1
|
Myszko M, Bychowski J, Skrzydlewska E, Łuczaj W. The Dual Role of Oxidative Stress in Atherosclerosis and Coronary Artery Disease: Pathological Mechanisms and Diagnostic Potential. Antioxidants (Basel) 2025; 14:275. [PMID: 40227238 PMCID: PMC11939617 DOI: 10.3390/antiox14030275] [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: 01/20/2025] [Revised: 02/18/2025] [Accepted: 02/24/2025] [Indexed: 04/15/2025] Open
Abstract
Oxidative stress plays a pivotal role in the pathogenesis of atherosclerosis and coronary artery disease (CAD), with both beneficial and detrimental effects on cardiovascular health. On one hand, the excessive production of reactive oxygen species (ROS) contributes to endothelial dysfunction, inflammation, and vascular remodeling, which are central to the development and progression of CAD. These pathological effects drive key processes such as atherosclerosis, plaque formation, and thrombosis. On the other hand, moderate levels of oxidative stress can have beneficial effects on cardiovascular health. These include regulating vascular tone by promoting blood vessel dilation, supporting endothelial function through nitric oxide production, and enhancing the immune response to prevent infections. Additionally, oxidative stress can stimulate cellular adaptation to stress, promote cell survival, and encourage angiogenesis, which helps form new blood vessels to improve blood flow. Oxidative stress also holds promise as a source of biomarkers that could aid in the diagnosis, prognosis, and monitoring of CAD. Specific oxidative markers, such as malondialdehyde (MDA), isoprostanes (isoP), ischemia-modified albumin, and antioxidant enzyme activity, have been identified as potential indicators of disease severity and therapeutic response. This review explores the dual nature of oxidative stress in atherosclerosis and CAD, examining its mechanisms in disease pathogenesis as well as its emerging role in clinical diagnostics and targeted therapies. The future directions for research aimed at harnessing the diagnostic and therapeutic potential of oxidative stress biomarkers are also discussed. Understanding the balance between the detrimental and beneficial effects of oxidative stress could lead to innovative approaches in the prevention and management of CAD.
Collapse
Affiliation(s)
- Marcin Myszko
- Department of Cardiology, Bialystok Regional Hospital, M. Skłodowskiej-Curie 25, 15-950 Bialystok, Poland; (M.M.); (J.B.)
| | - Jerzy Bychowski
- Department of Cardiology, Bialystok Regional Hospital, M. Skłodowskiej-Curie 25, 15-950 Bialystok, Poland; (M.M.); (J.B.)
| | - Elżbieta Skrzydlewska
- Department of Analytical Chemistry, Medical University of Bialystok, Mickiewicza 2d, 15-222 Bialystok, Poland;
| | - Wojciech Łuczaj
- Department of Analytical Chemistry, Medical University of Bialystok, Mickiewicza 2d, 15-222 Bialystok, Poland;
| |
Collapse
|
2
|
Williams H, Simmonds S, Bond A, Somos A, Li Z, Forbes T, Bianco R, Dugdale C, Brown Z, Rice H, Herman A, Johnson J, George S. CCN4 (WISP-1) reduces apoptosis and atherosclerotic plaque burden in an ApoE mouse model. Atherosclerosis 2024; 397:118570. [PMID: 39276419 PMCID: PMC7617386 DOI: 10.1016/j.atherosclerosis.2024.118570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 08/21/2024] [Accepted: 08/21/2024] [Indexed: 09/17/2024]
Abstract
BACKGROUND AND AIMS CCN4/WISP-1 regulates various cell behaviours that contribute to atherosclerosis progression, including cell adhesion, migration, proliferation and survival. We therefore hypothesised that CCN4 regulates the development and progression of atherosclerotic plaques. METHODS We used a high fat fed ApoE-/- mouse model to study atherosclerotic plaque progression in the brachiocephalic artery and aortic root. In protocol 1, male ApoE-/- mice with established plaques were given a CCN4 helper-dependent adenovirus to see the effect of treatment with CCN4, while in protocol 2 male CCN4-/-ApoE-/- were compared to CCN4+/+ApoE-/- mice to assess the effect of CCN4 deletion on plaque progression. RESULTS CCN4 overexpression resulted in reduced occlusion of the brachiocephalic artery with less apoptosis, fewer macrophages, and attenuated lipid core size. The amount of plaque found on the aortic root was also reduced. CCN4 deficiency resulted in increased apoptosis and occlusion of the brachiocephalic artery as well as increased plaque in the aortic root. Additionally, in vitro cells from CCN4-/-ApoE-/- mice had higher apoptotic levels. CCN4 deficiency did not significantly affect blood cholesterol levels or circulating myeloid cell populations. CONCLUSIONS We conclude that in an atherosclerosis model the most important action of CCN4 is the effect on cell apoptosis. CCN4 provides pro-survival signals and leads to reduced cell death, lower macrophage number, smaller lipid core size and reduced atherosclerotic plaque burden. As such, the pro-survival effect of CCN4 is worthy of further investigation, in a bid to find a therapeutic for atherosclerosis.
Collapse
Affiliation(s)
- Helen Williams
- Bristol Heart Institute, Bristol Medical School, University of Bristol, UK.
| | | | - Andrew Bond
- Bristol Heart Institute, Bristol Medical School, University of Bristol, UK
| | - Alexandros Somos
- Bristol Heart Institute, Bristol Medical School, University of Bristol, UK
| | - Ze Li
- Bristol Heart Institute, Bristol Medical School, University of Bristol, UK
| | - Tessa Forbes
- Bristol Heart Institute, Bristol Medical School, University of Bristol, UK
| | - Rosaria Bianco
- Bristol Heart Institute, Bristol Medical School, University of Bristol, UK
| | - Celyn Dugdale
- Flow Cytometry Facility, School of Cellular & Molecular Medicine, University of Bristol, UK
| | - Zoe Brown
- Flow Cytometry Facility, School of Cellular & Molecular Medicine, University of Bristol, UK
| | - Helen Rice
- Flow Cytometry Facility, School of Cellular & Molecular Medicine, University of Bristol, UK
| | - Andrew Herman
- Flow Cytometry Facility, School of Cellular & Molecular Medicine, University of Bristol, UK
| | - Jason Johnson
- Bristol Heart Institute, Bristol Medical School, University of Bristol, UK
| | - Sarah George
- Bristol Heart Institute, Bristol Medical School, University of Bristol, UK
| |
Collapse
|
3
|
Jia W, Yuan J, Zhang J, Li S, Lin W, Cheng B. Bioactive sphingolipids as emerging targets for signal transduction in cancer development. Biochim Biophys Acta Rev Cancer 2024; 1879:189176. [PMID: 39233263 DOI: 10.1016/j.bbcan.2024.189176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 08/28/2024] [Accepted: 08/28/2024] [Indexed: 09/06/2024]
Abstract
Sphingolipids, crucial components of cellular membranes, play a vital role in maintaining cellular structure and signaling integrity. Disruptions in sphingolipid metabolism are increasingly implicated in cancer development. Key bioactive sphingolipids, such as ceramides, sphingosine-1-phosphate (S1P), ceramide-1-phosphate (C1P), and glycosphingolipids, profoundly impact tumor biology. They influence the behavior of tumor cells, stromal cells, and immune cells, affecting tumor aggressiveness, angiogenesis, immune modulation, and extracellular matrix remodeling. Furthermore, abnormal expression of sphingolipids and their metabolizing enzymes modulates the secretion of tumor-derived extracellular vesicles (TDEs), which are key players in creating an immunosuppressive tumor microenvironment, remodeling the extracellular matrix, and facilitating oncogenic signaling within in situ tumors and distant pre-metastatic niches (PMNs). Understanding the role of sphingolipids in the biogenesis of tumor-derived extracellular vesicles (TDEs) and their bioactive contents can pave the way for new biomarkers in cancer diagnosis and prognosis, ultimately enhancing comprehensive tumor treatment strategies.
Collapse
Affiliation(s)
- Wentao Jia
- Department of General Practice, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; Oncology Department of Traditional Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; Faculty of Traditional Chinese Medicine, Naval Medical University, Shanghai 200043, China
| | - Jiaying Yuan
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Jinbo Zhang
- Department of Pharmacy, Tianjin Rehabilitation and Recuperation Center, Joint Logistics Support Force, Tianjin 300000, China
| | - Shu Li
- Department of Gastroenterology, Baoshan Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201900, China
| | - Wanfu Lin
- Oncology Department of Traditional Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; Faculty of Traditional Chinese Medicine, Naval Medical University, Shanghai 200043, China.
| | - Binbin Cheng
- Oncology Department of Traditional Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; Faculty of Traditional Chinese Medicine, Naval Medical University, Shanghai 200043, China.
| |
Collapse
|
4
|
Gianopoulos I, Daskalopoulou SS. Macrophage profiling in atherosclerosis: understanding the unstable plaque. Basic Res Cardiol 2024; 119:35-56. [PMID: 38244055 DOI: 10.1007/s00395-023-01023-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 11/01/2023] [Accepted: 11/01/2023] [Indexed: 01/22/2024]
Abstract
The development and rupture of atherosclerotic plaques is a major contributor to myocardial infarctions and ischemic strokes. The dynamic evolution of the plaque is largely attributed to monocyte/macrophage functions, which respond to various stimuli in the plaque microenvironment. To this end, macrophages play a central role in atherosclerotic lesions through the uptake of oxidized low-density lipoprotein that gets trapped in the artery wall, and the induction of an inflammatory response that can differentially affect the stability of the plaque in men and women. In this environment, macrophages can polarize towards pro-inflammatory M1 or anti-inflammatory M2 phenotypes, which represent the extremes of the polarization spectrum that include Mhem, M(Hb), Mox, and M4 populations. However, this traditional macrophage model paradigm has been redefined to include numerous immune and nonimmune cell clusters based on in-depth unbiased single-cell approaches. The goal of this review is to highlight (1) the phenotypic and functional properties of monocyte subsets in the circulation, and macrophage populations in atherosclerotic plaques, as well as their contribution towards stable or unstable phenotypes in men and women, and (2) single-cell RNA sequencing studies that have advanced our knowledge of immune, particularly macrophage signatures present in the atherosclerotic niche. We discuss the importance of performing high-dimensional approaches to facilitate the development of novel sex-specific immunotherapies that aim to reduce the risk of cardiovascular events.
Collapse
Affiliation(s)
- Ioanna Gianopoulos
- Division of Experimental Medicine, Department of Medicine, Faculty of Medicine and Health Sciences, Research Institute of the McGill University Health Centre, McGill University, Montreal, Canada
| | - Stella S Daskalopoulou
- Division of Experimental Medicine, Department of Medicine, Faculty of Medicine and Health Sciences, Research Institute of the McGill University Health Centre, McGill University, Montreal, Canada.
- Division of Internal Medicine, Department of Medicine, Faculty of Medicine and Health Sciences, McGill University Health Centre, McGill University, Montreal, Canada.
- Department of Medicine, Research Institute of the McGill University Health Centre, Glen Site, 1001 Decarie Boulevard, EM1.2210, Montreal, Quebec, H4A 3J1, Canada.
| |
Collapse
|
5
|
Atkinson G, Bianco R, Di Gregoli K, Johnson JL. The contribution of matrix metalloproteinases and their inhibitors to the development, progression, and rupture of abdominal aortic aneurysms. Front Cardiovasc Med 2023; 10:1248561. [PMID: 37799778 PMCID: PMC10549934 DOI: 10.3389/fcvm.2023.1248561] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 09/07/2023] [Indexed: 10/07/2023] Open
Abstract
Abdominal aortic aneurysms (AAAs) account for up to 8% of deaths in men aged 65 years and over and 2.2% of women. Patients with AAAs often have atherosclerosis, and intimal atherosclerosis is generally present in AAAs. Accordingly, AAAs are considered a form of atherosclerosis and are frequently referred to as atherosclerotic aneurysms. Pathological observations advocate inflammatory cell infiltration alongside adverse extracellular matrix degradation as key contributing factors to the formation of human atherosclerotic AAAs. Therefore, macrophage production of proteolytic enzymes is deemed responsible for the damaging loss of ECM proteins, especially elastin and fibrillar collagens, which characterise AAA progression and rupture. Matrix metalloproteinases (MMPs) and their regulation by tissue inhibitors metalloproteinases (TIMPs) can orchestrate not only ECM remodelling, but also moderate the proliferation, migration, and apoptosis of resident aortic cells, alongside the recruitment and subsequent behaviour of inflammatory cells. Accordingly, MMPs are thought to play a central regulatory role in the development, progression, and eventual rupture of abdominal aortic aneurysms (AAAs). Together, clinical and animal studies have shed light on the complex and often diverse effects MMPs and TIMPs impart during the development of AAAs. This dichotomy is underlined from evidence utilising broad-spectrum MMP inhibition in animal models and clinical trials which have failed to provide consistent protection from AAA progression, although more encouraging results have been observed through deployment of selective inhibitors. This review provides a summary of the supporting evidence connecting the contribution of individual MMPs to AAA development, progression, and eventual rupture. Topics discussed include structural, functional, and cell-specific diversity of MMP members; evidence from animal models of AAA and comparisons with findings in humans; the dual role of MMPs and the requirement to selectively target individual MMPs; and the advances in identifying aberrant MMP activity. As evidenced, our developing understanding of the multifaceted roles individual MMPs perform during the progression and rupture of AAAs, should motivate clinical trials assessing the therapeutic potential of selective MMP inhibitors, which could restrict AAA-related morbidity and mortality worldwide.
