1
|
Fiorotti AM, Gomes ACA, Bortoli AM, de Brito BB, Nunes KZ, Haraguchi FK, Bolsoni-Lopes A. Dynamic Changes in Adiponectin and Resistin Drive Remission of Cardiometabolic Risk Biomarkers in Individuals with Obesity Following Bariatric Surgery. Pharmaceuticals (Basel) 2024; 17:215. [PMID: 38399430 PMCID: PMC10893494 DOI: 10.3390/ph17020215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/17/2024] [Accepted: 01/22/2024] [Indexed: 02/25/2024] Open
Abstract
The remission of obesity-related diseases following bariatric surgery appears to result from the reorganization of metabolic and hormonal pathways involving adipokines. This study aimed to investigate the relationship between changes in body adiposity and serum adipokine levels, as well as the association between variations in adiponectin or resistin levels and cardiometabolic risk blood biomarkers before and after Roux-en-Y gastric bypass. A longitudinal and prospective study was conducted with bariatric surgery patients. Anthropometric, body composition and blood biochemical parameters were measured before and at 2 and 6 months post-surgery. The data were analyzed using ANOVA, Pearson or Spearman correlation, and simple linear regression with a significance level of p < 0.05. Among 36 mostly female patients aged 30 to 39 years, significant reductions in body weight (-26.8%), fat mass (-50%), waist circumference (-18%) and waist-to-height ratio (-22%) were observed post-surgery. Serum adiponectin levels increased (+107%), while resistin (-12.2%), TNF-α (-35%), and PAI-1 (-11.1%) decreased. Glucose, insulin, CRP, cholesterol, LDL-c, triglycerides, and vitamin D also decreased. Waist circumference variation showed a positive correlation with PAI-1 and TNF-α and a negative correlation with adiponectin. The total fat mass showed a positive correlation with PAI-1. Adiponectin variation correlated negatively with glucose, resistin, and CRP but positively with HDL-c. Resistin showed a positive correlation with insulin and CRP. In conclusion, 6 months post-bariatric surgery, reducing abdominal adiposity had a more significant impact on serum adipokine levels than total fat mass. Adiponectin increase and resistin decrease acted as endocrine mediators driving the remission of cardiometabolic risk biomarkers in individuals with obesity following Roux-en-Y gastric bypass.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Andressa Bolsoni-Lopes
- Postgraduate Program in Nutrition and Health, Health Sciences Center, Federal University of Espirito Santo, Vitoria 29047-105, Brazil (A.C.A.G.); (A.M.B.); (B.B.d.B.); (K.Z.N.); (F.K.H.)
| |
Collapse
|
2
|
Prater MC, Scheurell AR, Paton CM, Cooper JA. No Observed Difference in Inflammatory and Coagulation Markers Following Diets Rich in n-6 Polyunsaturated Fat vs Monounsaturated Fat in Adults With Untreated Hypercholesterolemia: A Randomized Trial. J Acad Nutr Diet 2024; 124:205-214.e1. [PMID: 37619782 DOI: 10.1016/j.jand.2023.08.127] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 08/08/2023] [Accepted: 08/17/2023] [Indexed: 08/26/2023]
Abstract
BACKGROUND Inflammatory and prothrombotic responses are hallmark to the progression of cardiovascular disease and may be influenced by the type of dietary fat. Cottonseed oil (CSO) is rich in n-6 polyunsaturated fats and improves traditional cardiovascular disease risk factors such as cholesterol profiles. However, some clinicians are still hesitant to promote n-6 polyunsaturated fats consumption despite growing evidence suggesting they may not be independently pro-inflammatory. OBJECTIVE To investigate the inflammatory and coagulation marker responses to an 8-week diet intervention rich in either CSO or olive oil (OO) (OO is rich in monounsaturated fat) in adults with untreated hypercholesterolemia. DESIGN This was a secondary analysis of a parallel-arm randomized clinical trial with the main outcome of cholesterol measures. PARTICIPANTS/SETTING Participants included in this analysis were 42 sedentary adults aged 30 to 75 years (62% women) in the Athens, GA, area, between May 2018 and June 2021, with untreated hypercholesterolemia or elevated blood lipids and body mass index >18.5. Hypercholesterolemia was defined as at least two blood lipid levels in a borderline undesirable/at risk range (total cholesterol level ≥180 mg/dL, low-density lipoprotein cholesterol level ≥110 mg/dL, high-density lipoprotein cholesterol level <50 mg/dL, or triglyceride level ≥130 mg/dL), or at least one in an undesirable range (total cholesterol level ≥240 mg/dL, low-density lipoprotein cholesterol level ≥160 mg/dL, high-density lipoprotein cholesterol level <40 mg/dL, or triglyceride level ≥200 mg/dL). INTERVENTION Participants were randomly assigned to either the CSO or OO group in a partial outpatient feeding trial. Meals from the study provided approximately 60% of their energy needs with 30% of energy needs from either CSO or OO for 8 weeks. Participants fulfilled their remaining energy needs with meals of their choosing. MAIN OUTCOME MEASURES Fasting plasma concentrations of inflammatory markers, including C-reactive protein, tumor necrosis factor-α, interleukin-6, and interleukin-1β were measured at baseline and 8 weeks. Markers of coagulation potential, including plasminogen activator inhibitor-1, and tissue factor were measured at the same time points. STATISTICAL ANALYSES PERFORMED Repeated measures linear mixed models were used with treatment and visit in the model for analyses of all biochemical markers. RESULTS There were no significant differences in fasting C-reactive protein (P = 0.70), tumor necrosis factor-α (P = 0.98), interleukin-6 (P = 0.21), interleukin-1β (P = 0.13), plasminogen activator inhibitor-1 (P = 0.29), or tissue factor (P = 0.29) between groups across the intervention. CONCLUSIONS Inflammation and coagulation marker responses to diets rich in CSO vs OO were not significantly different between groups, and neither group showed changes in these markers in adults with untreated hypercholesterolemia. This provides additional evidence suggesting that dietary n-6 polyunsaturated fats may not promote inflammation compared with monounsaturated fatty acids, even in adults at increased risk for cardiovascular disease.
Collapse
Affiliation(s)
- M Catherine Prater
- Department of Nutritional Sciences, University of Georgia, Athens, Georgia
| | - Alexis R Scheurell
- Department of Nutritional Sciences, University of Georgia, Athens, Georgia
| | - Chad M Paton
- Department of Nutritional Sciences, University of Georgia, Athens, Georgia; Department of Food Science and Technology, University of Georgia, Athens, Georgia
| | - Jamie A Cooper
- Department of Kinesiology, University of Georgia, Athens, Georgia.
| |
Collapse
|
3
|
Siebert AE, Brake MA, Verbeek SC, Johnston AJ, Morgan AP, Cleuren AC, Jurek AM, Schneider CD, Germain DM, Battistuzzi FU, Zhu G, Miller DR, Johnsen JM, Pardo-Manuel de Villena F, Rondina MT, Westrick RJ. Identification of genomic loci regulating platelet plasminogen activator inhibitor-1 in mice. J Thromb Haemost 2023; 21:2917-2928. [PMID: 37364776 PMCID: PMC10826891 DOI: 10.1016/j.jtha.2023.06.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 05/09/2023] [Accepted: 06/09/2023] [Indexed: 06/28/2023]
Abstract
BACKGROUND Plasminogen activator inhibitor-1 (PAI-1, Serpine1) is an important circulating fibrinolysis inhibitor. PAI-1 exists in 2 pools, packaged within platelet α-granules and freely circulating in plasma. Elevated plasma PAI-1 levels are associated with cardiovascular disease. However, little is known about the regulation of platelet PAI-1 (pPAI-1). OBJECTIVES We investigated the genetic control of pPAI-1 levels in mice and humans. METHODS We measured pPAI-1 antigen levels via enzyme-linked immunosorbent assay in platelets isolated from 10 inbred mouse strains, including LEWES/EiJ (LEWES) and C57BL/6J (B6). LEWES and B6 were crossed to produce the F1 generation, B6LEWESF1. B6LEWESF1 mice were intercrossed to produce B6LEWESF2 mice. These mice were subjected to genome-wide genetic marker genotyping followed by quantitative trait locus analysis to identify pPAI-1 regulatory loci. RESULTS We identified differences in pPAI-1 between several laboratory strains, with LEWES having pPAI-1 levels more than 10-fold higher than those in B6. Quantitative trait locus analysis of B6LEWESF2 offspring identified a major pPAI-1 regulatory locus on chromosome 5 from 136.1 to 137.6 Mb (logarithm of the odds score, 16.2). Significant pPAI-1 modifier loci on chromosomes 6 and 13 were also identified. CONCLUSION Identification of pPAI-1 genomic regulatory elements provides insights into platelet/megakaryocyte-specific and cell type-specific gene expression. This information can be used to design more precise therapeutic targets for diseases where PAI-1 plays a role.
