemiology, Institute of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark (H.W., S.P.J.); Department of Cardiothoracic and Vascular Surgery, Elitary Research Centre of Individualized Medicine of Arterial Disease, Odense University Hospital, Odense, Denmark (J.S.L.); Department of Medicine, University of California, San Francisco (X.H.); and Saha Cardiovascular Research Center, University of Kentucky, Lexington (A.D.)
| | - Jin-Ying Zhang
- From the Department of Cardiology, Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (C.-L.L., J.-Y.Z., G.-P.S.); Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (C.-L.L., Y.W., M.L., J.R., C.F., Y.Z., G.K.S., B.D.L., P.L., G.-P.S.); Department of Cardiology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Y.W.); Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China (M.L., X.C.); Department of Vascular Surgery, Viborg Regional Hospital, Viborg, Denmark (H.W.); Department of Hypertension, Beijing Anzhen Hospital, Capital Medical University, Beijing, China (J.R.); Department of Clinical Epidemiology, Institute of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark (H.W., S.P.J.); Department of Cardiothoracic and Vascular Surgery, Elitary Research Centre of Individualized Medicine of Arterial Disease, Odense University Hospital, Odense, Denmark (J.S.L.); Department of Medicine, University of California, San Francisco (X.H.); and Saha Cardiovascular Research Center, University of Kentucky, Lexington (A.D.)
| | - Xiang Cheng
- From the Department of Cardiology, Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (C.-L.L., J.-Y.Z., G.-P.S.); Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (C.-L.L., Y.W., M.L., J.R., C.F., Y.Z., G.K.S., B.D.L., P.L., G.-P.S.); Department of Cardiology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Y.W.); Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China (M.L., X.C.); Department of Vascular Surgery, Viborg Regional Hospital, Viborg, Denmark (H.W.); Department of Hypertension, Beijing Anzhen Hospital, Capital Medical University, Beijing, China (J.R.); Department of Clinical Epidemiology, Institute of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark (H.W., S.P.J.); Department of Cardiothoracic and Vascular Surgery, Elitary Research Centre of Individualized Medicine of Arterial Disease, Odense University Hospital, Odense, Denmark (J.S.L.); Department of Medicine, University of California, San Francisco (X.H.); and Saha Cardiovascular Research Center, University of Kentucky, Lexington (A.D.)
| | - Xiaozhu Huang
- From the Department of Cardiology, Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (C.-L.L., J.-Y.Z., G.-P.S.); Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (C.-L.L., Y.W., M.L., J.R., C.F., Y.Z., G.K.S., B.D.L., P.L., G.-P.S.); Department of Cardiology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Y.W.); Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China (M.L., X.C.); Department of Vascular Surgery, Viborg Regional Hospital, Viborg, Denmark (H.W.); Department of Hypertension, Beijing Anzhen Hospital, Capital Medical University, Beijing, China (J.R.); Department of Clinical Epidemiology, Institute of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark (H.W., S.P.J.); Department of Cardiothoracic and Vascular Surgery, Elitary Research Centre of Individualized Medicine of Arterial Disease, Odense University Hospital, Odense, Denmark (J.S.L.); Department of Medicine, University of California, San Francisco (X.H.); and Saha Cardiovascular Research Center, University of Kentucky, Lexington (A.D.)
| | - Alan Daugherty
- From the Department of Cardiology, Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (C.-L.L., J.-Y.Z., G.-P.S.); Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (C.-L.L., Y.W., M.L., J.R., C.F., Y.Z., G.K.S., B.D.L., P.L., G.-P.S.); Department of Cardiology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Y.W.); Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China (M.L., X.C.); Department of Vascular Surgery, Viborg Regional Hospital, Viborg, Denmark (H.W.); Department of Hypertension, Beijing Anzhen Hospital, Capital Medical University, Beijing, China (J.R.); Department of Clinical Epidemiology, Institute of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark (H.W., S.P.J.); Department of Cardiothoracic and Vascular Surgery, Elitary Research Centre of Individualized Medicine of Arterial Disease, Odense University Hospital, Odense, Denmark (J.S.L.); Department of Medicine, University of California, San Francisco (X.H.); and Saha Cardiovascular Research Center, University of Kentucky, Lexington (A.D.)