Collapse
Affiliation(s)
| | | | | | - Jason L. Johnson
- Laboratory of Cardiovascular Pathology, Department of Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
6
|
Cai C, Weng Y, Wang X, Wu Y, Li Y, Wang P, Zeng C, Yang Z, Jia B, Tang L, Chen L. Single-cell RNA landscape of cell heterogeneity and immune microenvironment in ligation-induced vascular remodeling in rat. Atherosclerosis 2023; 377:1-11. [PMID: 37343431 DOI: 10.1016/j.atherosclerosis.2023.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 06/08/2023] [Accepted: 06/08/2023] [Indexed: 06/23/2023]
Abstract
BACKGROUND AND AIMS Vascular remodeling is a common pathological basis for cardiovascular diseases. Although both immune and non-immune cells have been suggested to contribute to this process, the complex cellular heterogeneity and intercellular interactions remain largely uncharacterized. METHODS AND RESULTS In this study, we simulated early and late vascular remodeling by ligating the rat carotid artery for 1 week and 4 weeks, respectively. Using single-cell RNA-sequencing, we characterized gene expression signatures and driver signals of major cell types involved in vascular remodeling. Focused analysis revealed a novel sub-population of Selenbp1hi smooth muscle cells (SMCs) associated with vascular remodeling. Results of intercellular communication analyses predicted several ligand-receptor pairs between immune cells with SMCs and endothelial cells (ECs), implicating SMCs apoptosis and repair, ECs aging and inflammatory responses. CONCLUSIONS We present a comprehensive single-cell atlas of vascular cells in early and late stages of ligated rat carotid artery, providing valuable insights into the understanding of the initiation and progression of vascular remodeling.
Collapse
Affiliation(s)
- Changhong Cai
- Department of Cardiology, Fujian Heart Medical Center, Fujian Institute of Coronary Heart Disease, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Yingzheng Weng
- Department of Cardiology, Zhejiang Hospital, Hangzhou, 310013, China; Department of Medicine, The Second College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310013, China
| | - Xihao Wang
- Department of Medicine, The Second College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310013, China
| | - Yonghui Wu
- Department of Cardiology, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Municipal Central Hospital, Lishui, 323000, China
| | - Ya Li
- Department of Cardiology, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Municipal Central Hospital, Lishui, 323000, China
| | - Peipei Wang
- Department of Cardiology, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Municipal Central Hospital, Lishui, 323000, China
| | - Chunlai Zeng
- Department of Cardiology, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Municipal Central Hospital, Lishui, 323000, China
| | - Zhouxin Yang
- Zhejiang Provincial Key Lab of Geriatrics, Department of Geriatrics, Zhejiang Hospital, Hangzhou, 310013, China
| | - Bingbing Jia
- Zhejiang Provincial Key Lab of Geriatrics, Department of Geriatrics, Zhejiang Hospital, Hangzhou, 310013, China.
| | - Lijiang Tang
- Department of Cardiology, Zhejiang Hospital, Hangzhou, 310013, China.
| | - Lianglong Chen
- Department of Cardiology, Fujian Heart Medical Center, Fujian Institute of Coronary Heart Disease, Fujian Medical University Union Hospital, Fuzhou, 350001, China.
| |
Collapse
|
7
|
Bräuninger H, Krüger S, Bacmeister L, Nyström A, Eyerich K, Westermann D, Lindner D. Matrix metalloproteinases in coronary artery disease and myocardial infarction. Basic Res Cardiol 2023; 118:18. [PMID: 37160529 PMCID: PMC10169894 DOI: 10.1007/s00395-023-00987-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 05/11/2023]
Abstract
Cardiovascular diseases (CVDs) remain the leading cause of death worldwide. Most cardiovascular deaths are caused by ischaemic heart diseases such as myocardial infarction (MI). Hereby atherosclerosis in the coronary arteries often precedes disease manifestation. Since tissue remodelling plays an important role in the development and progression of atherosclerosis as well as in outcome after MI, regulation of matrix metalloproteinases (MMPs) as the major ECM-degrading enzymes with diverse other functions is crucial. Here, we provide an overview of the expression profiles of MMPs in coronary artery and left ventricular tissue using publicly available data from whole tissue to single-cell resolution. To approach an association between MMP expression and the development and outcome of CVDs, we further review studies investigating polymorphisms in MMP genes since polymorphisms are known to have an impact on gene expression. This review therefore aims to shed light on the role of MMPs in atherosclerosis and MI by summarizing current knowledge from publically available datasets, human studies, and analyses of polymorphisms up to preclinical and clinical trials of pharmacological MMP inhibition.
Collapse
Affiliation(s)
- Hanna Bräuninger
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Side Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Saskia Krüger
- Clinic for Cardiology, University Heart and Vascular Centre Hamburg, Hamburg, Germany
| | - Lucas Bacmeister
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany
| | - Alexander Nyström
- Department of Dermatology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Kilian Eyerich
- Department of Dermatology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Dirk Westermann
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany
| | - Diana Lindner
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany.
- German Centre for Cardiovascular Research (DZHK), Partner Side Hamburg/Kiel/Lübeck, Hamburg, Germany.
| |
Collapse
|
8
|
Giagtzidis I, Karkos C, Kadoglou FNPE, Spathis A, Papazoglou K. Serum levels of Matrix Metalloproteinases (MMPs) in patients undergoing endovascular intervention for peripheral arterial disease. Ann Vasc Surg 2023:S0890-5096(23)00250-9. [PMID: 37169253 DOI: 10.1016/j.avsg.2023.04.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 04/24/2023] [Accepted: 04/25/2023] [Indexed: 05/13/2023]
Abstract
OBJECTIVES Matrix metalloproteinases (MMPs) play a significant role in the development and progression of atherosclerotic vascular disease. The purpose of this study was to measure and document the profile of plasma circulating MMPs in patients with peripheral arterial disease (PAD) undergoing endovascular revascularization. METHODS This was a single centre prospective observational study with 80 patients with PAD enrolled. They underwent percutaneous balloon angioplasty and/or angioplasty with stent. Exclusion criteria were acute limb ischemia, active inflammation, wet gangrene, liver disease, end stage renal failure and cancer. Patients that underwent open or hybrid (open and endovascular) approach, were also excluded from the study. Venous blood samples were taken preoperatively, 24 hours and 6 months postoperatively. The values of MMP-2, MMP-3, MMP-7, MMP-9 and their inhibitors (Tissue Inhibitor of metalloproteinases, TIMP), TIMP-1 and TIMP-2 were measured. RESULTS The mean age was 67.1 years and 66 of them (82.5%) were male. During the clinical follow up (mean 35.8% months), 12 patients died (16.4%), 15 (20.5%) of them had a major adverse limb event (MALE) and 14 (19.2%) of them had a major adverse cardiovascular event (MACE). There was a statistically significant raise in the values of MMP-2. MMP-3 and MMP-7 at 6 months postoperatively, when compared to the preoperative and 24 hours postoperative values. There was no correlation of MMP and TIMP values with mortality, MALE and MACE events. CONCLUSIONS The present single-centre prospective study documented increased circulating levels of MMPs post-operatively in PAD patients undergoing endovascular treatment. Vascular trauma caused by angioplasty, could trigger expression of MMPs and TIMPs, but the absence of any association with clinical complications requires further investigation.
Collapse
Affiliation(s)
- Ioakeim Giagtzidis
- Aristotle University of Thessaloniki, Ippokratio General Hospital, 5(th) Surgical Department, Thessaloniki, Greece.
| | - Christos Karkos
- Aristotle University of Thessaloniki, Ippokratio General Hospital, 5(th) Surgical Department, Thessaloniki, Greece
| | | | - Aris Spathis
- Department of Cytopathology, "Attikon" University Hospital, Athens, Greece
| | - Konstantinos Papazoglou
- Aristotle University of Thessaloniki, Ippokratio General Hospital, 5(th) Surgical Department, Thessaloniki, Greece
| |
Collapse
|
9
|
Xiang W, Li L, Hong F, Zeng Y, Zhang J, Xie J, Shen G, Wang J, Fang Z, Qi W, Yang X, Gao G, Zhou T. N-cadherin cleavage: A critical function that induces diabetic retinopathy fibrosis via regulation of β-catenin translocation. FASEB J 2023; 37:e22878. [PMID: 36939278 DOI: 10.1096/fj.202201664rr] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 02/15/2023] [Accepted: 03/02/2023] [Indexed: 03/21/2023]
Abstract
Retinal fibrosis is a severe pathological change in the late stage of diabetic retinopathy and is also the leading cause of blindness. We have previously revealed that N-cadherin was significantly increased in type 1 and type 2 diabetic mice retinas and the fibrovascular membranes from proliferative diabetic retinopathy (PDR) patients. However, whether N-cadherin directly induces retinal fibrosis in DR and the related mechanism is unknown. Here, we investigated the pathogenic role of N-cadherin in mediating retinal fibrosis and further explored the relevant therapeutic targets. We found that the level of N-cadherin was significantly increased in PDR patients and STZ-induced diabetic mice and positively correlated with the fibrotic molecules Connective Tissue Growth Factor (CTGF) and fibronectin (FN). Moreover, intravitreal injection of N-cadherin adenovirus significantly increased the expression of FN and CTGF in normal mice retinas. Mechanistically, overexpression of N-cadherin promotes N-cadherin cleavage, and N-cadherin cleavage can further induce translocation of non-p-β-catenin in the nucleus and upregulation of fibrotic molecules. Furthermore, we found a novel N-cadherin cleavage inhibitor, pigment epithelial-derived factor (PEDF), which ameliorated the N-cadherin cleavage and subsequent retinal fibrosis in diabetic mice. Thus, our findings provide novel evidence that elevated N-cadherin level not only acts as a classic EMT maker but also plays a causative role in diabetic retinal fibrosis, and targeting N-cadherin cleavage may provide a strategy to inhibit retinal fibrosis in DR patients.
Collapse
Affiliation(s)
- Wei Xiang
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Department of Clinical Laboratory Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Longhui Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Fuyan Hong
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yongcheng Zeng
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jin Zhang
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jinye Xie
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Gang Shen
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jinhong Wang
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zhenzhen Fang
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Weiwei Qi
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Xia Yang
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Guoquan Gao
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Guangdong Engineering & Technology Research Center for Gene Manipulation and Biomacromolecular Products, Sun Yat-Sen University, Guangzhou, China
| | - Ti Zhou
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- China Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China
| |
Collapse
|
10
|
Rezaei M, Faramarzpour M, Shobeiri P, Seyedmirzaei H, Sarasyabi MS, Dabiri S. A systematic review, meta-analysis, and network analysis of diagnostic microRNAs in glaucoma. Eur J Med Res 2023; 28:137. [PMID: 36973823 PMCID: PMC10041737 DOI: 10.1186/s40001-023-01093-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 03/09/2023] [Indexed: 03/29/2023] Open
Abstract
Glaucoma is a chronic neurodegenerative process of the optic nerve that is the leading cause of blindness worldwide, and early diagnosis of the disease could greatly affect patients' prognoses. The pathophysiology of glaucoma is complicated by a combination of genetic and epigenetic factors. Deciphering the early diagnostic biomarkers in glaucoma could attenuate the disease's global burden and help us understand the exact mechanisms involved in glaucoma. The microRNAs are members of a larger family of non-coding RNAs that play an essential role in the epigenetic basis of glaucoma. A systematic study and meta-analysis of diagnostic microRNAs in glaucoma, jointly with network analysis of target genes, were carried out on published papers assessing differentially expressed microRNAs in human subjects. In total, 321 articles were found, and, after screening, six studies were eligible for further analysis. 52 differentially expressed microRNAs were found, of which 28 and 24 were up-regulated and down-regulated, respectively. Only 12 microRNAs were qualified for meta-analysis, with overall sensitivity and specificity of 80% and 74%, respectively. Then, using network analysis, it became apparent that the VEGF-A, AKT1, CXCL12, and HRAS genes were the most important targets for the microRNAs. Perturbations in WNT signaling, protein transport, and extracellular matrix organization pathways were discovered to be important in the etiology of glaucoma using the community detection approach. This study tries to uncover the promising microRNAs and their target genes that govern the epigenetics of glaucoma.
Collapse
Affiliation(s)
- Masoud Rezaei
- Research Center for Hydatid Disease in Iran, Kerman University of Medical Sciences, Kerman, Iran
- Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mahsa Faramarzpour
- Pathology and Stem Cell Research Center, Department of Pathology, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
- Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Parnian Shobeiri
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Endocrinology and Metabolism Population Sciences Institute, Non-Communicable Diseases Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Homa Seyedmirzaei
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mohammad Sharifi Sarasyabi
- Research Center for Hydatid Disease in Iran, Kerman University of Medical Sciences, Kerman, Iran
- Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
- Department of Radiology, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Shahriar Dabiri
- Pathology and Stem Cell Research Center, Department of Pathology, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
11
|
Jiang Y, Qian HY. Transcription factors: key regulatory targets of vascular smooth muscle cell in atherosclerosis. Mol Med 2023; 29:2. [PMID: 36604627 PMCID: PMC9817296 DOI: 10.1186/s10020-022-00586-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 12/05/2022] [Indexed: 01/07/2023] Open
Abstract
Atherosclerosis (AS), leading to gradual occlusion of the arterial lumen, refers to the accumulation of lipids and inflammatory debris in the arterial wall. Despite therapeutic advances over past decades including intervention or surgery, atherosclerosis is still the most common cause of cardiovascular diseases and the main mechanism of death and disability worldwide. Vascular smooth muscle cells (VSMCs) play an imperative role in the occurrence of atherosclerosis and throughout the whole stages. In the past, there was a lack of comprehensive understanding of VSMCs, but the development of identification technology, including in vivo single-cell sequencing technology and lineage tracing with the CreERT2-loxP system, suggests that VSMCs have remarkable plasticity and reevaluates well-established concepts about the contribution of VSMCs. Transcription factors, a kind of protein molecule that specifically recognizes and binds DNA upstream promoter regions or distal enhancer DNA elements, play a key role in the transcription initiation of the coding genes and are necessary for RNA polymerase to bind gene promoters. In this review, we highlight that, except for environmental factors, VSMC genes are transcriptionally regulated through complex interactions of multiple conserved cis-regulatory elements and transcription factors. In addition, through a series of transcription-related regulatory processes, VSMCs could undergo phenotypic transformation, proliferation, migration, calcification and apoptosis. Finally, enhancing or inhibiting transcription factors can regulate the development of atherosclerotic lesions, and the downstream molecular mechanism of transcriptional regulation has also been widely studied.