Collapse
Affiliation(s)
- Amy E Siebert
- Department of Biological Sciences, Oakland University, Rochester, Michigan, USA
| | - Marisa A Brake
- Department of Biological Sciences, Oakland University, Rochester, Michigan, USA
| | - Stephanie C Verbeek
- Department of Biological Sciences, Oakland University, Rochester, Michigan, USA
| | | | - Andrew P Morgan
- Department of Medicine, Duke University School of Medicine, Duke University, Durham, North Carolina, USA
| | - Audrey C Cleuren
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Adrianna M Jurek
- Department of Biological Sciences, Oakland University, Rochester, Michigan, USA
| | - Caitlin D Schneider
- Department of Biological Sciences, Oakland University, Rochester, Michigan, USA
| | - Derrik M Germain
- Department of Biological Sciences, Oakland University, Rochester, Michigan, USA
| | - Fabia U Battistuzzi
- Department of Biological Sciences, Oakland University, Rochester, Michigan, USA; Department of Bioengineering, Oakland University, Rochester, Michigan, USA; Centers for Data Science and Big Data Analytics and Biomedical Research, Oakland University, Rochester, Michigan, USA
| | - Guojing Zhu
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Darla R Miller
- Department of Genetics, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jill M Johnsen
- Department of Medicine, Institute for Stem Cell & Regenerative Medicine, and Center for Cardiovascular Biology, University of Washington, Seattle, Washington, USA
| | - Fernando Pardo-Manuel de Villena
- Department of Genetics, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Matthew T Rondina
- Molecular Medicine Program, Departments of Internal Medicine and Pathology, the University of Utah, Salt Lake City, Utah, USA; The George E. Wahlen Department of Medical Affairs Medical Center, Salt Lake City, Utah, USA
| | - Randal J Westrick
- Department of Biological Sciences, Oakland University, Rochester, Michigan, USA; Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, USA; Department of Bioengineering, Oakland University, Rochester, Michigan, USA; Centers for Data Science and Big Data Analytics and Biomedical Research, Oakland University, Rochester, Michigan, USA; Eye Research Center and Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, Michigan, USA.
| |
Collapse
|
4
|
Morrow GB, Mutch NJ. Past, Present, and Future Perspectives of Plasminogen Activator Inhibitor 1 (PAI-1). Semin Thromb Hemost 2023; 49:305-313. [PMID: 36522166 DOI: 10.1055/s-0042-1758791] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Plasminogen activator inhibitor 1 (PAI-1), a SERPIN inhibitor, is primarily known for its regulation of fibrinolysis. However, it is now known that this inhibitor functions and contributes to many (patho)physiological processes including inflammation, wound healing, cell adhesion, and tumor progression.This review discusses the past, present, and future roles of PAI-1, with a particular focus on the discovery of this inhibitor in the 1970s and subsequent characterization in health and disease. Throughout the past few decades diverse functions of this serpin have unraveled and it is now considered an important player in many disease processes. PAI-1 is expressed by numerous cell types, including megakaryocytes and platelets, adipocytes, endothelial cells, hepatocytes, and smooth muscle cells. In the circulation PAI-1 exists in two pools, within plasma itself and in platelet α-granules. Platelet PAI-1 is secreted following activation with retention of the inhibitor on the activated platelet membrane. Furthermore, these anucleate cells contain PAI-1 messenger ribonucleic acid to allow de novo synthesis.Outside of the traditional role of PAI-1 in fibrinolysis, this serpin has also been identified to play important roles in metabolic syndrome, obesity, diabetes, and most recently, acute respiratory distress syndrome, including coronavirus disease 2019 disease. This review highlights the complexity of PAI-1 and the requirement to ascertain a better understanding on how this complex serpin functions in (patho)physiological processes.
Collapse
Affiliation(s)
- Gael B Morrow
- Aberdeen Cardiovascular and Diabetes Centre, Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, United Kingdom
- Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Nicola J Mutch
- Aberdeen Cardiovascular and Diabetes Centre, Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, United Kingdom
| |
Collapse
|
5
|
Humphreys SJ, Whyte CS, Mutch NJ. "Super" SERPINs-A stabilizing force against fibrinolysis in thromboinflammatory conditions. Front Cardiovasc Med 2023; 10:1146833. [PMID: 37153474 PMCID: PMC10155837 DOI: 10.3389/fcvm.2023.1146833] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 03/31/2023] [Indexed: 05/09/2023] Open
Abstract
The superfamily of serine protease inhibitors (SERPINs) are a class of inhibitors that utilise a dynamic conformational change to trap and inhibit their target enzymes. Their powerful nature lends itself well to regulation of complex physiological enzymatic cascades, such as the haemostatic, inflammatory and complement pathways. The SERPINs α2-antiplasmin, plasminogen-activator inhibitor-1, plasminogen-activator inhibitor-2, protease nexin-1, and C1-inhibitor play crucial inhibitory roles in regulation of the fibrinolytic system and inflammation. Elevated levels of these SERPINs are associated with increased risk of thrombotic complications, obesity, type 2 diabetes, and hypertension. Conversely, deficiencies of these SERPINs have been linked to hyperfibrinolysis with bleeding and angioedema. In recent years SERPINs have been implicated in the modulation of the immune response and various thromboinflammatory conditions, such as sepsis and COVID-19. Here, we highlight the current understanding of the physiological role of SERPINs in haemostasis and inflammatory disease progression, with emphasis on the fibrinolytic pathway, and how this becomes dysregulated during disease. Finally, we consider the role of these SERPINs as potential biomarkers of disease progression and therapeutic targets for thromboinflammatory diseases.
Collapse
|
6
|
Romejko K, Rymarz A, Szamotulska K, Bartoszewicz Z, Rozmyslowicz T, Niemczyk S. Left Ventricular Diastolic Dysfunction in Chronic Kidney Disease Patients Not Treated with Dialysis. Nutrients 2022; 14:4664. [PMID: 36364925 PMCID: PMC9655426 DOI: 10.3390/nu14214664] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/31/2022] [Accepted: 11/02/2022] [Indexed: 11/25/2023] Open
Abstract
BACKGROUND Left ventricular diastolic dysfunction (LVDD) is observed in the early stages of chronic kidney disease (CKD) and may lead to heart failure with preserved ejection fraction (HFpEF). The purpose of our study was to investigate the association between metabolic, nutritional and inflammatory parameters and LVDD in CKD and non-CKD patients. METHODS Two groups of patients were recruited to the study: 93 men with CKD and eGFR lower than 60 mL/min/1.73 m2 and 40 men without kidney function decrease with eGFR ≥ 60 mL/min/1.73 m2. Transthoracic echocardiography was performed to evaluate the diastolic function of the left ventricle. Bioimpedance spectroscopy (BIS) was used to measure overhydration and lean body mass. We also measured the serum concentrations of albumin, glucose, haemoglobin A1c (HgbA1c), fibrinogen, C-reactive protein (CRP), tumor necrosis factor-alpha (TNF-alpha) and osteoprotegerin (OPG). RESULTS We observed that elevated serum fibrinogen and glucose concentrations were associated with LVDD independently of CKD status. Serum fibrinogen concentrations increased with the advancement of LVDD. Low albumin concentrations in CKD were related with LVDD. In the control group, lower muscle mass presented as lean tissue index (LTI) and lean tissue mass (LTM), and overhydration were associated with LVDD. In the group of patients without kidney function decrease the OPG concentrations were significantly higher in those with LVDD, and they rose with the advancement of LVDD. CONCLUSIONS Elevated inflammatory parameters, increased serum glucose concentrations and worse nutritional status are the states that may impair the diastolic function of the left ventricle in CKD and non-CKD patients. Serum OPG levels are elevated in patients without kidney function decrease and LVDD and its concentrations rise with the advancement of LVDD.
Collapse
Affiliation(s)
- Katarzyna Romejko
- Department of Internal Diseases, Nephrology and Dialysis, Military Institute of Medicine, 128 Szaserów Street, 04-141 Warsaw, Poland
| | - Aleksandra Rymarz
- Department of Internal Diseases, Nephrology and Dialysis, Military Institute of Medicine, 128 Szaserów Street, 04-141 Warsaw, Poland
| | - Katarzyna Szamotulska
- Department of Epidemiology and Biostatistics, Institute of Mother and Child, Kasprzaka Street, 17a, 01-211 Warsaw, Poland
| | - Zbigniew Bartoszewicz
- Department of Internal Diseases and Endocrinology, Medical University of Warsaw, 1a Banacha Street, 02-097 Warsaw, Poland
| | - Tomasz Rozmyslowicz
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, R.217 John Morgan Building, 3620 Hamilton Walk, Philadelphia, PA 19104, USA
| | - Stanisław Niemczyk
- Department of Internal Diseases, Nephrology and Dialysis, Military Institute of Medicine, 128 Szaserów Street, 04-141 Warsaw, Poland
| |
Collapse
|
7
|
Mossberg K, Olausson J, Fryk E, Jern S, Jansson PA, Brogren H. The role of the platelet pool of Plasminogen Activator Inhibitor-1 in well-controlled type 2 diabetes patients. PLoS One 2022; 17:e0267833. [PMID: 36044519 PMCID: PMC9432754 DOI: 10.1371/journal.pone.0267833] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 04/17/2022] [Indexed: 11/19/2022] Open
Abstract
Background The main inhibitor of the fibrinolytic system, Plasminogen Activator Inhibitor -1 (PAI-1), irreversibly binds tissue-type Plasminogen Activator (t-PA) and thereby inhibits the protective action of tPA against thrombus formation. Elevated levels of plasma PAI-1 are associated with an increased risk of cardiovascular events and are observed in subjects with type 2 diabetes (T2D) and obesity. Platelets contain the majority of PAI-1 present in blood and exhibit the ability to synthesis active PAI-1. Diabetic platelets are known to be hyper-reactive and larger in size; however, whether these features affect their contribution to the elevated levels of plasma PAI-1 in T2D is not established. Objectives To characterize the PAI-1 antigen content and the mRNA expression in platelets from T2D subjects compared to obese and lean control subjects, in order to elucidate the role of platelet PAI-1 in T2D. Methods Nine subjects with T2D and obesity were recruited from Primary Care Centers together with 15 healthy control subjects (8 lean subjects and 7 with obesity). PAI-1 antigen levels in plasma, serum and platelets were determined by ELISA, and PAI-1 mRNA expression was analyzed by qPCR. Results There was no significant difference in PAI-1 mRNA expression or PAI-1 antigen in platelets in T2D subject in comparison to obese and lean control subjects. An elevated level of plasma PAI-1 was seen in both T2D and obese subjects. PAI-1 gene expression was significantly higher in both obese groups compared to lean. Conclusion Similar levels of protein and mRNA expression of PAI-1 in platelets from T2D, obese and lean subjects indicate a limited role of platelets for the elevated plasma PAI-1 levels. However, an increased synthesis rate of mRNA transcripts in platelets from T2D and an increased release of PAI-1 could also result in similar mRNA and protein levels. Hence, synthesis and release rates of PAI-1 from platelets in T2D and obesity need to be investigated to further elucidate the role of platelets in obesity and T2D.