| | - Bruce D Levy
- From the Department of Cardiology, Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (C.-L.L., J.-Y.Z., G.-P.S.); Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (C.-L.L., Y.W., M.L., J.R., C.F., Y.Z., G.K.S., B.D.L., P.L., G.-P.S.); Department of Cardiology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Y.W.); Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China (M.L., X.C.); Department of Vascular Surgery, Viborg Regional Hospital, Viborg, Denmark (H.W.); Department of Hypertension, Beijing Anzhen Hospital, Capital Medical University, Beijing, China (J.R.); Department of Clinical Epidemiology, Institute of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark (H.W., S.P.J.); Department of Cardiothoracic and Vascular Surgery, Elitary Research Centre of Individualized Medicine of Arterial Disease, Odense University Hospital, Odense, Denmark (J.S.L.); Department of Medicine, University of California, San Francisco (X.H.); and Saha Cardiovascular Research Center, University of Kentucky, Lexington (A.D.)
| | - Peter Libby
- From the Department of Cardiology, Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (C.-L.L., J.-Y.Z., G.-P.S.); Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (C.-L.L., Y.W., M.L., J.R., C.F., Y.Z., G.K.S., B.D.L., P.L., G.-P.S.); Department of Cardiology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Y.W.); Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China (M.L., X.C.); Department of Vascular Surgery, Viborg Regional Hospital, Viborg, Denmark (H.W.); Department of Hypertension, Beijing Anzhen Hospital, Capital Medical University, Beijing, China (J.R.); Department of Clinical Epidemiology, Institute of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark (H.W., S.P.J.); Department of Cardiothoracic and Vascular Surgery, Elitary Research Centre of Individualized Medicine of Arterial Disease, Odense University Hospital, Odense, Denmark (J.S.L.); Department of Medicine, University of California, San Francisco (X.H.); and Saha Cardiovascular Research Center, University of Kentucky, Lexington (A.D.)
| | - Guo-Ping Shi
- From the Department of Cardiology, Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (C.-L.L., J.-Y.Z., G.-P.S.); Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (C.-L.L., Y.W., M.L., J.R., C.F., Y.Z., G.K.S., B.D.L., P.L., G.-P.S.); Department of Cardiology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Y.W.); Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China (M.L., X.C.); Department of Vascular Surgery, Viborg Regional Hospital, Viborg, Denmark (H.W.); Department of Hypertension, Beijing Anzhen Hospital, Capital Medical University, Beijing, China (J.R.); Department of Clinical Epidemiology, Institute of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark (H.W., S.P.J.); Department of Cardiothoracic and Vascular Surgery, Elitary Research Centre of Individualized Medicine of Arterial Disease, Odense University Hospital, Odense, Denmark (J.S.L.); Department of Medicine, University of California, San Francisco (X.H.); and Saha Cardiovascular Research Center, University of Kentucky, Lexington (A.D.).