Collapse
Affiliation(s)
- Yu Jiang
- grid.506261.60000 0001 0706 7839Center for Coronary Heart Disease, Department of Cardiology, Fu Wai Hospital, National Center for Cardiovascular Diseases of China, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Rd, Beijing, 100037 China
| | - Hai-Yan Qian
- grid.506261.60000 0001 0706 7839Center for Coronary Heart Disease, Department of Cardiology, Fu Wai Hospital, National Center for Cardiovascular Diseases of China, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Rd, Beijing, 100037 China
| |
Collapse
|
12
|
Asgari R, Yarani R, Mohammadi P, Emami Aleagha MS. HIF-1α in the Crosstalk Between Reactive Oxygen Species and Autophagy Process: A Review in Multiple Sclerosis. Cell Mol Neurobiol 2022; 42:2121-2129. [PMID: 34089426 PMCID: PMC11421632 DOI: 10.1007/s10571-021-01111-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 05/27/2021] [Indexed: 12/16/2022]
Abstract
Cellular stress can lead to the production of reactive oxygen species (ROS) while autophagy, as a catabolic pathway, protects the cells against stress. Autophagy in its turn plays a pivotal role in the pathophysiology of multiple sclerosis (MS). In the current review, we first summarized the contribution of ROS and autophagy to MS pathogenesis. Then probable crosstalk between these two pathways through HIF-1α for the first time has been proposed with the hope of employing a better understanding of MS pathophysiology and probable therapeutic approaches.
Collapse
Affiliation(s)
- Rezvan Asgari
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Reza Yarani
- Translational Type 1 Diabetes Biology, Department of Clinical Research, Steno Diabetes Center Copenhagen, Copenhagen, Denmark
| | - Pantea Mohammadi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | | |
Collapse
|
13
|
Kawai K, Vozenilek AE, Kawakami R, Sato Y, Ghosh SKB, Virmani R, Finn AV. Understanding the role of alternative macrophage phenotypes in human atherosclerosis. Expert Rev Cardiovasc Ther 2022; 20:689-705. [PMID: 35942866 DOI: 10.1080/14779072.2022.2111301] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Abstract
INTRODUCTION Atherosclerosis-based ischemic heart disease is still the primary cause of death throughout the world. Over the past decades there has been no significant changes in the therapeutic approaches to atherosclerosis, which are mainly based on lipid lowering therapies and management of comorbid conditions such as diabetes and hypertension. The involvement of macrophages in atherosclerosis has been recognized for decades. More recently, a more detailed and sophisticated understanding of their various phenotypes and roles in the atherosclerotic process has been recognized. This new data is revealing how specific subtypes of macrophage-induced inflammation may have distinct effects on atherosclerosis progression and may provide new approaches for treatment, based upon targeting of specific macrophage subtypes. AREAS COVERED We will comprehensively review the spectrum of macrophage phenotypes and how they contribute to atherosclerotic plaque development and progression. EXPERT OPINION Various signals derived from atherosclerotic lesions drive macrophages into complex subsets with different gene expression profiles, phenotypes, and functions, not all of which are understood. Macrophage phenotypes include those that enhance, heal, and regress the atherosclerotic lesions though various mechanisms. Targeting of specific macrophage phenotypes may provide a promising and novel approach to prevent atherosclerosis progression.
Collapse
Affiliation(s)
- Kenji Kawai
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD, USA
| | - Aimee E Vozenilek
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD, USA
| | - Rika Kawakami
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD, USA
| | - Yu Sato
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD, USA
| | | | - Renu Virmani
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD, USA
| | - Aloke V Finn
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD, USA.,University of Maryland, School of Medicine, Baltimore, MD, USA
| |
Collapse
|
14
|
Gkaliagkousi E, Lazaridis A, Dogan S, Fraenkel E, Tuna BG, Mozos I, Vukicevic M, Yalcin O, Gopcevic K. Theories and Molecular Basis of Vascular Aging: A Review of the Literature from VascAgeNet Group on Pathophysiological Mechanisms of Vascular Aging. Int J Mol Sci 2022; 23:ijms23158672. [PMID: 35955804 PMCID: PMC9368987 DOI: 10.3390/ijms23158672] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 07/25/2022] [Accepted: 07/29/2022] [Indexed: 11/29/2022] Open
Abstract
Vascular aging, characterized by structural and functional alterations of the vascular wall, is a hallmark of aging and is tightly related to the development of cardiovascular mortality and age-associated vascular pathologies. Over the last years, extensive and ongoing research has highlighted several sophisticated molecular mechanisms that are involved in the pathophysiology of vascular aging. A more thorough understanding of these mechanisms could help to provide a new insight into the complex biology of this non-reversible vascular process and direct future interventions to improve longevity. In this review, we discuss the role of the most important molecular pathways involved in vascular ageing including oxidative stress, vascular inflammation, extracellular matrix metalloproteinases activity, epigenetic regulation, telomere shortening, senescence and autophagy.
Collapse
Affiliation(s)
- Eugenia Gkaliagkousi
- 3rd Department of Internal Medicine, Papageorgiou Hospital, Faculty of Medicine, Aristotle University of Thessaloniki, 56429 Thessaloniki, Greece
- Correspondence: (E.G.); (K.G.)
| | - Antonios Lazaridis
- 3rd Department of Internal Medicine, Papageorgiou Hospital, Faculty of Medicine, Aristotle University of Thessaloniki, 56429 Thessaloniki, Greece
| | - Soner Dogan
- Department of Medical Biology, School of Medicine, Yeditepe University, 34755 Istanbul, Turkey
| | - Emil Fraenkel
- 1st Department of Internal Medicine, University Hospital, Pavol Jozef Šafárik University of Košice, Trieda SNP 1, 04066 Košice, Slovakia
| | - Bilge Guvenc Tuna
- Department of Biophysics, School of Medicine, Yeditepe University, 34755 Istanbul, Turkey
| | - Ioana Mozos
- Department of Functional Sciences-Pathophysiology, Center for Translational Research and Systems Medicine, “Victor Babes” University of Medicine and Pharmacy, 300173 Timisoara, Romania
| | - Milica Vukicevic
- Cardiac Surgery Clinic, Clinical Center of Serbia, 11000 Belgrade, Serbia
| | - Ozlem Yalcin
- Department of Physiology, School of Medicine, Koc University, 34450 Istanbul, Turkey
| | - Kristina Gopcevic
- Laboratory for Analytics of Biomolecules, Department of Chemistry in Medicine, Faculty of Medicine, 11000 Belgrade, Serbia
- Correspondence: (E.G.); (K.G.)
| |
Collapse
|
15
|
Liu Y, Li Y, Chen M, Liu Y, Liang J, Zhang Y, Qian ZJ. Mechanism of two alkaloids isolated from coral endophytic fungus for suppressing angiogenesis in atherosclerotic plaque in HUVEC. Int Immunopharmacol 2022; 109:108931. [PMID: 35704971 DOI: 10.1016/j.intimp.2022.108931] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/20/2022] [Accepted: 06/05/2022] [Indexed: 11/05/2022]
Abstract
Atherosclerosis is a significant cause of many cardiovascular diseases. Oxidized low-density lipoproteins (ox-LDL) are crucial in developing atherosclerosis. In this study, we researched the effects of two alkaloids epi-aszonalenin A (EAA) and aszonalenin (AZN) of an endophytic fungus Aspergillus terreus C23-3 from coral Pavona, on ox-LDL-induced inflammation, apoptosis and angiogenesis in HUVEC, and evaluated related factors and mechanism. The results reveal that EAA and AZN inhibit HUVEC migration, invasion, angiogenesis and reactive oxygen species (ROS) accumulation on a non-cytotoxic basis. Then, EAA and AZN suppressed the ox-LDL-induced of LOX-1, VEGF protein expression, MAPK and PI3K/AKT pathways phosphorylation. Furthermore, AZN suppressed the ox-LDL-induced inflammatory factors (IL-6, IL-1β, and TNF-α), intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1) and VEGF receptor VEGFR-2 and platelet-derived growth factor PDGF. In addition, it inhibited ox-LDL-induced atherosclerosis by blocking inflammation and apoptosis through nuclear factor κB (NF-κB), cleaved-caspase-3 and Bax/Bcl-2 pathways. Molecular docking results confirm that the effect of AZN on atherosclerosis inhibitory activity may be attributed to hydrogen bonds formed into LOX-1 and VEGFR-2. These data indicate that EAA and AZN can effectively prevent ox-LDL-induced HUVEC damage and angiogenesis by inhibiting inflammation and apoptosis. Therefore, EAA and AZN may have potential beneficial effects in regulating atherosclerosis and plaque angiogenesis.
Collapse
Affiliation(s)
- Yi Liu
- School of Chemistry and Environment, College of Food Science and Technology, Shenzhen Institute of Guangdong Ocean University, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Zhanjiang 524-088, China
| | - Yanmei Li
- School of Chemistry and Environment, College of Food Science and Technology, Shenzhen Institute of Guangdong Ocean University, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Zhanjiang 524-088, China
| | - Minqi Chen
- School of Chemistry and Environment, College of Food Science and Technology, Shenzhen Institute of Guangdong Ocean University, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Zhanjiang 524-088, China
| | - Yayue Liu
- School of Chemistry and Environment, College of Food Science and Technology, Shenzhen Institute of Guangdong Ocean University, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Zhanjiang 524-088, China
| | - Jinyue Liang
- School of Chemistry and Environment, College of Food Science and Technology, Shenzhen Institute of Guangdong Ocean University, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Zhanjiang 524-088, China
| | - Yi Zhang
- School of Chemistry and Environment, College of Food Science and Technology, Shenzhen Institute of Guangdong Ocean University, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Zhanjiang 524-088, China; Southern Marine Science and Engineering Guangdong Laboratory, Zhanjiang 524-088, China.
| | - Zhong-Ji Qian
- School of Chemistry and Environment, College of Food Science and Technology, Shenzhen Institute of Guangdong Ocean University, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Zhanjiang 524-088, China; Southern Marine Science and Engineering Guangdong Laboratory, Zhanjiang 524-088, China.
| |
Collapse
|
16
|
Larochelle J, Yang C, Liu L, Candelario-Jalil E. An Unexplored Role for MMP-7 (Matrix Metalloproteinase-7) in Promoting Gut Permeability After Ischemic Stroke. Stroke 2022; 53:3238-3242. [PMID: 35904018 DOI: 10.1161/strokeaha.122.040144] [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] [Indexed: 11/16/2022]
Abstract
Poststroke infections are common complications of stroke and are highly associated with poor outcomes for patients. Stroke induces profound immunodepression coupled with alterations to autonomic signaling, which together render the body more susceptible to infection from without (nosocomial/community-acquired infection) and from within (commensal bacterial infection). Critical to the hypothesis of commensal infection is the phenomenon of poststroke gut permeability and gut dysbiosis. Few studies have provided adequate explanations for the mechanisms underlying the molecular alterations that produce a more permeable gut and perturbed gut microbiota after stroke. A dysregulation in the production of matrix MMP-7 (metalloproteinase-7) may play a critical role in the progression of gut permeability after stroke. By cleaving junctional and extracellular matrix proteins, MMP-7 is capable of compromising gut barrier integrity. Because of MMP-7's unique abundance in the small intestine and its capacity to be induced in states of bacterial invasion and inflammation, along with its unique degradative capability, MMP-7 may be crucially important to the progression of gut permeability after ischemic stroke.
Collapse
Affiliation(s)
- Jonathan Larochelle
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville
| | - Changjun Yang
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville
| | - Lei Liu
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville
| | | |
Collapse
|
17
|
Kessler V, Klopf J, Eilenberg W, Neumayer C, Brostjan C. AAA Revisited: A Comprehensive Review of Risk Factors, Management, and Hallmarks of Pathogenesis. Biomedicines 2022; 10:94. [PMID: 35052774 PMCID: PMC8773452 DOI: 10.3390/biomedicines10010094] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 12/30/2021] [Indexed: 01/27/2023] Open
Abstract
Despite declining incidence and mortality rates in many countries, the abdominal aortic aneurysm (AAA) continues to represent a life-threatening cardiovascular condition with an overall prevalence of about 2-3% in the industrialized world. While the risk of AAA development is considerably higher for men of advanced age with a history of smoking, screening programs serve to detect the often asymptomatic condition and prevent aortic rupture with an associated death rate of up to 80%. This review summarizes the current knowledge on identified risk factors, the multifactorial process of pathogenesis, as well as the latest advances in medical treatment and surgical repair to provide a perspective for AAA management.