Collapse
Affiliation(s)
- Karin Mossberg
- Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, Göteborg, Sweden
- Department of Public Health and Community Medicine, Sahlgrenska University Hospital, Göteborg, Sweden
| | - Josefin Olausson
- Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, Göteborg, Sweden
- The Wallenberg Laboratory for Cardiovascular Research, Göteborg, Sweden
| | - Emanuel Fryk
- Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, Göteborg, Sweden
- The Wallenberg Laboratory for Cardiovascular Research, Göteborg, Sweden
| | - Sverker Jern
- Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, Göteborg, Sweden
- The Wallenberg Laboratory for Cardiovascular Research, Göteborg, Sweden
| | - Per-Anders Jansson
- Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, Göteborg, Sweden
- The Wallenberg Laboratory for Cardiovascular Research, Göteborg, Sweden
| | - Helén Brogren
- Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
- * E-mail:
| |
Collapse
|
8
|
Kanji R, Gue YX, Farag MF, Spencer NH, Mutch NJ, Gorog DA. Determinants of Endogenous Fibrinolysis in Whole Blood Under High Shear in Patients With Myocardial Infarction. JACC Basic Transl Sci 2022; 7:1069-1082. [PMID: 36687271 PMCID: PMC9849272 DOI: 10.1016/j.jacbts.2022.05.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/24/2022] [Accepted: 05/24/2022] [Indexed: 01/25/2023]
Abstract
Hypofibrinolysis is a recently-recognized risk factor for recurrent cardiovascular events in patients with ST-segment elevation myocardial infarction (STEMI), but the mechanistic determinants of this are not well understood. In patients with STEMI, we show that the effectiveness of endogenous fibrinolysis in whole blood is determined in part by fibrinogen level, high sensitivity C-reactive protein, and shear-induced platelet reactivity, the latter directly related to the speed of thrombin generation. Our findings strengthen the evidence for the role of cellular components and bidirectional crosstalk between coagulatory and inflammatory pathways as determinants of hypofibrinolysis.
Collapse
Affiliation(s)
- Rahim Kanji
- Faculty of Medicine, National Heart and Lung Institute, Imperial College, London, United Kingdom,Cardiology Department, East and North Hertfordshire NHS Trust, Stevenage, Hertfordshire, United Kingdom
| | - Ying X. Gue
- Cardiology Department, East and North Hertfordshire NHS Trust, Stevenage, Hertfordshire, United Kingdom,School of Life and Medical Sciences, Postgraduate Medical School, University of Hertfordshire, Hatfield, Hertfordshire, United Kingdom
| | - Mohamed F. Farag
- Cardiology Department, East and North Hertfordshire NHS Trust, Stevenage, Hertfordshire, United Kingdom,School of Life and Medical Sciences, Postgraduate Medical School, University of Hertfordshire, Hatfield, Hertfordshire, United Kingdom
| | - Neil H. Spencer
- Statistical Services and Consultancy Unit, Hertfordshire Business School, University of Hertfordshire, Hatfield, Hertfordshire, United Kingdom
| | - Nicola J. Mutch
- Aberdeen Cardiovascular and Diabetes Centre, Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, United Kingdom
| | - Diana A. Gorog
- Faculty of Medicine, National Heart and Lung Institute, Imperial College, London, United Kingdom,Cardiology Department, East and North Hertfordshire NHS Trust, Stevenage, Hertfordshire, United Kingdom,School of Life and Medical Sciences, Postgraduate Medical School, University of Hertfordshire, Hatfield, Hertfordshire, United Kingdom,Address for correspondence: Prof Diana A. Gorog, Faculty of Medicine, National Heart and Lung Institute, Imperial College, Dovehouse Street, London SW3 6LY, United Kingdom.
| |
Collapse
|
9
|
Loureirin B Alleviates Myocardial Ischemia/Reperfusion Injury via Inhibiting PAI-1/TGF- β1/Smad Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:9128210. [PMID: 35535157 PMCID: PMC9078770 DOI: 10.1155/2022/9128210] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 03/28/2022] [Accepted: 04/04/2022] [Indexed: 12/02/2022]
Abstract
Myocardial ischemia/reperfusion (MI/R) injury is a common clinical problem after myocardial infarction without effective therapy. Loureirin B (LrB) is a kind of flavonoid with anti-inflammatory and antifibrotic activities. However, the effect of LrB on MI/R and its underlying mechanism remains elusive. In the present study, a mouse model of MI/R was established by coronary artery occlusion. Administration of LrB (0.5 mg/kg or 1 mg/kg) for 4 weeks effectively improved left ventricular (LV) function and reduced myocardial infarction in MI/R mice. MI/R-induced expression of IL-6, TNF-α, and IL-1β in the hearts was reduced by LrB treatment. Histological analysis showed that LrB attenuated myocardial collagen deposition. LrB downregulated fibronectin, collagen I, collagen III, and α-SMA expression. Notably, LrB inhibited the expression of profibrotic plasminogen activator inhibitor-1 (PAI-1), transforming growth factor (TGF)-β1, TGF-β1R, and p-Smad2/3. Consistently, LrB inhibited the activation of TGF-β1/Smad signaling pathway and the expression of fibrosis-related proteins in angiotensin (Ang) II-treated cardiac fibroblasts (CFs). Overexpression of PAI-1 abolished the effects of LrB on Ang II-treated CFs, suggesting that LrB may function through regulating PAI-1. These results indicated that LrB may alleviate MI/R-induced myocardial fibrosis by inhibiting PAI-1/TGF-β1/Smad signaling pathway. Thus, LrB may be a potential drug in the treatment of MI/R injury.
Collapse
|
10
|
The Morphology of Coronary Artery Disease in South Asians versus White Caucasians and its Implications. Can J Cardiol 2022; 38:1570-1579. [PMID: 35568268 DOI: 10.1016/j.cjca.2022.05.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 04/27/2022] [Accepted: 05/05/2022] [Indexed: 01/09/2023] Open
Abstract
South Asians (SAs) experience a higher prevalence and earlier onset of coronary artery disease and have worse outcomes relative to White Caucasians (WCs) following invasive revascularization procedures, a mainstay of coronary artery disease (CAD) management. We sought to review the differences in the CAD pattern and risk factors between SA and WC patients and discuss their potential impact on the development of coronary disease, acute coronary syndrome and revascularization outcomes. SAs have a more diffuse pattern with multi-vessel involvement compared to WCs. However, less is known about other morphological characteristics such as calcification of atherosclerotic plaque and coronary diameter in SA populations. Despite a similar coronary calcification burden, higher non-calcified plaque composition, elevated thrombosis and inflammatory markers likely contribute to the disease pattern. While the current evidence on the role of coronary vessel size remains inconsistent, smaller coronary diameters in SAs could play a potential role in the higher disease prevalence. This is especially important given the impact of coronary artery diameter on revascularization outcomes. In conclusion, SAs have a unique CAD risk profile comprised of traditional and novel risk factors. Our findings highlight the need for additional awareness of healthcare professionals of this specific risk profile and potential therapeutic targets, as well as the need for further research in this vulnerable population.