| |
Collapse
|
22
|
Dale MA, Ruhlman MK, Baxter BT. Inflammatory cell phenotypes in AAAs: their role and potential as targets for therapy. Arterioscler Thromb Vasc Biol 2015; 35:1746-55. [PMID: 26044582 DOI: 10.1161/atvbaha.115.305269] [Citation(s) in RCA: 173] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 05/11/2015] [Indexed: 02/06/2023]
Abstract
Abdominal aortic aneurysms (AAAs) are characterized by chronic inflammatory cell infiltration. AAA is typically an asymptomatic disease and caused ≈15 000 deaths annually in the United States. Previous studies have examined both human and murine aortic tissue for the presence of various inflammatory cell types. Studies show that in both human and experimental AAAs, prominent inflammatory cell infiltration, such as CD4(+) T cells and macrophages, occurs in the damaged aortic wall. These cells have the ability to undergo phenotypic modulation based on microenvironmental cues, potentially influencing disease progression. Proinflammatory CD4(+) T cells and classically activated macrophages dominate the landscape of aortic infiltrates. The skew to proinflammatory phenotypes alters disease progression and plays a role in causing chronic inflammation. The local cytokine production and presence of inflammatory mediators, such as extracellular matrix breakdown products, influence the uneven balance of the inflammatory infiltrate phenotypes. Understanding and developing new strategies that target the proinflammatory phenotype could provide useful therapeutic targets for a disease with no current pharmacological intervention.
Collapse
Affiliation(s)
- Matthew A Dale
- From the Department of Pathology and Microbiology (M.A.D.) and Department of Surgery, University of Nebraska Medical Center, Omaha (M.A.D., M.K.R., B.T.B.)
| | - Melissa K Ruhlman
- From the Department of Pathology and Microbiology (M.A.D.) and Department of Surgery, University of Nebraska Medical Center, Omaha (M.A.D., M.K.R., B.T.B.)
| | - B Timothy Baxter
- From the Department of Pathology and Microbiology (M.A.D.) and Department of Surgery, University of Nebraska Medical Center, Omaha (M.A.D., M.K.R., B.T.B.)
| |
Collapse
|
23
|
Carbone F, Montecucco F. Inflammation in arterial diseases. IUBMB Life 2015; 67:18-28. [PMID: 25631520 DOI: 10.1002/iub.1344] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 12/28/2014] [Indexed: 12/26/2022]
Affiliation(s)
- Federico Carbone
- First Clinic of Internal Medicine; Department of Internal Medicine; University of Genoa School of Medicine, IRCCS Azienda Ospedaliera Universitaria San Martino-IST Istituto Nazionale per la Ricerca sul Cancro; Genoa Italy
- Division of Cardiology; Foundation for Medical Researches; Department of Medical Specialties; University of Geneva; Geneva Switzerland
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine; Department of Internal Medicine; University of Genoa School of Medicine, IRCCS Azienda Ospedaliera Universitaria San Martino-IST Istituto Nazionale per la Ricerca sul Cancro; Genoa Italy
- Division of Cardiology; Foundation for Medical Researches; Department of Medical Specialties; University of Geneva; Geneva Switzerland
- Division of Laboratory Medicine; Department of Genetics and Laboratory Medicine; Geneva University Hospitals; Geneva Switzerland
| |
Collapse
|
24
|
Swaminathan G, Gadepalli VS, Stoilov I, Mecham RP, Rao RR, Ramamurthi A. Pro-elastogenic effects of bone marrow mesenchymal stem cell-derived smooth muscle cells on cultured aneurysmal smooth muscle cells. J Tissue Eng Regen Med 2014; 11:679-693. [DOI: 10.1002/term.1964] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Revised: 08/18/2014] [Accepted: 09/25/2014] [Indexed: 01/09/2023]
Affiliation(s)
- Ganesh Swaminathan
- Department of Biomedical Engineering; Cleveland Clinic; Cleveland OH USA
- Department of Biology; University of Akron; Akron OH USA
| | - Venkat S. Gadepalli