Collapse
Affiliation(s)
| | | | | | | | - Christine Brostjan
- Department of General Surgery, Division of Vascular Surgery, Medical University of Vienna, Vienna General Hospital, 1090 Vienna, Austria; (V.K.); (J.K.); (W.E.); (C.N.)
| |
Collapse
|
18
|
Vascular Pathobiology: Atherosclerosis and Large Vessel Disease. Cardiovasc Pathol 2022. [DOI: 10.1016/b978-0-12-822224-9.00006-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
19
|
Kremastiotis G, Handa I, Jackson C, George S, Johnson J. Disparate effects of MMP and TIMP modulation on coronary atherosclerosis and associated myocardial fibrosis. Sci Rep 2021; 11:23081. [PMID: 34848763 PMCID: PMC8632906 DOI: 10.1038/s41598-021-02508-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 11/12/2021] [Indexed: 11/12/2022] Open
Abstract
Matrix metalloproteinase (MMP) activity is tightly regulated by the endogenous tissue inhibitors (TIMPs), and dysregulated activity contributes to extracellular matrix remodelling. Accordingly, MMP/TIMP balance is associated with atherosclerotic plaque progression and instability, alongside adverse post-infarction cardiac fibrosis and subsequent heart failure. Here, we demonstrate that prolonged high-fat feeding of apolipoprotein (Apo)e-deficient mice triggered the development of unstable coronary artery atherosclerosis alongside evidence of myocardial infarction and progressive sudden death. Accordingly, the contribution of select MMPs and TIMPs to the progression of both interrelated pathologies was examined in Apoe-deficient mice with concomitant deletion of Mmp7, Mmp9, Mmp12, or Timp1 and relevant wild-type controls after 36-weeks high-fat feeding. Mmp7 deficiency increased incidence of sudden death, while Mmp12 deficiency promoted survival, whereas Mmp9 or Timp1 deficiency had no effect. While all mice harboured coronary disease, atherosclerotic burden was reduced in Mmp7-deficient and Mmp12-deficient mice and increased in Timp1-deficient animals, compared to relevant controls. Significant differences in cardiac fibrosis were only observed in Mmp-7-deficient mice and Timp1-deficient animals, which was associated with reduced capillary number. Adopting therapeutic strategies in Apoe-deficient mice, TIMP-2 adenoviral-overexpression or administration (delayed or throughout) of a non-selective MMP inhibitor (RS-130830) had no effect on coronary atherosclerotic burden or cardiac fibrosis. Taken together, our findings emphasise the divergent roles of MMPs on coronary plaque progression and associated post-MI cardiac fibrosis, highlighting the need for selective therapeutic approaches to target unstable atherosclerosis alongside adverse cardiac remodelling while negating detrimental adverse effects on either pathology, with targeting of MMP-12 seeming a suitable target.
Collapse
Affiliation(s)
- Georgios Kremastiotis
- Laboratory of Cardiovascular Pathology, Translational Health Sciences, Bristol Medical School, Faculty of Health Sciences, University of Bristol, Level 7, Bristol Royal Infirmary, Bristol, BS2 8HW, England, UK
| | - Ishita Handa
- Laboratory of Cardiovascular Pathology, Translational Health Sciences, Bristol Medical School, Faculty of Health Sciences, University of Bristol, Level 7, Bristol Royal Infirmary, Bristol, BS2 8HW, England, UK
| | - Christopher Jackson
- Laboratory of Cardiovascular Pathology, Translational Health Sciences, Bristol Medical School, Faculty of Health Sciences, University of Bristol, Level 7, Bristol Royal Infirmary, Bristol, BS2 8HW, England, UK
| | - Sarah George
- Laboratory of Cardiovascular Pathology, Translational Health Sciences, Bristol Medical School, Faculty of Health Sciences, University of Bristol, Level 7, Bristol Royal Infirmary, Bristol, BS2 8HW, England, UK
| | - Jason Johnson
- Laboratory of Cardiovascular Pathology, Translational Health Sciences, Bristol Medical School, Faculty of Health Sciences, University of Bristol, Level 7, Bristol Royal Infirmary, Bristol, BS2 8HW, England, UK.
| |
Collapse
|
20
|
TSGA10 as a Potential Key Factor in the Process of Spermatid Differentiation/Maturation: Deciphering Its Association with Autophagy Pathway. Reprod Sci 2021; 28:3228-3240. [PMID: 34232471 DOI: 10.1007/s43032-021-00648-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 05/30/2021] [Indexed: 10/20/2022]
Abstract
Testis-specific gene antigen 10 (TSGA10) plays an important role in spermatogenesis. However, the exact TSGA10 role and its relationship with the autophagy pathway in the process of spermatids differentiation/maturation is still not clear. Therefore, the present study evaluates the role of TSGA10 gene in the spermatid differentiation/maturation through its effect on autophagy and explores possible underlying pathway(s). Sperm samples from patients with teratospermia were collected. The mRNA and protein level of TSGA10 in these samples were assessed by real-time PCR and western blotting. Using the ingenuity pathway analysis (IPA) software, the gene network and interactions of TSGA10 involved in sperm maturation and autophagy were investigated. Based on these analyses, the expression levels of identified genes in patient's samples and healthy controls were further evaluated. Moreover, using flow cytometry analysis, the levels of reactive oxygen species (ROS( production in teratospermic sperm samples were evaluated. The results showed that the expression levels of TSGA10 mRNA and protein decreased significantly in the teratospermic patients compared to controls (P < 0.05). Moreover, a significant reduction in the expression of the important genes involved in sperm maturation and autophagy was observed (P < 0.05). Also, the levels of ROS production in teratospermic sperm samples were shown to be significantly higher compared to those in normal sperms (P < 0.05). Our findings provide new evidence that simultaneous decrease in TSGA10 and autophagy beside the increased level of ROS production in sperm cells might be associated with the abnormalities in the spermatids differentiation/maturation and the formation of sperms with abnormal morphology.
Collapse
|
21
|
Mozzini C, Girelli D, Cominacini L, Soresi M. An Exploratory Look at Bicuspid Aortic Valve (Bav) Aortopathy: Focus on Molecular and Cellular Mechanisms. Curr Probl Cardiol 2021; 46:100425. [PMID: 31097209 DOI: 10.1016/j.cpcardiol.2019.04.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 04/16/2019] [Indexed: 01/12/2023]
Abstract
Bicuspid aortic valve (BAV) is the most common congenital heart malformation. BAV patients are at increased risk for aortic valve disease (stenosis/regurgitation), infective endocarditis, thrombi formation and, in particular, aortic dilatation, aneurysm and dissection. This review aims at exploring the possible interplay among genetics, extracellular matrix remodeling, abnormal signaling pathways, oxidative stress and inflammation in contributing to BAV-associated aortopathy (BAV-A-A). Novel circulating biomarkers have been proposed as diagnostic tools able to improve risk stratification in BAV-A-A. However, to date, the precise molecular and cellular mechanisms that lead to BAV-A-A remain unknown. Genetic, hemodynamic and cardiovascular risk factors have been implicated in the development and progression of BAV-A-A. Oxidative stress may also play a role, similarly to what observed in atherosclerosis and vulnerable plaque formation. The identification of common pathways between these 2 conditions may provide a platform for future therapeutic solutions.
Collapse
|
22
|
Wang MD, Xing H, Li C, Liang L, Wu H, Xu XF, Sun LY, Wu MC, Shen F, Yang T. A novel role of Krüppel-like factor 8 as an apoptosis repressor in hepatocellular carcinoma. Cancer Cell Int 2020; 20:422. [PMID: 32874135 PMCID: PMC7456055 DOI: 10.1186/s12935-020-01513-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 08/21/2020] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Krüppel-like factor 8 (KLF8), a cancer-promoting factor that regulates critical gene transcription and cellular cancer-related events, has been implicated in tumor development and progression. However, the functional role of KLF8 in the pathogenesis of hepatocellular carcinoma (HCC) remains largely unknown. METHODS The gene expression patterns and genome-wide regulatory profiles of HCC cells after KLF8 knockout were analyzed by using RNA sequencing (RNA-seq) and chromatin immunoprecipitation sequencing (ChIP-seq) of histone H3 lysine 27 acetylation (H3K27ac) combined with bioinformatics analysis. Transcription factor-binding motifs that recognized by KLF8 were evaluated by motif analysis. For the predicted target genes, transcriptional changes were examined by ChIP, and loss of function experiments were conducted by siRNA transfection. RESULTS KLF8 functioned as a transcription repressor in HCC and mainly regulated apoptotic-related genes directly. A total of 1,816 differentially expressed genes after KLF8 knockout were identified and significantly corresponded to global changes in H3K27ac status. Furthermore, two predicted target genes, high-mobility group AT-hook 2 (HMGA2) and matrix metalloproteinase 7 (MMP7), were identified as important participants in KLF8-mediated anti-apoptotic effect in HCC. Knockout of KLF8 enhanced cell apoptosis process and caused increase in the associated H3K27ac, whereas suppression HMGA2 or MMP7 attenuated these biological effects. CONCLUSIONS Our work suggests a novel role and mechanism for KLF8 in the regulation of cell apoptosis in HCC and facilitates the discovery of potential therapeutic targets for HCC treatment.
Collapse
Affiliation(s)
- Ming-Da Wang
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University (Navy Medical University), No. 225, Changhai Road, Shanghai, 200438 China
| | - Hao Xing
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University (Navy Medical University), No. 225, Changhai Road, Shanghai, 200438 China
| | - Chao Li
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University (Navy Medical University), No. 225, Changhai Road, Shanghai, 200438 China
| | - Lei Liang
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University (Navy Medical University), No. 225, Changhai Road, Shanghai, 200438 China
| | - Han Wu
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University (Navy Medical University), No. 225, Changhai Road, Shanghai, 200438 China
| | - Xin-Fei Xu
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University (Navy Medical University), No. 225, Changhai Road, Shanghai, 200438 China
| | - Li-Yang Sun
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University (Navy Medical University), No. 225, Changhai Road, Shanghai, 200438 China
- Department of Clinical Medicine, Second Military Medical University (Navy Medical University), Shanghai, China
| | - Meng-Chao Wu
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University (Navy Medical University), No. 225, Changhai Road, Shanghai, 200438 China
| | - Feng Shen
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University (Navy Medical University), No. 225, Changhai Road, Shanghai, 200438 China
| | - Tian Yang
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University (Navy Medical University), No. 225, Changhai Road, Shanghai, 200438 China
| |
Collapse
|
23
|
Su CC, Ho WT, Peng FT, Gao CM, Jou TS, Wang IJ. Exploring a peptidomimetic approach of N-cadherin in modulating fibroblast growth factor receptor signaling for corneal endothelial regeneration. FASEB J 2020; 34:11698-11713. [PMID: 32654299 DOI: 10.1096/fj.201902525rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 06/14/2020] [Accepted: 06/17/2020] [Indexed: 11/11/2022]
Abstract
Endothelial rejection and a critical shortage of corneal transplants present an unmet medical need in corneal regeneration research area. Although basic fibroblast growth factor (bFGF) is a potent mitogenic factor for corneal ex vivo expansion, it is also a morphogen eliciting unfavorable endothelial-mesenchymal transition (EnMT) of corneal endothelial cells. A pharmacological reagent that retains the beneficial proliferative effect while lacking the EnMT effect of bFGF would be of great potential in corneal regeneration. In present study, we demonstrated that bFGF not only activated the canonical fibroblast growth factor receptor 1 (FGFR1) tyrosine kinase pathway, but also further upregulated matrix metalloproteinase activity to cleave N-cadherin into N-terminus and C-terminus fragments, which activated the classical FGFR1 tyrosine kinase pathway and a cryptic β-catenin pathway to affect corneal proliferation and EnMT, respectively. We generated the synthetic peptides resembling a critical motif in the ectodomain of N-cadherin and found these peptides enhanced downstream proliferative signaling of FGFR1 but without seemingly EnMT effect. The potential of these peptides can be demonstrated on both ex vivo cell culture and in vivo rat cryo-injury model. Our study indicated this peptidomimetic approach of N-cadherin can stimulate corneal regeneration and offer a promising therapeutic option to treat corneal endothelial dysfunction.
Collapse
Affiliation(s)
- Chien-Chia Su
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan R.O.C.,Institute of Clinical Medicine, National Taiwan University, Taipei, Taiwan R.O.C.,College of Medicine, National Taiwan University, Taipei, Taiwan R.O.C
| | - Wei-Ting Ho
- Department of Ophthalmology, Far Eastern Memorial Hospital, New Taipei City, Taiwan R.O.C
| | - Fu-Ti Peng
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan R.O.C
| | - Chia-Mao Gao
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan R.O.C
| | - Tzuu-Shuh Jou
- Institute of Clinical Medicine, National Taiwan University, Taipei, Taiwan R.O.C.,College of Medicine, National Taiwan University, Taipei, Taiwan R.O.C.,Center of Precision Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan R.O.C
| | - I-Jong Wang
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan R.O.C.,Institute of Clinical Medicine, National Taiwan University, Taipei, Taiwan R.O.C.,College of Medicine, National Taiwan University, Taipei, Taiwan R.O.C
| |
Collapse
|
24
|
Matrix Metalloproteinases as Biomarkers of Atherosclerotic Plaque Instability. Int J Mol Sci 2020; 21:ijms21113946. [PMID: 32486345 PMCID: PMC7313469 DOI: 10.3390/ijms21113946] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 05/27/2020] [Accepted: 05/29/2020] [Indexed: 02/07/2023] Open
Abstract
Matrix metalloproteinases (MMPs) are a family of zinc-dependent endopeptidases responsible for tissue remodeling and degradation of extracellular matrix (ECM) proteins. MMPs may modulate various cellular and signaling pathways in atherosclerosis responsible for progression and rupture of atherosclerotic plaques. The effect of MMPs polymorphisms and the expression of MMPs in both the atherosclerotic plaque and plasma was shown. They are independent predictors of atherosclerotic plaque instability in stable coronary heart disease (CHD) patients. Increased levels of MMPs in patients with advanced cardiovascular disease (CAD) and acute coronary syndrome (ACS) was associated with future risk of cardiovascular events. These data confirm that MMPs may be biomarkers in plaque instability as they target in potential drug therapies for atherosclerosis. They provide important prognostic information, independent of traditional risk factors, and may turn out to be useful in improving risk stratification.