Collapse
|
11
|
Basak S, Banerjee A, Pathak S, Duttaroy AK. Dietary Fats and the Gut Microbiota: Their impacts on lipid-induced metabolic syndrome. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
12
|
Eriksen PL, Thomsen KL, Sørensen M, Vilstrup H, Hvas AM. Impaired fibrinolysis without hypercoagulability characterises patients with non-alcoholic fatty liver disease. Thromb Res 2022; 213:9-15. [DOI: 10.1016/j.thromres.2022.02.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 02/14/2022] [Accepted: 02/24/2022] [Indexed: 12/12/2022]
|
13
|
Ye RZ, Richard G, Gévry N, Tchernof A, Carpentier AC. Fat Cell Size: Measurement Methods, Pathophysiological Origins, and Relationships With Metabolic Dysregulations. Endocr Rev 2022; 43:35-60. [PMID: 34100954 PMCID: PMC8755996 DOI: 10.1210/endrev/bnab018] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Indexed: 11/19/2022]
Abstract
The obesity pandemic increasingly causes morbidity and mortality from type 2 diabetes, cardiovascular diseases and many other chronic diseases. Fat cell size (FCS) predicts numerous obesity-related complications such as lipid dysmetabolism, ectopic fat accumulation, insulin resistance, and cardiovascular disorders. Nevertheless, the scarcity of systematic literature reviews on this subject is compounded by the use of different methods by which FCS measurements are determined and reported. In this paper, we provide a systematic review of the current literature on the relationship between adipocyte hypertrophy and obesity-related glucose and lipid dysmetabolism, ectopic fat accumulation, and cardiovascular disorders. We also review the numerous mechanistic origins of adipocyte hypertrophy and its relationship with metabolic dysregulation, including changes in adipogenesis, cell senescence, collagen deposition, systemic inflammation, adipokine secretion, and energy balance. To quantify the effect of different FCS measurement methods, we performed statistical analyses across published data while controlling for body mass index, age, and sex.
Collapse
Affiliation(s)
- Run Zhou Ye
- Division of Endocrinology, Department of Medicine, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Gabriel Richard
- Division of Endocrinology, Department of Medicine, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Nicolas Gévry
- Department of Biology, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - André Tchernof
- Québec Heart and Lung Research Institute, Laval University, Québec, Québec, Canada
| | - André C Carpentier
- Division of Endocrinology, Department of Medicine, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec, Canada
| |
Collapse
|
14
|
Taheri A, Mirzababaei A, Setayesh L, Yarizadeh H, Shiraseb F, Imani H, C T Clark C, Mirzaei K. The relationship between Dietary approaches to stop hypertension diet adherence and inflammatory factors and insulin resistance in overweight and obese women: A cross-sectional study. Diabetes Res Clin Pract 2021; 182:109128. [PMID: 34785301 DOI: 10.1016/j.diabres.2021.109128] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 10/29/2021] [Accepted: 11/02/2021] [Indexed: 11/21/2022]
Abstract
BACKGROUND Although there is abundant evidence for an association between dietary pattern, weight, and other related factors, such as homeostatic model assessment for insulin resistance (HOMA-IR) and inflammatory markers; there is limited information pertaining to levels of plasminogen activator inhibitor-1 (PAI-1) and Monocyte Chemoattractant Protein -1 (MCP-1). Therefore, this study sought to examine the association between adherence to the dietary approaches to stop hypertension (DASH) diet and levels of inflammatory factors PAI-1, MCP-1, and HOMA-IR. METHODS This cross-sectional study was performed on 305 obese and overweight women. The typical food intake of individuals was assessed using the 147 items semi-quantitative food frequency questionnaire (FFQ). Body components were measured for all participants. Insulin resistance was estimated by homeostasis model assessment (HOMA), and biochemical parameters were examined. RESULTS No significant relationship was observed between the DASH diet and MCP-1 (P-trend = 0.70), PAI-1 (P-trend = 0.92), or HOMA-IR (P-trend = 0.08) in the crude model. However, there was a significant inverse relationship between the DASH diet and HOMA-IR (P-trend = 0.03) after adjusting for age, BMI, and physical activity. CONCLUSION This study showed that higher adherence to DASH pattern is inversely correlated with HOMA-IR in overweight and obese women.
Collapse
Affiliation(s)
- Akram Taheri
- Department of Nutrition, Faculty of Medicine, Sciences and Research Branch, Islamic Azad, University, Tehran, Iran
| | - Atieh Mirzababaei
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Leila Setayesh
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Habib Yarizadeh
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Farideh Shiraseb
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Hossein Imani
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Iran
| | - Cain C T Clark
- Centre for Intelligent Healthcare, Coventry University, Coventry CV1 5FB, UK
| | - Khadijeh Mirzaei
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran.
| |
Collapse
|
15
|
Wong YK, Tse HF. Circulating Biomarkers for Cardiovascular Disease Risk Prediction in Patients With Cardiovascular Disease. Front Cardiovasc Med 2021; 8:713191. [PMID: 34660715 PMCID: PMC8517145 DOI: 10.3389/fcvm.2021.713191] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 09/08/2021] [Indexed: 12/23/2022] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death globally. Risk assessment is crucial for identifying at-risk individuals who require immediate attention as well as to guide the intensity of medical therapy to reduce subsequent risk of CVD. In the past decade, many risk prediction models have been proposed to estimate the risk of developing CVD. However, in patients with a history of CVD, the current models that based on traditional risk factors provide limited power in predicting recurrent cardiovascular events. Several biomarkers from different pathophysiological pathways have been identified to predict cardiovascular events, and the incorporation of biomarkers into risk assessment may contribute to enhance risk stratification in secondary prevention. This review focuses on biomarkers related to cardiovascular and metabolic diseases, including B-type natriuretic peptide, high-sensitivity cardiac troponin I, adiponectin, adipocyte fatty acid-binding protein, heart-type fatty acid-binding protein, lipocalin-2, fibroblast growth factor 19 and 21, retinol-binding protein 4, plasminogen activator inhibitor-1, 25-hydroxyvitamin D, and proprotein convertase subtilisin/kexin type 9, and discusses the potential utility of these biomarkers in cardiovascular risk prediction among patients with CVD. Many of these biomarkers have shown promise in improving risk prediction of CVD. Further research is needed to assess the validity of biomarker and whether the strategy for incorporating biomarker into clinical practice may help to optimize decision-making and therapeutic management.
Collapse
Affiliation(s)
- Yuen-Kwun Wong
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, China
| | - Hung-Fat Tse
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, China.,Department of Medicine, Shenzhen Hong Kong University Hospital, Shenzhen, China.,Hong Kong-Guangdong Joint Laboratory on Stem Cell and Regenerative Medicine, The University of Hong Kong, Hong Kong, China.,Shenzhen Institutes of Research and Innovation, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
16
|
Chen J, Zhai C, Wang Z, Li R, Wu W, Hou K, Alzogool M, Wang Y, Cong H. The susceptibility of SERPINE1 rs1799889 SNP in diabetic vascular complications: a meta-analysis of fifty-one case-control studies. BMC Endocr Disord 2021; 21:195. [PMID: 34592988 PMCID: PMC8482645 DOI: 10.1186/s12902-021-00837-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 08/10/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND The serine protease inhibitor-1 (SERPINE1) rs1799889 single nucleotide polymorphism (SNP) has been constantly associated with diabetes mellitus (DM) and its vascular complications. The aim of this meta-analysis was to evaluate this association with combined evidences. METHODS The systematic search was performed for studies published up to March 2021 which assess the associations between SERPINE1 rs1799889 SNP and the risks of DM, diabetic retinopathy (DR), diabetic cardiovascular disease (CVD) and diabetic nephropathy (DN). Only case-control studies were identified, and the linkage between SERPINE1 rs1799889 polymorphism and diabetic vascular risks were evaluated using genetic models. RESULTS 51 comparisons were enrolled. The results revealed a significant association with diabetes risk in overall population (allelic: OR = 1.34, 95 % CI = 1.14-1.57, homozygous: OR = 1.66, 95 % CI = 1.23-2.14, heterozygous: OR = 1.35, 95 % CI = 1.08-1.69, dominant: OR = 1.49, 95 % CI = 1.18-1.88, recessive: OR = 1.30, 95 % CI = 1.06-1.59) as well as in Asian descents (allelic: OR = 1.45, 95 % CI = 1.16-1.82, homozygous: OR = 1.88, 95 % CI = 1.29-2.75, heterozygous: OR = 1.47, 95 % CI = 1.08-2.00, dominant: OR = 1.64, 95 % CI = 1.21-2.24, recessive: OR = 1.46, 95 % CI = 1.09-1.96). A significant association was observed with DR risk (homozygous: OR = 1.25, 95 % CI = 1.01-1.56, recessive: OR = 1.20, 95 % CI = 1.01-1.43) for overall population, as for the European subgroup (homozygous: OR = 1.32, 95 % CI = 1.02-1.72, recessive: OR = 1.38, 95 % CI = 1.11-1.71). A significant association were shown with DN risk for overall population (allelic: OR = 1.48, 95 % CI = 1.15-1.90, homozygous: OR = 1.92, 95 % CI = 1.26-2.95, dominant: OR = 1.41, 95 % CI = 1.01-1.97, recessive: OR = 1.78, 95 % CI = 1.27-2.51) and for Asian subgroup (allelic: OR = 1.70, 95 % CI = 1.17-2.47, homozygous: OR = 2.46, 95 % CI = 1.30-4.66, recessive: OR = 2.24, 95 % CI = 1.40-3.59) after ethnicity stratification. No obvious association was implied with overall diabetic CVD risk in any genetic models, or after ethnicity stratification. CONCLUSIONS SERPINE1 rs1799889 4G polymorphism may outstand for serving as a genetic synergistic factor in overall DM and DN populations, positively for individuals with Asian descent. The association of SERPINE1 rs1799889 SNP and DR or diabetic CVD risks was not revealed.