- Department of Chemical and Life Science Engineering; Virginia Commonwealth University; Richmond VA USA
| | - Ivan Stoilov
- Department of Cell Biology and Physiology; Washington University; St. Louis MO USA
| | - Robert P. Mecham
- Department of Cell Biology and Physiology; Washington University; St. Louis MO USA
| | - Raj R. Rao
- Department of Chemical and Life Science Engineering; Virginia Commonwealth University; Richmond VA USA
| | - Anand Ramamurthi
- Department of Biomedical Engineering; Cleveland Clinic; Cleveland OH USA
- Department of Biology; University of Akron; Akron OH USA
| |
Collapse
|
25
|
T lymphocytes and aortic aneurysms. SCIENCE CHINA-LIFE SCIENCES 2014; 57:795-801. [DOI: 10.1007/s11427-014-4699-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 06/19/2014] [Indexed: 11/27/2022]
|
26
|
Ma Y, Tan G, Zhao Z, Li W, Huang L, Liu G. Therapeutic effectiveness of endoscopic vidian neurectomy for the treatment of vasomotor rhinitis. Acta Otolaryngol 2014; 134:260-7. [PMID: 24433056 DOI: 10.3109/00016489.2013.831478] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
CONCLUSION Our results indicate that vidian neurectomy may be recommended as an effective method for the treatment of vasomotor rhinitis (VMR). OBJECTIVE The aim of this work was to study the feasibility and effectiveness of vidian neurectomy treatment under the nasal endoscope for VMR. METHODS The study included 45 patients with VMR. They were all assigned to functional endoscopic surgery with vidian neurectomy. RESULTS The obtained data showed that, using the rhinoconjunctivitis quality of life questionnaire, vidian neurectomy treatment relieved the symptoms of VMR in 82.2% of the patients. Vidian neurectomy also led to the reduction of expression of several cytokines, including vasoactive intestinal polypeptide, calcitonin gene-related peptide, substance P, interleukin (IL)-4, and IL-5.
Collapse
|
27
|
Jeon YJ, Kim JH, Lee BE, Rah H, Shin JE, Kang H, Choi DH, Yoon TK, Lee WS, Shim SH, Kim NK. Association between Polymorphisms in the Renin-Angiotensin System Genes and Prevalence of Spontaneously Aborted Fetuses. Am J Reprod Immunol 2013; 70:238-45. [DOI: 10.1111/aji.12110] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Accepted: 02/06/2013] [Indexed: 11/29/2022] Open
Affiliation(s)
- Young Joo Jeon
- Institute for Clinical Research; CHA Bundang Medical Center; CHA University; Seongnam South Korea
- Department of Biomedical Science; College of Life Science; CHA University; Seongnam South Korea
| | - Ji Hyang Kim
- Department of Obstetrics and Gynecology; CHA Bundang Medical Center; CHA University; Seongnam South Korea
| | - Bo Eun Lee
- Institute for Clinical Research; CHA Bundang Medical Center; CHA University; Seongnam South Korea
- Department of Biomedical Science; College of Life Science; CHA University; Seongnam South Korea
| | - HyungChul Rah
- Institute for Clinical Research; CHA Bundang Medical Center; CHA University; Seongnam South Korea
- Department of Biomedical Science; College of Life Science; CHA University; Seongnam South Korea
| | - Ji Eun Shin
- Department of Obstetrics and Gynecology; CHA Bundang Medical Center; CHA University; Seongnam South Korea
| | - Hojeong Kang
- Department of Obstetrics and Gynecology; CHA Bundang Medical Center; CHA University; Seongnam South Korea
| | - Dong Hee Choi
- Department of Obstetrics and Gynecology; CHA Bundang Medical Center; CHA University; Seongnam South Korea
| | - Tae Ki Yoon
- Fertility Center of CHA Gangnam Medical Center; CHA University; Seoul South Korea
| | - Woo Sik Lee
- Fertility Center of CHA Gangnam Medical Center; CHA University; Seoul South Korea
| | - Sung Han Shim
- Fertility Center of CHA Gangnam Medical Center; CHA University; Seoul South Korea
| | - Nam Keun Kim
- Institute for Clinical Research; CHA Bundang Medical Center; CHA University; Seongnam South Korea
- Department of Biomedical Science; College of Life Science; CHA University; Seongnam South Korea
| |
Collapse
|