Collapse
|
25
|
Cuvelliez M, Vandewalle V, Brunin M, Beseme O, Hulot A, de Groote P, Amouyel P, Bauters C, Marot G, Pinet F. Circulating proteomic signature of early death in heart failure patients with reduced ejection fraction. Sci Rep 2019; 9:19202. [PMID: 31844116 PMCID: PMC6914779 DOI: 10.1038/s41598-019-55727-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 12/03/2019] [Indexed: 12/11/2022] Open
Abstract
Heart failure (HF) remains a main cause of mortality worldwide. Risk stratification of patients with systolic chronic HF is critical to identify those who may benefit from advanced HF therapies. The aim of this study is to identify plasmatic proteins that could predict the early death (within 3 years) of HF patients with reduced ejection fraction hospitalized in CHRU de Lille. The subproteome targeted by an aptamer-based technology, the Slow Off-rate Modified Aptamer (SOMA) scan assay of 1310 proteins, was profiled in blood samples from 168 HF patients, and 203 proteins were significantly modulated between patients who died of cardiovascular death and patients who were alive after 3 years of HF evaluation (Wilcoxon test, FDR 5%). A molecular network was built using these 203 proteins, and the resulting network contained 2281 molecules assigned to 34 clusters annotated to biological pathways by Gene Ontology. This network model highlighted extracellular matrix organization as the main mechanism involved in early death in HF patients. In parallel, an adaptive Least Absolute Shrinkage and Selection Operator (LASSO) was performed on these 203 proteins, and six proteins were selected as candidates to predict early death in HF patients: complement C3, cathepsin S and F107B were decreased and MAPK5, MMP1 and MMP7 increased in patients who died of cardiovascular causes compared with patients living 3 years after HF evaluation. This proteomic signature of 6 circulating plasma proteins allows the identification of systolic HF patients with a risk of early death.
Collapse
Affiliation(s)
- Marie Cuvelliez
- Univ. Lille, CHU Lille, Inserm, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, F-59000, Lille, France.,FHU REMOD-HF, Lille, France
| | - Vincent Vandewalle
- Univ. Lille, CHU Lille, Inria Lille Nord-Europe, EA2694 - MODAL - MOdels for Data Analysis and Learning, F-59000, Lille, France.,Univ. Lille, « Institut Français de Bioinformatique », « Billille- plateforme de bioinformatique et bioanalyse de Lille », F-59000, Lille, France
| | - Maxime Brunin
- Univ. Lille, « Institut Français de Bioinformatique », « Billille- plateforme de bioinformatique et bioanalyse de Lille », F-59000, Lille, France
| | - Olivia Beseme
- Univ. Lille, CHU Lille, Inserm, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, F-59000, Lille, France.,FHU REMOD-HF, Lille, France
| | - Audrey Hulot
- Univ. Lille, « Institut Français de Bioinformatique », « Billille- plateforme de bioinformatique et bioanalyse de Lille », F-59000, Lille, France
| | - Pascal de Groote
- Univ. Lille, CHU Lille, Inserm, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, F-59000, Lille, France.,FHU REMOD-HF, Lille, France
| | - Philippe Amouyel
- Univ. Lille, CHU Lille, Inserm, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, F-59000, Lille, France
| | - Christophe Bauters
- Univ. Lille, CHU Lille, Inserm, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, F-59000, Lille, France.,FHU REMOD-HF, Lille, France
| | - Guillemette Marot
- Univ. Lille, CHU Lille, Inria Lille Nord-Europe, EA2694 - MODAL - MOdels for Data Analysis and Learning, F-59000, Lille, France.,Univ. Lille, « Institut Français de Bioinformatique », « Billille- plateforme de bioinformatique et bioanalyse de Lille », F-59000, Lille, France
| | - Florence Pinet
- Univ. Lille, CHU Lille, Inserm, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, F-59000, Lille, France. .,FHU REMOD-HF, Lille, France.
| |
Collapse
|
26
|
The Many Roles of Cell Adhesion Molecules in Hepatic Fibrosis. Cells 2019; 8:cells8121503. [PMID: 31771248 PMCID: PMC6952767 DOI: 10.3390/cells8121503] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 11/17/2019] [Accepted: 11/18/2019] [Indexed: 01/09/2023] Open
Abstract
Fibrogenesis is a progressive scarring event resulting from disrupted regular wound healing due to repeated tissue injury and can end in organ failure, like in liver cirrhosis. The protagonists in this process, either liver-resident cells or patrolling leukocytes attracted to the site of tissue damage, interact with each other by soluble factors but also by direct cell–cell contact mediated by cell adhesion molecules. Since cell adhesion molecules also support binding to the extracellular matrix, they represent excellent biosensors, which allow cells to modulate their behavior based on changes in the surrounding microenvironment. In this review, we focus on selectins, cadherins, integrins and members of the immunoglobulin superfamily of adhesion molecules as well as some non-classical cell adhesion molecules in the context of hepatic fibrosis. We describe their liver-specific contributions to leukocyte recruitment, cell differentiation and survival, matrix remodeling or angiogenesis and touch on their suitability as targets in antifibrotic therapies.
Collapse
|
27
|
Abstract
OBJECTIVE Chronic obstructive pulmonary disease (COPD) is a common chronic respiratory disease of human beings characterized by not fully reversible airflow limitation. Emphysema is the main pathological feature of COPD which causes high mortality worldwide every year and consumes a large amount of medical expenses. This paper was to review the establishment and evaluation methods of animal models of emphysema or COPD, and put forward some new ideas on animal selection, method of modeling, and model evaluation. DATA SOURCES The author retrieved information from the PubMed database up to July 2019, using various combinations of search terms, including emphysema, model, and animal. STUDY SELECTION Original articles, reviews, and other articles were searched and reviewed for animal models of emphysema. RESULTS This review summarized animal models of emphysema from the perspectives of animal selection, emphysema mechanism, modeling method and model evaluation, and found that passive smoking is the classic method for developing animal model of emphysema, mice are more suitable for experimental study on emphysema. Compared with pulmonary function indicators, airway inflammation indicators and oxidative stress indicators, pathomorphological indicators of lung tissue are the most important parameters for evaluating the establishment of the animal model of emphysema. CONCLUSIONS Mice model induced by passive smoking is the classic animal model of emphysema. Pathomorphological indicators are the most important parameters for evaluating the establishment of the animal model of emphysema.
Collapse
Affiliation(s)
- Gui-Bin Liang
- Department of Intensive Care Unit, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | | |
Collapse
|
28
|
Ezhov M, Safarova M, Afanasieva O, Mitroshkin M, Matchin Y, Pokrovsky S. Matrix Metalloproteinase 9 as a Predictor of Coronary Atherosclerotic Plaque Instability in Stable Coronary Heart Disease Patients with Elevated Lipoprotein(a) Levels. Biomolecules 2019; 9:biom9040129. [PMID: 30934954 PMCID: PMC6523150 DOI: 10.3390/biom9040129] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 03/24/2019] [Accepted: 03/26/2019] [Indexed: 11/22/2022] Open
Abstract
We sought to investigate whether levels of matrix metalloproteinases (MMPs) and their inhibitors predict coronary atherosclerotic plaque instability, as assessed by intravascular ultrasound (IVUS) virtual histology during coronary angiography. Blood samples were collected before angiography in 32 subjects (mean age 56 ± 8 years) with stable coronary heart disease (CHD) and elevated lipoprotein(a) (Lp(a), 94 ± 35 mg/dL). Levels of high-sensitivity C-reactive protein (hsCRP), apolipoprotein B100 (apoB100), MMP-7, MMP-9, tissue inhibitor of metalloproteinases (TIMP)-1, and TIMP-2 were determined using commercially available enzyme-linked immunosorbent assay kits. Results. The morphology of a total of sixty coronary lesions was assessed by virtual histology IVUS imaging. Eleven (18%) plaques in nine (28%) patients were classified as plaques with an unstable phenotype or a thin-cap fibroatheroma. Age, low-density lipoprotein cholesterol, apoB100, MMP-7, and MMP-9 levels were positively associated with necrotic core volume. Conversely, there was a negative relationship between MMP-7 and -9 levels and fibrous and fibro-fatty tissue volume. Multivariate regression analysis revealed that MMP-9 is a strong independent predictor of atherosclerotic plaque instability in stable CHD patients. In stable CHD patients with elevated Lp(a), MMP-9 levels are positively associated with the size of the necrotic core of coronary atherosclerotic plaques.
Collapse
Affiliation(s)
- Marat Ezhov
- Federal State Budget Institution "National Cardiology Research Center" of Ministry of Health of the Russian Federation; 15A, 3d Cherepkovskaya street, 121552 Moscow, Russia.
| | - Maya Safarova
- Federal State Budget Institution "National Cardiology Research Center" of Ministry of Health of the Russian Federation; 15A, 3d Cherepkovskaya street, 121552 Moscow, Russia.
| | - Olga Afanasieva
- Federal State Budget Institution "National Cardiology Research Center" of Ministry of Health of the Russian Federation; 15A, 3d Cherepkovskaya street, 121552 Moscow, Russia.
| | - Maksim Mitroshkin
- Federal State Budget Institution "National Cardiology Research Center" of Ministry of Health of the Russian Federation; 15A, 3d Cherepkovskaya street, 121552 Moscow, Russia.
| | - Yuri Matchin
- Federal State Budget Institution "National Cardiology Research Center" of Ministry of Health of the Russian Federation; 15A, 3d Cherepkovskaya street, 121552 Moscow, Russia.
| | - Sergei Pokrovsky
- Federal State Budget Institution "National Cardiology Research Center" of Ministry of Health of the Russian Federation; 15A, 3d Cherepkovskaya street, 121552 Moscow, Russia.
| |
Collapse
|
29
|
Asgari R, Mansouri K, Bakhtiari M, Vaisi-Raygani A. CD147 as an apoptosis regulator in spermatogenesis: deciphering its association with matrix metalloproteinases' pathway. Mol Biol Rep 2019; 46:1099-1105. [PMID: 30600459 DOI: 10.1007/s11033-018-4568-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 12/10/2018] [Indexed: 12/27/2022]
Abstract
CD147 plays an important role in germ cells migration and survival/apoptosis during the spermatogenesis process. However, to best of our knowledge, there is no report on the exact role of CD147 gene in the regulation of germ cells apoptosis through matrix metalloproteinases (MMPs). So, the current study aims to evaluate the role of CD147 gene expression in the regulation of germ cells apoptosis in conjunction with MMPs. Real-Time PCR was applied to investigate the expression of CD147, MMP2, MMP7, and MMP9 genes in the azoospermic patients and fertile males. Receiver-operating characteristic curve was used to interpret gene expression data. According to our results, a significant decrease in the expression of CD147 gene and an increase in MMPs genes expression were observed in infertile patients compared to fertile males. These results proved this fact that the CD147 gene has an important role in the regulation of germ cells apoptosis via a MMPs-dependent pathway.
Collapse
Affiliation(s)
- Rezvan Asgari
- Medical Biology Research Center (MBRC), Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Kamran Mansouri
- Medical Biology Research Center (MBRC), Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mitra Bakhtiari
- Department of Anatomical Sciences & Cell Biology, Kermanshah University of Medical Sciences, Kermanshah, Iran.
- Fertility and Sterility Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Asad Vaisi-Raygani
- Department of Clinical Biochemistry, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
30
|
Myasoedova VA, Chistiakov DA, Grechko AV, Orekhov AN. Matrix metalloproteinases in pro-atherosclerotic arterial remodeling. J Mol Cell Cardiol 2018; 123:159-167. [PMID: 30172754 DOI: 10.1016/j.yjmcc.2018.08.026] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 08/07/2018] [Accepted: 08/29/2018] [Indexed: 12/20/2022]
Abstract
Matrix metalloproteinases (MMPs) is a family of Zn2+ endopeptidases that process various components of the extracellular matrix. These enzymes are also involved in activation and inhibition of signaling cascades through proteolytic cleavage of surface receptors. Moreover, MMPs play a key role in tissue remodeling and and repair. Dysregulation of MMPs is observed in patholofgical conditions, including atherosclerosis, which is associated with hyperactivation of MMPs, aberrant tissue remodeling and neovascularization of the growing atherosclerotic plaques. This makes MMPs interesting therapeutic targets that can be employed for developing novel therapies to treat atherosclerosis and its complications. Currently, a growing number of synthetic MMP inhibitors is available. In this review, we will discuss the role of these enzymes in atherosclerosis pathology and the ways of their pothential therapeutic use.