Collapse
Affiliation(s)
- JingYi Chen
- School of Medicine, NanKai University, Weijin Road No. 94, Nankai District, 300071 Tianjin, China
- Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Gansu Road No. 4, Heping District, 300020 Tianjin, China
| | - ChuanNan Zhai
- Department of Cardiology, Tianjin Chest Hospital, Taierzhuang south Road No. 291, Jinnan District, 300350 Tianjin, China
| | - ZhiQian Wang
- Department of Optometry, Shenyang Eye Institute, The 4th People’s Hospital of Shenyang, No 20. Huanghe South Avenue, Huanggu District, 110031 Shenyang, Liaoning China
| | - Rui Li
- Tianjin GongAn Hospital, Nanjing Road No. 78, Heping District, 300042 Tianjin, China
| | - WenJing Wu
- Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Gansu Road No. 4, Heping District, 300020 Tianjin, China
| | - Kai Hou
- Department of Cardiology, Tianjin Chest Hospital, Taierzhuang south Road No. 291, Jinnan District, 300350 Tianjin, China
| | - Mohammad Alzogool
- School of Medicine, NanKai University, Weijin Road No. 94, Nankai District, 300071 Tianjin, China
- Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Gansu Road No. 4, Heping District, 300020 Tianjin, China
| | - Yan Wang
- School of Medicine, NanKai University, Weijin Road No. 94, Nankai District, 300071 Tianjin, China
- Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Gansu Road No. 4, Heping District, 300020 Tianjin, China
| | - HongLiang Cong
- Department of Cardiology, Tianjin Chest Hospital, Taierzhuang south Road No. 291, Jinnan District, 300350 Tianjin, China
| |
Collapse
|
17
|
Siniarski A, Baker SR, Duval C, Malinowski KP, Gajos G, Nessler J, Ariëns RAS. Quantitative analysis of clot density, fibrin fiber radius, and protofibril packing in acute phase myocardial infarction. Thromb Res 2021; 205:110-119. [PMID: 34298252 DOI: 10.1016/j.thromres.2021.06.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 05/04/2021] [Accepted: 06/30/2021] [Indexed: 11/18/2022]
Abstract
INTRODUCTION Coronary artery disease is associated with impaired clot structure. The aim of this study was to investigate acute phase myocardial infarction (AMI) and provide detailed quantitative analysis of clot ultrastructure. MATERIALS AND METHODS Clot formation and breakdown, pore size, fiber density, fiber radius and protofibril packing were investigated in plasma clots from AMI patients. These data were compared to those from healthy controls. RESULTS Analysis on clot formation using turbidity showed increased lag time, suggesting changes in protofibril packing and increased fiber size for AMI patients compared to healthy controls. Additionally, increased average rate of clotting and decreased time to maximum absorbance in AMI patients suggest that clots formed more quickly. Moreover, we observed increased time from max OD to max rate of lysis. Increased fibrinogen and decreased plasminogen in AMI patients were accounted for in represented significant differences. AMI samples showed increased time to 25% and 50% lysis, but no change in 75% lysis, representative of delayed lysis onset, but expediated lysis once initiated. These data suggest that AMI patients formed less porous clots made from more densely packed fibers with decreased numbers of protofibrils, which was confirmed using decreased permeation and increased fiber density, and decreased turbidimetry. CONCLUSIONS AMI plasma formed clots that were denser, less permeable, and lysed more slowly than healthy controls. These findings were confirmed by detailed analysis of clot ultrastructure, fiber size, and protofibril packing. Dense clot structures that are resistant to lysis may contribute to a prothrombotic milieu in AMI.
Collapse
Affiliation(s)
- Aleksander Siniarski
- Department of Coronary Disease and Heart Failure, Institute of Cardiology, Jagiellonian University Medical College, Krakow, Poland; John Paul II Hospital, Krakow, Poland
| | - Stephen R Baker
- Leeds Thrombosis Collective, Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK; Department of Physics, Wake Forest University, Winston Salem, NC, USA.
| | - Cédric Duval
- Leeds Thrombosis Collective, Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | | | - Grzegorz Gajos
- Department of Coronary Disease and Heart Failure, Institute of Cardiology, Jagiellonian University Medical College, Krakow, Poland; John Paul II Hospital, Krakow, Poland
| | - Jadwiga Nessler
- Department of Coronary Disease and Heart Failure, Institute of Cardiology, Jagiellonian University Medical College, Krakow, Poland; John Paul II Hospital, Krakow, Poland
| | - Robert A S Ariëns
- Leeds Thrombosis Collective, Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK.
| |
Collapse
|
18
|
Morrow GB, Whyte CS, Mutch NJ. A Serpin With a Finger in Many PAIs: PAI-1's Central Function in Thromboinflammation and Cardiovascular Disease. Front Cardiovasc Med 2021; 8:653655. [PMID: 33937363 PMCID: PMC8085275 DOI: 10.3389/fcvm.2021.653655] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 02/23/2021] [Indexed: 12/27/2022] Open
Abstract
Plasminogen activator inhibitor 1 (PAI-1) is a member of the serine protease inhibitor (serpin) superfamily. PAI-1 is the principal inhibitor of the plasminogen activators, tissue plasminogen activator (tPA), and urokinase-type plasminogen activator (uPA). Turbulence in the levels of PAI-1 tilts the balance of the hemostatic system resulting in bleeding or thrombotic complications. Not surprisingly, there is strong evidence that documents the role of PAI-1 in cardiovascular disease. The more recent uncovering of the coalition between the hemostatic and inflammatory pathways has exposed a distinct role for PAI-1. The storm of proinflammatory cytokines liberated during inflammation, including IL-6 and TNF-α, directly influence PAI-1 synthesis and increase circulating levels of this serpin. Consequently, elevated levels of PAI-1 are commonplace during infection and are frequently associated with a hypofibrinolytic state and thrombotic complications. Elevated PAI-1 levels are also a feature of metabolic syndrome, which is defined by a cluster of abnormalities including obesity, type 2 diabetes, hypertension, and elevated triglyceride. Metabolic syndrome is in itself defined as a proinflammatory state associated with elevated levels of cytokines. In addition, insulin has a direct impact on PAI-1 synthesis bridging these pathways. This review describes the key physiological functions of PAI-1 and how these become perturbed during disease processes. We focus on the direct relationship between PAI-1 and inflammation and the repercussion in terms of an ensuing hypofibrinolytic state and thromboembolic complications. Collectively, these observations strengthen the utility of PAI-1 as a viable drug target for the treatment of various diseases.
Collapse
Affiliation(s)
- Gael B Morrow
- Aberdeen Cardiovascular and Diabetes Centre, Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, United Kingdom.,Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Claire S Whyte
- Aberdeen Cardiovascular and Diabetes Centre, Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, United Kingdom
| | - Nicola J Mutch
- Aberdeen Cardiovascular and Diabetes Centre, Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, United Kingdom
| |
Collapse
|
19
|
Vahidi M, Asgari S, Tohidi M, Azizi F, Hadaegh F. Macrosomia is a risk factor for incident maternal chronic kidney disease. BMC Pregnancy Childbirth 2021; 21:210. [PMID: 33726706 PMCID: PMC7968264 DOI: 10.1186/s12884-021-03695-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 03/03/2021] [Indexed: 11/29/2022] Open
Abstract
Background Gestational diabetes mellitus (GDM) and macrosomia are associated with several adverse outcomes including diabetes mellitus and cardiovascular diseases, however, the relationship between GDM/macrosomia with incident chronic kidney disease (CKD) is a matter of debate. The purpose of this study was to examine the association between the history of macrosomia with or without GDM and incident maternal CKD. Methods The study population includes 2669 women aged 18–50 years without known diabetes mellitus and CKD from participants of the Tehran Lipid and Glucose Study. The study population was categorized into 3 groups; group 1: GDM/macrosomia and without diabetes mellitus (n = 204), group 2: newly diagnosed incident diabetes mellitus (NDM) in the presence or abcence of GDM/Macrosomia (n = 113), and, group 3: the reference group including women without prior history of GDM/macrosomia and free of NDM (n = 2352). CKD was defined as an estimated glomerular filtration rate (eGFR) < 60 ml/min/1.73 m2. Multivariable Cox proportional hazard regression adjusted for baseline values of age, body mass index, waist circumference, parity numbers, smoking, educational level, gestational hypertension, eGFR, systolic and diastolic blood pressures (SBP and DBP, respectively), anti-hypertensive medication, and family history of diabetes mellitus was applied for data analyses. Results During a median follow-up of 11.9 years, 613 incident CKD cases were identified. The multivariable hazard ratio (HR) and 95% confidence interval (CI) on GDM/macrosomia group was [1.32 (1.02–1.72)]; the risk was more prominent among non-hypertensive women [1.41 (1.07–1.85); P for interaction: 0.046]. Moreover, the history of macrosomia alone also showed a significant risk [1.36 (1.04–1.78)]; however, history of GDM alone did not have a significant risk [0.92 (0.34–2.46)]. Age, current smoking, eGFR, and SBP remained as independent risk factors for incident CKD. Conclusions A history of GDM/macrosomia or macrosomia alone, independent of subsequent diabetes mellitus was associated with significant risk for incident maternal CKD. Pregnancy may provide a unique situation to identify high-risk women at risk for CKD that could benefit from regular monitoring of kidney function and providing risk modifying strategies.