Collapse
Affiliation(s)
- Veronika A Myasoedova
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 125315 Moscow, Russia
| | - Dimitry A Chistiakov
- Department of Basic and Applied Neurobiology, Serbsky Federal Medical Research Center for Psychiatry and Narcology, 119991 Moscow, Russia
| | - Andrey V Grechko
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 109240 Moscow, Russia
| | - Alexander N Orekhov
- Department of Basic and Applied Neurobiology, Serbsky Federal Medical Research Center for Psychiatry and Narcology, 119991 Moscow, Russia; Institute for Atherosclerosis Research, Skolkovo Innovative Center, 121609 Moscow, Russia.
| |
Collapse
|
31
|
Söderholm M, Nordin Fredrikson G, Nilsson J, Engström G. High Serum Level of Matrix Metalloproteinase-7 Is Associated With Increased Risk of Spontaneous Subarachnoid Hemorrhage. Stroke 2018; 49:1626-1631. [PMID: 29880550 DOI: 10.1161/strokeaha.118.020660] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 04/19/2018] [Accepted: 05/04/2018] [Indexed: 12/18/2022]
Abstract
BACKGROUND AND PURPOSE Increased degradation of the extracellular matrix in the arterial wall by matrix metalloproteinases (MMPs) may be an important mechanism in the pathogenesis of intracranial aneurysms and subarachnoid hemorrhage (SAH). MMP-2 and MMP-9 have been suggested to be involved in matrix degradation preceding SAH. We studied serum levels of MMP-1, -2, -3, -7, -9, -10, and -12 and the risk of incident SAH. METHODS A nested case-control study within the population-based cohort, Malmö Diet and Cancer study, was performed including incident cases of spontaneous SAH (n=79) and controls matched by age, sex, and follow-up time (n=232). MMPs were measured in serum from the baseline examination in 1991 to 1996. MMPs were compared between cases and controls, using conditional logistic regression adjusting for risk factors. RESULTS Baseline levels of MMP-7, MMP-10, and MMP-12 were significantly higher in incident SAH cases compared with controls. Odds ratios (95% confidence interval) for SAH per 1 SD increase of MMP-7, MMP-10, and MMP-12 were 1.78 (1.31-2.41), 1.45 (1.11-1.91), and 1.53 (1.17-2.01), respectively. After adjustment for SAH risk factors, MMP-7 was still significantly associated with SAH (odds ratio: 1.64; 95% confidence interval: 1.19-2.27; P=0.0026), whereas associations for MMP-10 and MMP-12 were attenuated and nonsignificant. We did not find any association between high serum levels of MMP-2 or MMP-9 and SAH risk. CONCLUSIONS High serum level of MMP-7 was associated with increased risk of incident spontaneous SAH, independently of the main risk factors for SAH. High serum levels of MMP-2 and MMP-9 did not predict SAH risk.
Collapse
Affiliation(s)
- Martin Söderholm
- From the Department of Clinical Sciences Malmö, Lund University, Sweden (M.S., G.N.F., J.N., G.E.) .,Department of Neurology and Rehabilitation Medicine, Skåne University Hospital, Malmö and Lund, Sweden (M.S.)
| | | | - Jan Nilsson
- From the Department of Clinical Sciences Malmö, Lund University, Sweden (M.S., G.N.F., J.N., G.E.)
| | - Gunnar Engström
- From the Department of Clinical Sciences Malmö, Lund University, Sweden (M.S., G.N.F., J.N., G.E.)
| |
Collapse
|
32
|
Frismantiene A, Philippova M, Erne P, Resink TJ. Cadherins in vascular smooth muscle cell (patho)biology: Quid nos scimus? Cell Signal 2018; 45:23-42. [DOI: 10.1016/j.cellsig.2018.01.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 01/23/2018] [Accepted: 01/23/2018] [Indexed: 12/16/2022]
|
33
|
Goikuria H, Vandenbroeck K, Alloza I. Inflammation in human carotid atheroma plaques. Cytokine Growth Factor Rev 2018; 39:62-70. [PMID: 29396056 DOI: 10.1016/j.cytogfr.2018.01.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 01/11/2018] [Indexed: 12/31/2022]
Abstract
Inflammation in carotid atherosclerotic plaque is linked to plaque rupture and cerebrovascular accidents. The balance between pro- and anti-inflammatory mediators governs development of the plaque, and may mediate enhancement of lesion broadening or, on the contrary, delay progression. In addition to macrophages and endothelial cells, smooth muscle cells (SMCs), which are the dominant cell subset in advanced plaques, are crucial players in carotid atherosclerosis development given their ability to differentiate into distinct phenotypes in reponse to specific signals received from the environment of the lesion. Carotid atheroma SMCs actively contribute to the inflammation in the lesion because of their acquired capacity to produce inflammatory mediators. We review the successive stages of carotid atheroma plaque formation via fatty streak early-stage toward more advanced rupture-prone lesions and document involvement of cytokines and chemokines and their cellular sources and targets in plaque progression and rupture.
Collapse
Affiliation(s)
- Haize Goikuria
- Neurogenomiks, Neuroscience Department, Faculty of Medicine and Odontology, Basque Country University (UPV/EHU), 48940 Leioa, Spain; ACHUCARRO, Basque Centre for Neuroscience, Science Park of the Basque Country University (UPV/EHU), SEDE Building, 3rd, 48940 Leioa, Spain
| | - Koen Vandenbroeck
- Neurogenomiks, Neuroscience Department, Faculty of Medicine and Odontology, Basque Country University (UPV/EHU), 48940 Leioa, Spain; ACHUCARRO, Basque Centre for Neuroscience, Science Park of the Basque Country University (UPV/EHU), SEDE Building, 3rd, 48940 Leioa, Spain; Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Spain
| | - Iraide Alloza
- Neurogenomiks, Neuroscience Department, Faculty of Medicine and Odontology, Basque Country University (UPV/EHU), 48940 Leioa, Spain; ACHUCARRO, Basque Centre for Neuroscience, Science Park of the Basque Country University (UPV/EHU), SEDE Building, 3rd, 48940 Leioa, Spain.
| |
Collapse
|
34
|
Yu H, Fellows A, Foote K, Yang Z, Figg N, Littlewood T, Bennett M. FOXO3a (Forkhead Transcription Factor O Subfamily Member 3a) Links Vascular Smooth Muscle Cell Apoptosis, Matrix Breakdown, Atherosclerosis, and Vascular Remodeling Through a Novel Pathway Involving MMP13 (Matrix Metalloproteinase 13). Arterioscler Thromb Vasc Biol 2018; 38:555-565. [PMID: 29326312 PMCID: PMC5828387 DOI: 10.1161/atvbaha.117.310502] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 01/02/2018] [Indexed: 01/02/2023]
Abstract
OBJECTIVE Vascular smooth muscle cell (VSMC) apoptosis accelerates atherosclerosis and promotes breakdown of the extracellular matrix, but the mechanistic links between these 2 processes are unknown. The forkhead protein FOXO3a (forkhead transcription factor O subfamily member 3a) is activated in human atherosclerosis and induces a range of proapoptotic and other transcriptional targets. We, therefore, determined the mechanisms and consequences of FOXO3a activation in atherosclerosis and arterial remodeling after injury. APPROACH AND RESULTS Expression of a conditional FOXO3a allele (FOXO3aA3ER) potently induced VSMC apoptosis, expression and activation of MMP13 (matrix metalloproteinase 13), and downregulation of endogenous TIMPs (tissue inhibitors of MMPs). mmp13 and mmp2 were direct FOXO3a transcriptional targets in VSMCs. Activation of endogenous FOXO3a also induced MMP13, extracellular matrix degradation, and apoptosis, and MMP13-specific inhibitors and fibronectin reduced FOXO3a-mediated apoptosis. FOXO3a activation in mice with VSMC-restricted FOXO3aA3ER induced MMP13 expression and activity and medial VSMC apoptosis. FOXO3a activation in FOXO3aA3ER/ApoE-/- (apolipoprotein E deficient) mice increased atherosclerosis, increased necrotic core and reduced fibrous cap areas, and induced features of medial degeneration. After carotid artery ligation, FOXO3a activation increased VSMC apoptosis, VSMC proliferation, and neointima formation, all of which were reduced by MMP13 inhibition. CONCLUSIONS FOXO3a activation induces VSMC apoptosis and extracellular matrix breakdown, in part, because of transcriptional activation of MMP13. FOXO3a activation promotes atherosclerosis and medial degeneration and increases neointima after injury that is partly dependent on MMP13. FOXO3a-induced MMP activation represents a direct mechanistic link between VSMC apoptosis and matrix breakdown in vascular disease.
Collapse
MESH Headings
- Animals
- Apoptosis
- Atherosclerosis/enzymology
- Atherosclerosis/genetics
- Atherosclerosis/pathology
- Carotid Artery Injuries/enzymology
- Carotid Artery Injuries/genetics
- Carotid Artery Injuries/pathology
- Cells, Cultured
- Disease Models, Animal
- Extracellular Matrix/enzymology
- Extracellular Matrix/pathology
- Fibrosis
- Forkhead Box Protein O3/genetics
- Forkhead Box Protein O3/metabolism
- Humans
- Male
- Matrix Metalloproteinase 13/genetics
- Matrix Metalloproteinase 13/metabolism
- Mice, Inbred C57BL
- Mice, Inbred CBA
- Mice, Knockout, ApoE
- Mice, Transgenic
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/pathology
- Mutation
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/pathology
- Necrosis
- Rats, Wistar
- Signal Transduction
- Transcriptional Activation
- Vascular Remodeling
Collapse
Affiliation(s)
- Haixiang Yu
- From the Division of Cardiovascular Medicine, Addenbrooke's Hospital (H.Y., A.F., K.F., N.F., M.B.) and Department of Biochemistry (T.L.), University of Cambridge, United Kingdom; and Institute of Medical Biology, Peking Union Medical College, Chinese Academy of Medical Sciences, Kunming, Yunnan Province, China (Z.Y.)
| | - Adam Fellows
- From the Division of Cardiovascular Medicine, Addenbrooke's Hospital (H.Y., A.F., K.F., N.F., M.B.) and Department of Biochemistry (T.L.), University of Cambridge, United Kingdom; and Institute of Medical Biology, Peking Union Medical College, Chinese Academy of Medical Sciences, Kunming, Yunnan Province, China (Z.Y.)
| | - Kirsty Foote
- From the Division of Cardiovascular Medicine, Addenbrooke's Hospital (H.Y., A.F., K.F., N.F., M.B.) and Department of Biochemistry (T.L.), University of Cambridge, United Kingdom; and Institute of Medical Biology, Peking Union Medical College, Chinese Academy of Medical Sciences, Kunming, Yunnan Province, China (Z.Y.)
| | - Zhaoqing Yang
- From the Division of Cardiovascular Medicine, Addenbrooke's Hospital (H.Y., A.F., K.F., N.F., M.B.) and Department of Biochemistry (T.L.), University of Cambridge, United Kingdom; and Institute of Medical Biology, Peking Union Medical College, Chinese Academy of Medical Sciences, Kunming, Yunnan Province, China (Z.Y.)
| | - Nichola Figg
- From the Division of Cardiovascular Medicine, Addenbrooke's Hospital (H.Y., A.F., K.F., N.F., M.B.) and Department of Biochemistry (T.L.), University of Cambridge, United Kingdom; and Institute of Medical Biology, Peking Union Medical College, Chinese Academy of Medical Sciences, Kunming, Yunnan Province, China (Z.Y.)
| | - Trevor Littlewood
- From the Division of Cardiovascular Medicine, Addenbrooke's Hospital (H.Y., A.F., K.F., N.F., M.B.) and Department of Biochemistry (T.L.), University of Cambridge, United Kingdom; and Institute of Medical Biology, Peking Union Medical College, Chinese Academy of Medical Sciences, Kunming, Yunnan Province, China (Z.Y.)
| | - Martin Bennett
- From the Division of Cardiovascular Medicine, Addenbrooke's Hospital (H.Y., A.F., K.F., N.F., M.B.) and Department of Biochemistry (T.L.), University of Cambridge, United Kingdom; and Institute of Medical Biology, Peking Union Medical College, Chinese Academy of Medical Sciences, Kunming, Yunnan Province, China (Z.Y.).
| |
Collapse
|
35
|
Aneurysm Severity is Increased by Combined Mmp-7 Deletion and N-cadherin Mimetic (EC4-Fc) Over-Expression. Sci Rep 2017; 7:17342. [PMID: 29229950 PMCID: PMC5725451 DOI: 10.1038/s41598-017-17700-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 11/28/2017] [Indexed: 11/24/2022] Open
Abstract
There is an unmet need for treatments to reduce abdominal aortic aneurysm (AAA) progression. Vascular smooth muscle cell (VSMC) apoptosis precipitates AAA formation, whereas VSMC proliferation repairs the vessel wall. We previously demonstrated that over-expression of EC4-Fc (truncated N-cadherin), or deletion of matrix-metalloproteinase-7 (Mmp-7) reduced VSMC apoptosis in mouse atherosclerotic plaques. Additionally, MMP-7 promotes VSMC apoptosis by cleavage of N-cadherin. We investigated their combined effect on AAA formation. Increased apoptosis and proliferation were observed in human AAA (HAAA) sections compared to normal aortae (HA). This coincided with increased MMP-7 activity and reduced N-cadherin protein levels in HAAA sections compared to HA. Using a mouse model of aneurysm formation, we showed that the combination of Mmp-7 deletion and EC4-Fc overexpression significantly increased AAA severity. Medial apoptosis and proliferation were both significantly reduced in these mice compared to control mice. In vitro, MMP-7 inhibition and EC4-Fc administration significantly supressed human aortic VSMC apoptosis (via activation of PI-3 kinase/Akt signalling) and proliferation. In conclusion, combined Mmp-7 deletion and systemic over-expression of EC4-Fc reduced both proliferation and apoptosis. Reduced proliferation-mediated repair over-rides any benefit of reduced apoptosis, increasing aneurysm severity. Future studies should therefore focus on retarding VSMC apoptosis whilst promoting VSMC proliferation.