Collapse
Affiliation(s)
- Mohammad Vahidi
- Prevention of Metabolic Disorders Research Center, Research Institute for Endocrine Sciences Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Samaneh Asgari
- Prevention of Metabolic Disorders Research Center, Research Institute for Endocrine Sciences Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Tohidi
- Prevention of Metabolic Disorders Research Center, Research Institute for Endocrine Sciences Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Fereidoun Azizi
- Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farzad Hadaegh
- Prevention of Metabolic Disorders Research Center, Research Institute for Endocrine Sciences Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
20
|
Koca N, Ayar K, Bal Ö, Ersoy C. The evaluation of the role of BMI and insulin resistance on inflammatory markers, PAI-1 levels and arterial stiffness in newly diagnosed type 2 diabetes mellitus patients. Minerva Endocrinol (Torino) 2021; 46:116-123. [PMID: 33779113 DOI: 10.23736/s2724-6507.20.03158-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Increased cardiovascular risk, represented by endothelial inflammation, probably starts with the very first course of type-2 diabetes (T2DM). Almost 85.2% of all T2DM patients are overweight or obese. Thrombosis accounts 80% of all deaths in patients with diabetes. The thrombotic-fibrinolytic equilibrium shifts in favor of thrombosis by plasminogen activator inhibitor-1 (PAI-1). PAI-1 secretion is induced primarily by CRP. PAI-1 overexpression predisposes unstable plaque development. The contribution of obesity and diabetes to this process is not clearly understood. In this study, we aimed to investigate comparison of inflammatory markers, PAI-1 levels and arterial stiffness according to BMI and impaired glucose metabolism in patient with newly diagnosed T2DM. METHODS Newly diagnosed 60 T2DM patients were enrolled. Demographics and measurements were noted. Liver (AST, ALT), kidney (urea, creatinine, albumin/creatinine ratio), metabolic (fasting blood glucose, post-prandial blood glucose, insulin, c-peptide, HbA1c, total cholesterol, low-density lipoprotein [LDL], high-density lipoprotein [HDL], triglyceride) parameters, inflammatory markers [hsCRP, fibrinogen]), PAI-1 levels and pulse wave velocity was measured from all participants. The results were compared. RESULTS Inflammatory markers and PAI-1 levels were significantly elevated in obese group compared to overweight participants. The correlation analysis showed that waist and hip circumferences, high-sensitive CRP, fibrinogen and PAI-1 levels were positively correlated with BMI but not with HbA1c levels. CONCLUSIONS The results of our study showed that lipid levels, glycemic and blood pressure values of the obese and overweight patients were similar. BMI affects inflammatory markers and PAI-1 levels independent of glucose regulation and insulin resistance in newly diagnosed T2DM. According to the current study BMI is found to be more prominent in terms of inflammatory markers and PAI-1 levels compared to insulin resistance and impaired glucose metabolism in newly diagnosed T2DM.
Collapse
Affiliation(s)
- Nizameddin Koca
- Department of Internal Medicine, University of Health Sciences, Bursa Yuksek Ihtisas Training and Research Hospital, Bursa, Turkey -
| | - Koray Ayar
- Department of Rheumatology, University of Health Sciences, Bursa Yuksek Ihtisas Training and Research Hospital, Bursa, Turkey
| | - Öznur Bal
- Department of Medical Oncology, Ankara Numune Training and Research Hospital, Ankara, Turkey
| | - Canan Ersoy
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Uludag University Hospital, Bursa, Turkey
| |
Collapse
|
21
|
Xie W, You J, Zhi C, Li L. The toxicity of ambient fine particulate matter (PM2.5) to vascular endothelial cells. J Appl Toxicol 2021; 41:713-723. [DOI: 10.1002/jat.4138] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 12/17/2020] [Accepted: 12/27/2020] [Indexed: 12/20/2022]
Affiliation(s)
- Wei Xie
- Clinical Anatomy & Reproductive Medicine Application Institute University of South China Hengyang China
| | - Jia You
- Clinical Anatomy & Reproductive Medicine Application Institute University of South China Hengyang China
| | - Chenxi Zhi
- Clinical Anatomy & Reproductive Medicine Application Institute University of South China Hengyang China
| | - Liang Li
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards University of South China Hengyang China
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Medical Research Center, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study University of South China Hengyang China
| |
Collapse
|
22
|
Wang W, Jia L. Regulatory Mechanism of MicroRNA-30b on Neonatal Hypoxic-Ischemic Encephalopathy (HIE). J Stroke Cerebrovasc Dis 2020; 30:105553. [PMID: 33360521 DOI: 10.1016/j.jstrokecerebrovasdis.2020.105553] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 12/02/2020] [Accepted: 12/12/2020] [Indexed: 01/24/2023] Open
Abstract
OBJECTIVE This study is to investigate the role of microRNA (miR)-30b in the pathogenesis of hypoxic-ischemic encephalopathy (HIE) in neonates. METHODS Totally 26 cases of neonatal HIE were included in this study. The protein expression levels of CD26P and PAI-1 were detected with ELISA. Serum levels of miR-30b and PAI-1 mRNA was measured by quantitative real-time PCR. Human brain microvascular endothelial cells (HBMECs) were cultured under hypoxic condition, and the intracellular expression levels of miR-30b and PAI-1 were evaluated. Dual-luciferase reporter assay was performed to confirm the interaction between miR-30b and PAI-1. RESULTS Compared with the control group, both the mRNA and protein expression levels of PAI-1 in the serum were up-regulated in the neonates with HIE, together with up-regulated serum CD26P levels. However, the serum expression level of miR-30b was down-regulated in neonatal HIE. In hypoxia-induced HBMECs, the mRNA and protein expression levels of PAI-1 were significantly up-regulated, while the miR-30b expression level was significantly down-regulated. Dual-luciferase reporter assay showed that PAI-1 was the direct target of miR-30b. CONCLUSION Neonatal HIE is accompanied with abnormal platelet activation, significantly up-regulated serum PAI-1 expression levels, and down-regulated miR-30b expression. MiR-30b might regulate the disease pathogenesis and immune responses via modulating PAI-1.
Collapse
Affiliation(s)
- Weiwei Wang
- Department of Pediatrics, Tianjin hospital, Tianjin 300211, China.
| | - Lifang Jia
- Department of Pediatrics, Tianjin hospital, Tianjin hospital of Tianjin University, Tianjin 300211, China.
| |
Collapse
|
23
|
Translational insight into prothrombotic state and hypercoagulation in nonalcoholic fatty liver disease. Thromb Res 2020; 198:139-150. [PMID: 33340925 DOI: 10.1016/j.thromres.2020.12.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 11/17/2020] [Accepted: 12/07/2020] [Indexed: 02/08/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is an emerging and threatening pathological condition, ranging from fatty liver (FL) to chronic steatohepatitis (NASH), liver cirrhosis, and eventually to hepatocellular carcinoma (HCC). Recent findings suggest that patients with NAFLD have a higher risk of cardiovascular events and thromboembolism and that this risk is independent of metabolic diseases that are frequently associated with NAFLD, such as diabetes, hyperlipidaemia, and obesity. The vascular involvement of NAFLD might be considered its systemic burden, conditioning higher mortality in patients affected by the disease. These clinical findings suggested the existence of a prothrombotic state in NAFLD, which is partially unexplored and whose underlying mechanisms are to date not completely understood. Here, we review the mechanisms involved in the pathogenesis of the prothrombotic state in NAFLD across the progression from the healthy liver through the different stages of the disease. We focused on the possible role of several metabolic features of NAFLD possibly leading to hypercoagulation other than endothelial and platelet activation, such as insulin-resistance, nitric oxide production regulation, and gut microbiota homeostasis. Also, we analysed the involvement of plasminogen activator inhibitor-1 (PAI-1) and thromboinflammation taking place in NAFLD. Finally, we described factors striking a prothrombotic imbalance in NASH cirrhosis, with a particular focus on the pathogenesis of portal vein thrombosis.
Collapse
|
24
|
Liu K, Xu J, Tao L, Yang K, Sun Y, Guo X. Platelet counts are associated with arterial stiffness in Chinese Han population: a longitudinal study. BMC Cardiovasc Disord 2020; 20:353. [PMID: 32731902 PMCID: PMC7393731 DOI: 10.1186/s12872-020-01634-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 07/20/2020] [Indexed: 11/13/2022] Open
Abstract
Background Determining the risk factors for brachial-ankle pulse wave velocity (baPWV) may help to identify people susceptible to diabetic atherosclerosis and could prevent diabetic macrovascular complications in the early stages. We aim to comprehensively investigate risk factors contributing to arterial stiffness in patients with and without diabetes. Methods BaPWV was measured in 5651 individuals who attended health check-ups at baseline and follow-up. Lasso regression was used to screen for risk factors. Mixed models and multiple linear regressions were subsequently established to evaluate the effect size of the potential risk factors on baPWV and PWV change rates. All analyses were stratified by diabetes. Mediation analysis was also conducted to demonstrate the mechanisms of arterial stiffness in patients with diabetes. Results In lasso regression, postprandial 2-h glucose (P2hG), systolic blood pressure (SBP) and age were associated with baPWV regardless of diabetes. Platelet counts (PLT), mean corpuscular volume (MCV) and coronary heart disease (CHD) were associated with baPWV in patients with diabetes. In the mixed models, PLT were positively associated with baPWV in patients with diabetes (βplatelet, perSD = 25.80; 18.26–33.33). Elevated PLTs could also significantly increase the PWV change rate in patients with diabetes (βplatelet, perSD = 54.05; 10.00–107.10). In mediation analysis, diabetes had a significant average direct effect on baPWV. The average causal mediation effect (ACME) of PLTs was 1.76, with a range of 0.17 to 3.70. Conclusions Elevated PLT counts can increase baPWV in diabetes and are a potential mediator between diabetes and atherosclerosis.
Collapse
Affiliation(s)
- Kuo Liu
- Department of Epidemiology and Health statistics, School of Public Health, Capital Medical University, No.10 Xitoutiao, Youanmenwai Avenue, Beijing, 100069, People's Republic of China.,Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Junfeng Xu
- Beijing Xiaotangshan Hospital, Xiaotangshan Town, Changping District, Beijing, 102211, China
| | - Lixin Tao
- Department of Epidemiology and Health statistics, School of Public Health, Capital Medical University, No.10 Xitoutiao, Youanmenwai Avenue, Beijing, 100069, People's Republic of China.,Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Kun Yang
- Department of Epidemiology and Health statistics, School of Public Health, Capital Medical University, No.10 Xitoutiao, Youanmenwai Avenue, Beijing, 100069, People's Republic of China.,Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Yang Sun
- Department of Epidemiology and Health statistics, School of Public Health, Capital Medical University, No.10 Xitoutiao, Youanmenwai Avenue, Beijing, 100069, People's Republic of China.,Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Xiuhua Guo
- Department of Epidemiology and Health statistics, School of Public Health, Capital Medical University, No.10 Xitoutiao, Youanmenwai Avenue, Beijing, 100069, People's Republic of China. .,Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China.
| |
Collapse
|
25
|
Yasuda MT, Watanabe Y, Hosono T, Seki T, Suzuki TA, Sakakibara H, Shimoi K. Nocturnal light exposure stimulates the cardiac fibrinolysis system and stress responses in C3H/He mice. Thromb Res 2020; 188:79-81. [PMID: 32109771 DOI: 10.1016/j.thromres.2020.02.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 02/03/2020] [Accepted: 02/05/2020] [Indexed: 11/29/2022]
Affiliation(s)
- Michiko T Yasuda
- Department of Human Nutrition, School of Life Studies, Sugiyama Jogakuen University, 17-3 Hoshigaoka Motomachi, Chikusa-ku, Nagoya 464-8662, Japan; Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan.
| | - Yuto Watanabe
- Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Takashi Hosono
- Department of Chemistry and Life Science, College of Bioresource Sciences, Nihon University, 1866 Kameino, Fujisawa, Kanagawa 252-0880, Japan
| | - Taiichiro Seki
- Department of Chemistry and Life Science, College of Bioresource Sciences, Nihon University, 1866 Kameino, Fujisawa, Kanagawa 252-0880, Japan
| | - Taka-Aki Suzuki
- Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; Industrial Research Institute of Shizuoka Prefecture, 2078 Makigaya, Aoi-ku, Shizuoka 421-1298, Japan
| | - Hiroyuki Sakakibara
- Faculty of Agriculture, University of Miyazaki, 1-1 Gakuen-kibanadai-nishi, Miyazaki 889-2192, Japan
| | - Kayoko Shimoi
- Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; School of Food and Nutritional Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan.
| |
Collapse
|
26
|
Beck R, Stachs O, Koschmieder A, Mueller-Lierheim WGK, Peschel S, van Setten GB. Hyaluronic Acid as an Alternative to Autologous Human Serum Eye Drops: Initial Clinical Results with High-Molecular-Weight Hyaluronic Acid Eye Drops. Case Rep Ophthalmol 2019; 10:244-255. [PMID: 31692577 PMCID: PMC6760367 DOI: 10.1159/000501712] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 06/11/2019] [Indexed: 12/05/2022] Open
Abstract
Introduction Autologous serum eye drops (ASED) are used in the treatment of most severe stages of dry eye. Once introduced, it is currently considered impossible to return to other lubricating eye drops or other commercially available therapeutic regimen. Materials and Methods In a randomized study, non-preserved high-molecular-weight hyaluronic acid eye drops were offered as an alternative to 11 patients using autologous serum treatment for at least 3 months. The control group (n = 5) continued their treatment with ASED. The verum group (n = 6) used very-high-molecular-weight hyaluronic eye drops (Comfort Shield®) instead of the ASED. Results From four of initially six patients in the verum group that finished the study, 2 (50%) preferred to stay with the very-high-molecular-weight hyaluronic acid eye drops beyond the trial period, the other two returned to the earlier therapy with ASED. The control group continued their treatment as before and finished the study after 8 weeks. Conclusion For the first time, artificial eye drops, i.e., high-molecular-weight hyaluronic acid eye drops, offered an acceptable alternative to ASED. Some patients perceived these drops as even better than the patient's own serum. This is the first evidence that optimization of the molecular structure of hyaluronic acid can be used to create eye drops that are perceived to be better than other tested tear substitutes and even patients' own serum. This offers a new treatment perspective for patients with very severe dry eye disease.
Collapse
Affiliation(s)
- Ria Beck
- University Eye Hospital, Rostock, Germany
| | | | | | | | | | - Gysbert-Botho van Setten
- St Eriks Eye Hospital, Karolinska Institutet, Stockholm, Sweden.,Institute of Wound Research and Department of Ophthalmology, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
27
|
Guedes AF, Moreira C, Nogueira JB, Santos NC, Carvalho FA. Fibrinogen-erythrocyte binding and hemorheology measurements in the assessment of essential arterial hypertension patients. NANOSCALE 2019; 11:2757-2766. [PMID: 30672545 DOI: 10.1039/c8nr04398a] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Some studies have reported a positive association between plasma fibrinogen levels, erythrocyte aggregation and essential arterial hypertension (EAH). The aim of this study was to understand how the interaction between fibrinogen and its erythrocyte membrane receptor is altered in EAH. EAH patients (n = 31) and healthy blood donors (n = 65) were enrolled in the study. EAH patients were therapeutically controlled for the disease, presenting a systolic blood pressure between 108 and 180 mmHg and a diastolic blood pressure between 66 and 123 mmHg. Clinical evaluation included blood pressure monitoring, electrocardiography, echocardiography and blood cell count. The hemorheological parameters were also analyzed. Fibrinogen-erythrocyte binding force and frequency were evaluated quantitatively, at the single-molecule level, using atomic force microscopy (AFM). Changes in erythrocyte elasticity were also evaluated. Force spectroscopy data showed that the average fibrinogen-erythrocyte binding forces increase from 40.4 ± 3.0 pN in healthy donors to 73.8 ± 8.1 pN in patients with EAH, despite a lower binding frequency for patients compared to the control group (7.9 ± 1.6% vs. 27.6 ± 4.2%, respectively). Elasticity studies revealed an increase of erythrocyte stiffness in the patients. The stronger fibrinogen binding to erythrocytes from EAH patients and alteration in cell elasticity may lead to changes in the whole blood flow. The patients' altered hemorheological parameters may also contribute to these blood flow perturbations. The transient bridging of two erythrocytes, by the simultaneous binding of fibrinogen to both of them, promoting erythrocyte aggregation, could represent an important cardiovascular risk factor.
Collapse
Affiliation(s)
- Ana Filipa Guedes
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.
| | | | | | | | | |
Collapse
|
28
|
Tsujimoto T, Kajio H. Thrombotic/Thrombolytic Balance as a Cardiac Treatment Determinant in Patients With Diabetes Mellitus and Coronary Artery Disease. J Am Heart Assoc 2019; 8:e011207. [PMID: 30646801 PMCID: PMC6497335 DOI: 10.1161/jaha.118.011207] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background This study aimed to assess whether the plasminogen activator inhibitor‐1/tissue plasminogen activator (PAI‐1/tPA) ratio as a prothrombotic state is useful for optimizing cardiac treatment strategy. Methods and Results Using BARI 2D (Bypass Angioplasty Revascularization Investigation 2 Diabetes) trial data, we used a Cox proportional hazard model to calculate hazard ratios with 95% CIs for cardiac events in patients receiving early revascularization (percutaneous coronary intervention or coronary artery bypass grafting) or medical therapy, separately in patients with low (n=1276) and high (n=894) PAI‐1/tPA ratios. The primary outcome was major cardiac events, which was a composite end point including cardiac death and nonfatal myocardial infarction. The mean±SD follow‐up period was 4.1±1.7 years. The risk of major cardiac events in patients with high PAI‐1/tPA ratio was significantly higher when receiving percutaneous coronary intervention (hazard ratio, 1.84; 95% CI, 1.16–2.93; P=0.01) than when receiving medical therapy, whereas that in patients with low PAI‐1/tPA ratio did not differ significantly between the groups (hazard ratio, 0.95; 95% CI, 0.66–1.36; P=0.77); the interaction between the cardiac treatment strategy and PAI‐1/tPA ratio was significant (P=0.02). However, regardless of the PAI‐1/tPA ratio, major cardiac event risk seemed to be lower in patients receiving coronary artery bypass grafting than in those receiving medical therapy. Conclusions In patients with type 2 diabetes mellitus and coronary artery disease, this study demonstrated that those with high PAI‐1/tPA ratio were at higher risks of major cardiac events when treated with percutaneous coronary intervention than when treated with intensive medical therapy.