Collapse
|
36
|
Metalloproteinases in atherosclerosis. Eur J Pharmacol 2017; 816:93-106. [DOI: 10.1016/j.ejphar.2017.09.007] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 08/31/2017] [Accepted: 09/08/2017] [Indexed: 11/20/2022]
|
37
|
Nadanaka S, Kinouchi H, Kitagawa H. Chondroitin sulfate-mediated N-cadherin/β-catenin signaling is associated with basal-like breast cancer cell invasion. J Biol Chem 2017; 293:444-465. [PMID: 29183998 DOI: 10.1074/jbc.m117.814509] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 11/20/2017] [Indexed: 12/20/2022] Open
Abstract
Tumor metastasis involves cancer cell invasion across basement membranes and interstitial tissues. The initial invasion step consists of adherence of the tumor cell to the extracellular matrix (ECM), and this binding transduces a variety of signals from the ECM to the tumor cell. Accordingly, it is critical to establish the mechanisms by which extracellular cues influence the intracellular activities that regulate tumor cell invasion. Here, we found that invasion of the basal-like breast cancer cell line BT-549 is enhanced by the ECM component chondroitin sulfates (CSs). CSs interacted with and induced proteolytic cleavage of N-cadherin in the BT-549 cells, yielding a C-terminal intracellular N-cadherin fragment that formed a complex with β-catenin. Of note, the cleavage of N-cadherin increased cytoplasmic and nuclear β-catenin levels; induced the matrix metalloproteinase 9 (MMP9) gene, a target of β-catenin nuclear signaling; and augmented the invasion potential of the cells. We also found that CS-induced N-cadherin proteolysis requires caveolae-mediated endocytosis. An inhibitor of that process, nystatin, blocked both the endocytosis and proteolytic cleavage of N-cadherin induced by CS and also suppressed BT-549 cell invasion. Knock-out of chondroitin 4-O-sulfotransferase-1 (C4ST-1), a key CS biosynthetic enzyme, suppressed activation of the N-cadherin/β-catenin pathway through N-cadherin endocytosis and significantly decreased BT-549 cell invasion. These results suggest that CSs produced by C4ST-1 might be useful therapeutic targets in the management of basal-like breast cancers.
Collapse
Affiliation(s)
- Satomi Nadanaka
- From the Department of Biochemistry, Kobe Pharmaceutical University, Higashinada-ku, Kobe 658-8558, Japan
| | - Hiroki Kinouchi
- From the Department of Biochemistry, Kobe Pharmaceutical University, Higashinada-ku, Kobe 658-8558, Japan
| | - Hiroshi Kitagawa
- From the Department of Biochemistry, Kobe Pharmaceutical University, Higashinada-ku, Kobe 658-8558, Japan
| |
Collapse
|
38
|
Conant K, Daniele S, Bozzelli PL, Abdi T, Edwards A, Szklarczyk A, Olchefske I, Ottenheimer D, Maguire-Zeiss K. Matrix metalloproteinase activity stimulates N-cadherin shedding and the soluble N-cadherin ectodomain promotes classical microglial activation. J Neuroinflammation 2017; 14:56. [PMID: 28302163 PMCID: PMC5356362 DOI: 10.1186/s12974-017-0827-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 02/27/2017] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Matrix metalloproteinases (MMPs) are a family of enzymes that are typically released from intracellular stores to act on specific extracellular substrates. MMP expression and activity can be increased in a neuronal activity-dependent manner, and further increased in response to tissue injury. MMP substrates include cell adhesion molecules (CAMs) that are abundantly expressed in the brain and well positioned for membrane proximal cleavage. Importantly, CAM integrity is important to synaptic structure and axon-myelin interactions, and shed ectodomains may themselves influence cellular function. METHODS In the present study, we have examined proteolysis of N-cadherin (N-cdh) by MMP-7, a family member that has been implicated in disorders including HIV dementia, multiple sclerosis, and major depression. With in vitro digest assays, we tested N-cdh cleavage by increasing concentrations of recombinant enzyme. We also tested MMP-7 for its potential to stimulate N-cdh shedding from cultured neural cells. Since select CAM ectodomains may interact with cell surface receptors that are expressed on microglial cells, we subsequently tested the N-cdh ectodomain for its ability to stimulate activation of this cell type as determined by nuclear translocation of NF-κB, Iba-1 expression, and TNF-α release. RESULTS We observed that soluble N-cdh increased Iba-1 levels in microglial lysates, and also increased microglial release of the cytokine TNF-α. Effects were associated with increased NF-κB immunoreactivity in microglial nuclei and diminished by an inhibitor of the toll-like receptor adaptor protein, MyD88. CONCLUSIONS Together, these in vitro results suggest that soluble N-cdh may represent a novel effector of microglial activation, and that disorders with increased MMP levels may stimulate a cycle in which the products of excess proteolysis further exacerbate microglial-mediated tissue injury. Additional in vivo studies are warranted to address this issue.
Collapse
Affiliation(s)
- Katherine Conant
- Department of Neuroscience, Georgetown University School of Medicine, Washington, D.C., USA
| | - Stefano Daniele
- Department of Neuroscience, Georgetown University School of Medicine, Washington, D.C., USA
| | - P. Lorenzo Bozzelli
- Department of Neuroscience, Georgetown University School of Medicine, Washington, D.C., USA
| | - Tsion Abdi
- Department of Neuroscience, Georgetown University School of Medicine, Washington, D.C., USA
| | - Amanda Edwards
- Department of Neuroscience, Georgetown University School of Medicine, Washington, D.C., USA
| | | | - India Olchefske
- Department of Neuroscience, Georgetown University School of Medicine, Washington, D.C., USA
| | - David Ottenheimer
- Department of Neuroscience, Georgetown University School of Medicine, Washington, D.C., USA
| | - Kathleen Maguire-Zeiss
- Department of Neuroscience, Georgetown University School of Medicine, Washington, D.C., USA
| |
Collapse
|
39
|
Brown BA, Williams H, George SJ. Evidence for the Involvement of Matrix-Degrading Metalloproteinases (MMPs) in Atherosclerosis. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 147:197-237. [PMID: 28413029 DOI: 10.1016/bs.pmbts.2017.01.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Atherosclerosis leads to blockage of arteries, culminating in myocardial infarction, and stroke. The involvement of matrix-degrading metalloproteinases (MMPs) in atherosclerosis is established and many studies have highlighted the importance of various MMPs in this process. MMPs were first implicated in atherosclerosis due to their ability to degrade extracellular matrix components, which can lead to increased plaque instability. However, more recent work has highlighted a multitude of roles for MMPs in addition to breakdown of extracellular matrix proteins. MMPs are now known to be involved in various stages of plaque progression: from initial macrophage infiltration to plaque rupture. This chapter summarizes the development and progression of atherosclerotic plaques and the contribution of MMPs. We provide data from human studies showing the effect of MMP polymorphisms and the expression of MMPs in both the atherosclerotic plaque and within plasma. We also discuss work in animal models of atherosclerosis that show the effect of gain or loss of function of MMPs. Together, the data provided from these studies illustrate that MMPs are ideal targets as both biomarkers and potential drug therapies for atherosclerosis.
Collapse
Affiliation(s)
- Bethan A Brown
- School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
| | - Helen Williams
- School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
| | - Sarah J George
- School of Clinical Sciences, University of Bristol, Bristol, United Kingdom.
| |
Collapse
|
40
|
Ruddy JM, Ikonomidis JS, Jones JA. Multidimensional Contribution of Matrix Metalloproteinases to Atherosclerotic Plaque Vulnerability: Multiple Mechanisms of Inhibition to Promote Stability. J Vasc Res 2016; 53:1-16. [PMID: 27327039 PMCID: PMC7196926 DOI: 10.1159/000446703] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 05/07/2016] [Indexed: 12/17/2022] Open
Abstract
The prevalence of atherosclerotic disease continues to increase, and despite significant reductions in major cardiovascular events with current medical interventions, an additional therapeutic window exists. Atherosclerotic plaque growth is a complex integration of cholesterol penetration, inflammatory cell infiltration, vascular smooth muscle cell (VSMC) migration, and neovascular invasion. A family of matrix-degrading proteases, the matrix metalloproteinases (MMPs), contributes to all phases of vascular remodeling. The contribution of specific MMPs to endothelial cell integrity and VSMC migration in atherosclerotic lesion initiation and progression has been confirmed by the increased expression of these proteases in plasma and plaque specimens. Endogenous blockade of MMPs by the tissue inhibitors of metalloproteinases (TIMPs) may attenuate proteolysis in some regions, but the progression of matrix degeneration suggests that MMPs predominate in atherosclerotic plaque, precipitating vulnerability. Plaque neovascularization also contributes to instability and, coupling the known role of MMPs in angiogenesis to that of atherosclerotic plaque growth, interest in targeting MMPs to facilitate plaque stabilization continues to accumulate. This article aims to review the contributions of MMPs and TIMPs to atherosclerotic plaque expansion, neovascularization, and rupture vulnerability with an interest in promoting targeted therapies to improve plaque stabilization and decrease the risk of major cardiovascular events.
Collapse
Affiliation(s)
- Jean Marie Ruddy
- Division of Vascular Surgery, Department of Surgery, Medical University of South Carolina, Charleston, S.C., USA
| | | | | |
Collapse
|
41
|
Di Gregoli K, George SJ, Jackson CL, Newby AC, Johnson JL. Differential effects of tissue inhibitor of metalloproteinase (TIMP)-1 and TIMP-2 on atherosclerosis and monocyte/macrophage invasion. Cardiovasc Res 2016; 109:318-30. [PMID: 26645981 PMCID: PMC4724937 DOI: 10.1093/cvr/cvv268] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 11/29/2015] [Indexed: 12/30/2022] Open
Abstract
AIMS MMPs contribute to atherosclerotic plaque progression and instability, but the relative potency of their endogenous tissue inhibitors of metalloproteinases (TIMPs) as protective factors has not been defined. We therefore investigated the impact of TIMP-1 and TIMP-2 knockout on atherosclerotic plaque burden and composition in apolipoprotein E-knockout (Apoe(-/-)) mice and studied the underlying effects on monocyte/macrophage behaviour. METHODS AND RESULTS Analysis of brachiocephalic artery plaques revealed comparable atherosclerotic lesion areas between TIMP-1(-/-) Apoe(-/-) or TIMP-2(-/-) Apoe(-/-) double deficient mice and relevant age-matched, strain-matched Apoe(-/-) controls after 8 weeks of high-fat feeding. However, lesions from TIMP-2(-/-) Apoe(-/-) mice had higher levels of markers associated with plaque vulnerability, including increased macrophage: vascular smooth muscle cell ratios, larger necrotic core areas, reduced collagen contents, increased macrophage proliferation, and apoptosis frequencies, compared with TIMP-1(-/-)Apoe(-/-) and controls. In contrast, TIMP-1(-/-) Apoe(-/-) animals only had a significant reduction in vascular smooth muscle cell content compared with Apoe(-/-) controls. In vitro and in vivo findings implicated heightened monocyte/macrophage invasion in the detrimental effects observed on atherosclerotic plaque composition in TIMP-2(-/-) Apoe(-/-) mice. Moreover, TIMP-2 specifically decreased MMP-14-dependent monocyte/macrophage infiltration into sites of experimentally induced inflammation and established atherosclerotic lesions. CONCLUSION Our data demonstrate that TIMP-2 plays a greater protective role than TIMP-1 during the pathogenesis of atherosclerosis, in part by suppressing MMP-14-dependent monocyte/macrophage accumulation into plaques.
Collapse
|
42
|
|
43
|
Aspirin Inhibits LPS-Induced Expression of PI3K/Akt, ERK, NF-κB, CX3CL1, and MMPs in Human Bronchial Epithelial Cells. Inflammation 2015; 39:643-50. [DOI: 10.1007/s10753-015-0289-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
44
|
Afkarian M, Zelnick LR, Ruzinski J, Kestenbaum B, Himmelfarb J, de Boer IH, Mehrotra R. Urine matrix metalloproteinase-7 and risk of kidney disease progression and mortality in type 2 diabetes. J Diabetes Complications 2015; 29:1024-31. [PMID: 26412030 PMCID: PMC5389898 DOI: 10.1016/j.jdiacomp.2015.08.024] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 08/25/2015] [Accepted: 08/30/2015] [Indexed: 02/06/2023]
Abstract
AIMS The renin-angiotensin-aldosterone system (RAAS), bone morphogenetic protein (BMP) and WNT pathways are dysregulated in diabetic kidney disease (DKD). Urine excretion of angiotensinogen, gremlin-1 and matrix metalloproteinase-7 (MMP-7), components of the RAAS, BMP and WNT pathways, respectively, is increased in DKD. We asked if this increase is associated with subsequent progression to end-stage renal disease (ESRD) or death. METHODS Using time-to-event analyses, we examined the association of baseline urine concentration of these proteins with progression to ESRD or death in a predominantly Mexican-American cohort with type 2 diabetes and proteinuric DKD (n=141). RESULTS Progression to ESRD occurred for 38 participants over a median follow-up of 3.0years; 39 participants died over a median follow-up of 3.6years. Urine MMP-7 and gremlin-1 were associated with increased risk of ESRD after adjustment for demographic and clinical covariates. Angiotensinogen showed a U-shaped relationship with ESRD, with the middle tertile associated with lowest risk of ESRD. After additional adjustment for glomerular filtration rate and albuminuria, all associations with ESRD lost significance. Only urine MMP-7 was associated with mortality, and this association remained robust in the fully adjusted model with a Hazard ratio of 3.59 (95% confidence interval 1.31 to 9.85) for highest vs. lowest tertile. Serum MMP-7 was not associated with mortality and did not attenuate the association of urine MMP-7 with mortality (HR 4.03 for highest vs. lowest urine MMP-7 tertile). CONCLUSIONS Among people with type 2 diabetes and proteinuric DKD, urine MMP-7 concentration was strongly associated with subsequent mortality.
Collapse
MESH Headings
- Aged
- Cohort Studies
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/ethnology
- Diabetes Mellitus, Type 2/mortality
- Diabetes Mellitus, Type 2/urine
- Diabetic Nephropathies/complications
- Diabetic Nephropathies/epidemiology
- Diabetic Nephropathies/mortality
- Diabetic Nephropathies/physiopathology
- Disease Progression
- Female
- Follow-Up Studies
- Glomerular Filtration Rate
- Hospitals, Public
- Hospitals, Urban
- Humans
- Kidney/physiopathology
- Kidney Failure, Chronic/complications
- Kidney Failure, Chronic/epidemiology
- Kidney Failure, Chronic/mortality
- Kidney Failure, Chronic/physiopathology
- Los Angeles/epidemiology
- Male
- Matrix Metalloproteinase 7/urine
- Mexican Americans
- Middle Aged
- Prospective Studies
- Renal Insufficiency/complications
- Renal Insufficiency/epidemiology
- Renal Insufficiency/mortality
- Renal Insufficiency/physiopathology
- Risk
- Up-Regulation
Collapse
Affiliation(s)
- Maryam Afkarian
- Kidney Research Institute and Division of Nephrology, Department of Medicine, University of Washington, Seattle, WA.
| | - Leila R Zelnick
- Kidney Research Institute and Division of Nephrology, Department of Medicine, University of Washington, Seattle, WA; Department of Biostatistics, University of Washington
| | - John Ruzinski
- Kidney Research Institute and Division of Nephrology, Department of Medicine, University of Washington, Seattle, WA
| | - Bryan Kestenbaum
- Kidney Research Institute and Division of Nephrology, Department of Medicine, University of Washington, Seattle, WA
| | - Jonathan Himmelfarb
- Kidney Research Institute and Division of Nephrology, Department of Medicine, University of Washington, Seattle, WA
| | - Ian H de Boer
- Kidney Research Institute and Division of Nephrology, Department of Medicine, University of Washington, Seattle, WA
| | - Rajnish Mehrotra
- Kidney Research Institute and Division of Nephrology, Department of Medicine, University of Washington, Seattle, WA
| |
Collapse
|
45
|
Conant K, Allen M, Lim ST. Activity dependent CAM cleavage and neurotransmission. Front Cell Neurosci 2015; 9:305. [PMID: 26321910 PMCID: PMC4531370 DOI: 10.3389/fncel.2015.00305] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 07/27/2015] [Indexed: 12/13/2022] Open
Abstract
Spatially localized proteolysis represents an elegant means by which neuronal activity dependent changes in synaptic structure, and thus experience dependent learning and memory, can be achieved. In vitro and in vivo studies suggest that matrix metalloproteinase and adamalysin activity is concentrated at the cell surface, and emerging evidence suggests that increased peri-synaptic expression, release and/or activation of these proteinases occurs with enhanced excitatory neurotransmission. Synaptically expressed cell adhesion molecules (CAMs) could therefore represent important targets for neuronal activity-dependent proteolysis. Several CAM subtypes are expressed at the synapse, and their cleavage can influence the efficacy of synaptic transmission through a variety of non-mutually exclusive mechanisms. In the following review, we discuss mechanisms that regulate neuronal activity-dependent synaptic CAM shedding, including those that may be calcium dependent. We also highlight CAM targets of activity-dependent proteolysis including neuroligin and intercellular adhesion molecule-5 (ICAM-5). We include discussion focused on potential consequences of synaptic CAM shedding, with an emphasis on interactions between soluble CAM cleavage products and specific pre- and post-synaptic receptors.
Collapse
Affiliation(s)
- Katherine Conant
- Department of Neuroscience and Interdisciplinary Program in Neuroscience, Georgetown University Medical Center Washington, DC, USA
| | - Megan Allen
- Department of Neuroscience and Interdisciplinary Program in Neuroscience, Georgetown University Medical Center Washington, DC, USA
| | - Seung T Lim
- Department of Neuroscience and Interdisciplinary Program in Neuroscience, Georgetown University Medical Center Washington, DC, USA
| |
Collapse
|
46
|
Ho WT, Chang JS, Su CC, Chang SW, Hu FR, Jou TS, Wang IJ. Inhibition of Matrix Metalloproteinase Activity Reverses Corneal Endothelial-Mesenchymal Transition. THE AMERICAN JOURNAL OF PATHOLOGY 2015. [DOI: 10.1016/j.ajpath.2015.04.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
47
|
Goncalves I, Bengtsson E, Colhoun HM, Shore AC, Palombo C, Natali A, Edsfeldt A, Dunér P, Fredrikson GN, Björkbacka H, Östling G, Aizawa K, Casanova F, Persson M, Gooding K, Strain D, Khan F, Looker HC, Adams F, Belch J, Pinnoli S, Venturi E, Kozakova M, Gan LM, Schnecke V, Nilsson J. Elevated Plasma Levels of MMP-12 Are Associated With Atherosclerotic Burden and Symptomatic Cardiovascular Disease in Subjects With Type 2 Diabetes. Arterioscler Thromb Vasc Biol 2015; 35:1723-31. [PMID: 25953645 DOI: 10.1161/atvbaha.115.305631] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 04/22/2015] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Matrix metalloproteinases (MMPs) degrade extracellular matrix proteins and play important roles in development and tissue repair. They have also been shown to have both protective and pathogenic effects in atherosclerosis, and experimental studies have suggested that MMP-12 contributes to plaque growth and destabilization. The objective of this study was to investigate the associations between circulating MMPs, atherosclerosis burden, and incidence of cardiovascular disease with a particular focus on type 2 diabetes mellitus. APPROACH AND RESULTS Plasma levels of MMP-1, -3, -7, -10, and -12 were analyzed by the Proximity Extension Assay technology in 1500 subjects participating in the SUMMIT (surrogate markers for micro- and macrovascular hard end points for innovative diabetes tools) study, 384 incident coronary cases, and 409 matched controls in the Malmö Diet and Cancer study and in 205 carotid endarterectomy patients. Plasma MMP-7 and -12 were higher in subjects with type 2 diabetes mellitus, increased with age and impaired renal function, and was independently associated with prevalent cardiovascular disease, atherosclerotic burden (as assessed by carotid intima-media thickness and ankle-brachial pressure index), arterial stiffness, and plaque inflammation. Baseline MMP-7 and -12 levels were increased in Malmö Diet and Cancer subjects who had a coronary event during follow-up. CONCLUSIONS The plasma level of MMP-7 and -12 are elevated in type 2 diabetes mellitus, associated with more severe atherosclerosis and an increased incidence of coronary events. These observations provide clinical support to previous experimental studies, demonstrating a role for these MMPs in plaque development, and suggest that they are potential biomarkers of atherosclerosis burden and cardiovascular disease risk.
Collapse
Affiliation(s)
- Isabel Goncalves
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Eva Bengtsson
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Helen M Colhoun
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Angela C Shore
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Carlo Palombo
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Andrea Natali
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Andreas Edsfeldt
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Pontus Dunér
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Gunilla Nordin Fredrikson
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Harry Björkbacka
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Gerd Östling
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Kunihiko Aizawa
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Francesco Casanova
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Margaretha Persson
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Kim Gooding
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - David Strain
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Faisel Khan
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Helen C Looker
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Fiona Adams
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Jill Belch
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Silvia Pinnoli
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Elena Venturi
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Michaela Kozakova
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Li-Ming Gan
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Volker Schnecke
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Jan Nilsson
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.).
| | | |
Collapse
|
48
|
Drebert Z, Bracke M, Beck IM. Glucocorticoids and the non-steroidal selective glucocorticoid receptor modulator, compound A, differentially affect colon cancer-derived myofibroblasts. J Steroid Biochem Mol Biol 2015; 149:92-105. [PMID: 25666906 DOI: 10.1016/j.jsbmb.2015.02.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 01/27/2015] [Accepted: 02/04/2015] [Indexed: 12/21/2022]
Abstract
The glucocorticoid receptor functions as a ligand-dependent transcription factor that positively or negatively regulates the transcription of various specific target genes. Not only steroidal glucocorticoids can bind and activate the glucocorticoid receptor, but also the intensively examined non-steroidal selective glucocorticoid receptor modulators can do so, albeit with a select effector profile skewed to glucocorticoid receptor transrepression. Glucocorticoids are widely used to treat inflammatory afflictions, but also as anti-cancer therapies or adjuvants thereof. As the impact of glucocorticoids and selective glucocorticoid receptor modulators has scarcely been researched in this setting, we focused on colon cancer and its stromal environment, in particular the stromal myofibroblasts, which are known to influence cancer cells via paracrine signaling. In these myofibroblasts, the glucocorticoid dexamethasone is able to drive the glucocorticoid receptor into the nucleus and thus negatively regulates the expression of particular pro-inflammatory genes in TNFα-stimulated cells. The selective glucocorticoid receptor modulator compound A has an impaired ability to translocate GR, presumably underpinning its modest anti-inflammatory properties in these cells. Only dexamethasone, and not compound A, can upregulate the glucocorticoid receptor transactivation-dependent GILZ expression. Neither dexamethasone, nor compound A affects myofibroblast cell viability. However, compound A retards the growth of this myofibroblast cell line. Additionally, dexamethasone can inhibit the expression of Tenascin C, hepatocyte growth factor, and TGFβ, which are all factors known for their impact on colon cancer cell invasion, in a glucocorticoid receptor-dependent manner. In contrast, compound A can only slightly diminish the expression of just hepatocyte growth factor, and not tenascin C or TGFβ. Combined, our results expose new tumor microenvironment-modulating effects of glucocorticoids and the selective GR modulator compound A.
Collapse
Affiliation(s)
- Zuzanna Drebert
- Laboratory of Experimental Cancer Research, Department of Radiation Oncology & Experimental Cancer Research, Ghent University, Gent, Belgium
| | - Marc Bracke
- Laboratory of Experimental Cancer Research, Department of Radiation Oncology & Experimental Cancer Research, Ghent University, Gent, Belgium
| | - Ilse M Beck
- Laboratory of Experimental Cancer Research, Department of Radiation Oncology & Experimental Cancer Research, Ghent University, Gent, Belgium.
| |
Collapse
|
49
|
Polyakov LM, Lushnikova EL, Karpova AA, Russkikh GS, Poteryaeva ON, Nikityuk DB, Nepomnyashchikh LM, Belonogova ZI, Sumenkova DV, Nepomnyashchikh RD, Pichigin VI. Content of circulating extracellular DNA, plasma activities of matrix metalloproteinases, and ultrastructure of the myocardium in hypothyroid rats with hypercholesterolemia. Bull Exp Biol Med 2015; 158:632-7. [PMID: 25778649 DOI: 10.1007/s10517-015-2824-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Indexed: 11/29/2022]
Abstract
Free circulating extracellular DNA, plasma activities of matrix metalloproteinases in hypothyroid rats, and ultrastructural changes in the myocardium were studied under conditions of experimental hypercholesterolemia. For suppression of thyroid function, the animals received antithyroid drug mercazolyl under conditions of cholesterol diet. Hypercholesterolemia in hypothyroid rats (thyroxine concentration 2-fold below the normal) was paralleled by a pronounced increase of the concentration of free circulating extracellular DNA and total matrix metalloproteinases 2 and 7 activity. These changes were associated with lytic and destructive changes in cardiomyocytes and blood capillary endotheliocytes. Changes in the cardiomyocyte and endotheliocyte ultrastructure were more pronounced in hypothyroid rats.
Collapse
Affiliation(s)
- L M Polyakov
- Research Institute of Biochemistry, Siberian Division of the Russian Academy of Medical Sciences, Novosibirsk, Russia,
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Erbel C, Wolf A, Lasitschka F, Linden F, Domschke G, Akhavanpoor M, Doesch AO, Katus HA, Gleissner CA. Prevalence of M4 macrophages within human coronary atherosclerotic plaques is associated with features of plaque instability. Int J Cardiol 2015; 186:219-25. [PMID: 25828120 DOI: 10.1016/j.ijcard.2015.03.151] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 02/05/2015] [Accepted: 03/15/2015] [Indexed: 01/08/2023]
Abstract
BACKGROUND The platelet chemokine CXCL4 induces monocyte differentiation resulting in a macrophage phenotype called "M4", which co-expresses CD68, MMP7, and S100A8. We hypothesized that M4 macrophages are associated with plaque destabilization. METHODS Atherosclerotic arteries were obtained from explanted hearts of patients with severe coronary artery disease (CAD, n = 32) and of patients with dilated cardiomyopathy and no or mild CAD (controls, n = 19). Coronary arteries were stained with H&E, and immuno-fluorescence was performed against CD68, MMP7, and S100A8. RESULTS Both CD68(+) macrophages representing the entire macrophage population and MMP7(+)S100A8(+)CD68(+) M4 macrophages could be reproducibly identified within all arterial layers. The average proportion of the M4 macrophage phenotype amongst all CD68(+) macrophages was 31.7 ± 16.2%. The highest number of M4 macrophages was found in the adventitia, followed by the intima. CD68(+) and M4 macrophage numbers were significantly higher in patients with severe CAD. The presence of M4 macrophages within the intima and the media was significantly associated with plaque instability as determined by Stary class. Multivariate analysis showed a highly significant contribution of cardiovascular risk factors (P = 0.008) to plaque instability, while only trends were observed for age (P = 0.060) and intimal prevalence of M4 macrophages (P = 0.098). CONCLUSIONS We demonstrate for the first time that M4 macrophages can be reproducibly found in coronary artery plaques. The prevalence of M4 macrophages is associated with indexes of plaque instability, most likely representing a surrogate marker of inflammatory activity. These findings suggest a pathogenetic role of M4 macrophages in vulnerable atherosclerotic plaques.
Collapse
Affiliation(s)
- Christian Erbel
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg, Germany
| | - Antonia Wolf
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany
| | - Felix Lasitschka
- Institute of Pathology, University of Heidelberg, Heidelberg, Germany
| | - Fabian Linden
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany
| | - Gabriele Domschke
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany
| | | | - Andreas O Doesch
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg, Germany
| | - Hugo A Katus
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg, Germany
| | - Christian A Gleissner
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg, Germany.
| |
Collapse
|