Collapse
Affiliation(s)
- Tetsuro Tsujimoto
- 1 Department of Diabetes, Endocrinology, and Metabolism Center Hospital National Center for Global Health and Medicine Tokyo Japan
| | - Hiroshi Kajio
- 1 Department of Diabetes, Endocrinology, and Metabolism Center Hospital National Center for Global Health and Medicine Tokyo Japan
| |
Collapse
|
29
|
Pavlov M, Ćelap I. Plasminogen activator inhibitor 1 in acute coronary syndromes. Clin Chim Acta 2019; 491:52-58. [PMID: 30659821 DOI: 10.1016/j.cca.2019.01.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 01/14/2019] [Accepted: 01/15/2019] [Indexed: 11/24/2022]
Abstract
Plasminogen activator inhibitor 1 (PAI-1) is the main regulator of endogenous fibrinolysis, overriding the impact of other constituents of fibrinolysis. In plasma, it can be found in three forms: active, latent and inactive. There are numerous commercially available tests, analysing the activity of PAI-1 or the antigen level, with variable correlations between the two. PAI-1 has been extensively studied regarding incidence and outcomes of acute coronary syndromes, and showed positive association with both in numerous studies. Higher PAI-1 has been associated with worse short- and long-term outcomes. Studies are more consistent in the primary percutaneous coronary intervention era. Higher rise of PAI-1 within the first 24 h of acute myocardial infarction has been linked to some of its high-risk features. The circadian pattern of PAI-1 kinetics has been previously described, and the mechanisms behind this phenomenon and its impact on the incidence of acute coronary syndromes are well known. Further investigations are needed to test the safety and efficacy of PAI-1 as a pharmacological target in cardiovascular diseases.
Collapse
Affiliation(s)
- Marin Pavlov
- Department of Cardiology, Sestre milosrdnice University Hospital Centre, Vinogradska cesta 29, 10000 Zagreb, Croatia.
| | - Ivana Ćelap
- Department of Clinical Chemistry, Sestre milosrdnice University Hospital Centre, Vinogradska cesta 29, 10000 Zagreb, Croatia
| |
Collapse
|
30
|
An ATF6-tPA pathway in hepatocytes contributes to systemic fibrinolysis and is repressed by DACH1. Blood 2018; 133:743-753. [PMID: 30504459 DOI: 10.1182/blood-2018-07-864843] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 11/13/2018] [Indexed: 01/18/2023] Open
Abstract
Tissue-type plasminogen activator (tPA) is a major mediator of fibrinolysis and, thereby, prevents excessive coagulation without compromising hemostasis. Studies on tPA regulation have focused on its acute local release by vascular cells in response to injury or other stimuli. However, very little is known about sources, regulation, and fibrinolytic function of noninjury-induced systemic plasma tPA. We explore the role and regulation of hepatocyte-derived tPA as a source of basal plasma tPA activity and as a contributor to fibrinolysis after vascular injury. We show that hepatocyte tPA is downregulated by a pathway in which the corepressor DACH1 represses ATF6, which is an inducer of the tPA gene Plat Hepatocyte-DACH1-knockout mice show increases in liver Plat, circulating tPA, fibrinolytic activity, bleeding time, and time to thrombosis, which are reversed by silencing hepatocyte Plat Conversely, hepatocyte-ATF6-knockout mice show decreases in these parameters. The inverse correlation between DACH1 and ATF6/PLAT is conserved in human liver. These findings reveal a regulated pathway in hepatocytes that contributes to basal circulating levels of tPA and to fibrinolysis after vascular injury.
Collapse
|
31
|
Peroxisome proliferator-activated receptor α attenuates high-cholesterol diet-induced toxicity and pro-thrombotic effects in mice. Arch Toxicol 2018; 93:149-161. [PMID: 30341732 DOI: 10.1007/s00204-018-2335-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 10/10/2018] [Indexed: 02/07/2023]
Abstract
Peroxisome proliferator-activated receptor α (PPARα) is involved in the regulation of fatty acid and cholesterol metabolism. A high-cholesterol (HC) diet increases the risk of developing cardiovascular diseases (CVD); however, it is unclear whether the toxic effects of cholesterol involve changes in thrombotic factor expression, and whether PPARα is necessary for such effects. To investigate this possibility, we fed a HC diet to wild-type (WT) and Ppara-null mice and measured cholesterol and triglyceride contents, liver histology, serum/plasma levels of coagulation factors, hepatic expression of the coagulation factors, liver/serum sulfatide levels, hepatic sulfatide metabolism, hepatic expression of lipid transporters, and hepatic oxidative stress and its relating enzymes. In Ppara-null mice, the HC diet caused triglyceride accumulation and exacerbated inflammation and oxidative stress in liver, increased levels of coagulation factors, including tissue factor, plasminogen activator inhibitor-1 and carboxypeptidase B2 in blood and liver, and decreased levels of anti-thrombotic sulfatides in serum and liver. These changes were much less marked in WT mice. These findings imply that cholesterol overload exerts its toxic effects at least in part by enhancing thrombosis, secondary to abnormal hepatic lipid metabolism, inflammation, and oxidative stress. Moreover, we reveal for the first time that PPARα can attenuate these toxic effects by transcriptional regulation of coagulation factors and sulfatides, in addition to its known effects of controlling lipid homeostasis and suppressing inflammation and oxidative stress. Therapies aimed at activating PPARα might prevent HC diet-induced CVD through modulating various pro- and anti-thrombotic factors.
Collapse
|
32
|
Corban MT, Prasad A, Nesbitt L, Loeffler D, Herrmann J, Lerman LO, Lerman A. Local Production of Soluble Urokinase Plasminogen Activator Receptor and Plasminogen Activator Inhibitor-1 in the Coronary Circulation Is Associated With Coronary Endothelial Dysfunction in Humans. J Am Heart Assoc 2018; 7:e009881. [PMID: 30371230 PMCID: PMC6201458 DOI: 10.1161/jaha.118.009881] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 06/21/2018] [Indexed: 01/11/2023]
Abstract
Background Soluble urokinase plasminogen activator receptor (su PAR ) is a proinflammatory biomarker associated with immune activation and fibrinolysis inhibition. Plasminogen activator inhibitor ( PAI -1) is associated with excessive fibrin accumulation, thrombus formation, and atherosclerosis. The relationship between cross-coronary su PAR and PAI -1 production and endothelial dysfunction remains unknown. Methods and Results Seventy-nine patients (age 53±10 years, 75% women) with angina and normal coronary arteries or mild coronary artery disease (<40% stenosis) on angiogram underwent acetylcholine assessment of epicardial endothelial dysfunction (mid-left anterior descending coronary artery diameter decrease >20% after acetylcholine) and mircovascular endothelial dysfunction (coronary blood flow change <50% after acetylcholine). Simultaneous left main and coronary sinus su PAR and PAI -1 levels were measured in each patient before acetylcholine administration, and cross-coronary su PAR and PAI -1 production rates were calculated. Patients' characteristics, except for age (51±10 versus 57±9, P=0.02), and resting coronary hemodynamics were not significantly different between patients with (26%) versus without (74%) epicardial endothelial dysfunction. Patients' characteristics and resting coronary hemodynamics were not significantly different between those with (62%) and those without (38%) mircovascular endothelial dysfunction. Patients with mircovascular endothelial dysfunction demonstrated local coronary su PAR production versus su PAR extraction in patients with normal microvascular function (median 25.8 [interquartile range 121.6, -23.7] versus -12.7 [52.0, -74.8] ng/min, P=0.03). Patients with epicardial endothelial dysfunction had higher median coronary PAI -1 production rates compared with those with normal epicardial endothelial function (1224.7 [12 940.7, -1915.4] versus -187.4 [4444.7, -4535.8] ng/min, P=0.03). Conclusions su PAR is released in coronary circulation of patients with mircovascular endothelial dysfunction and extracted in those with normal microvascular function. Cross-coronary PAI -1 release is higher in humans with epicardial endothelial dysfunction.
Collapse
Affiliation(s)
- Michel T. Corban
- Department of Cardiovascular DiseasesMayo Clinic College of Medicine and ScienceRochesterMN
| | - Abhiram Prasad
- Department of Cardiovascular DiseasesMayo Clinic College of Medicine and ScienceRochesterMN
| | - Lisa Nesbitt
- Department of Cardiovascular DiseasesMayo Clinic College of Medicine and ScienceRochesterMN
| | - Darrell Loeffler
- Department of Cardiovascular DiseasesMayo Clinic College of Medicine and ScienceRochesterMN
| | - Joerg Herrmann
- Department of Cardiovascular DiseasesMayo Clinic College of Medicine and ScienceRochesterMN
| | - Lilach O. Lerman
- Department of Cardiovascular DiseasesMayo Clinic College of Medicine and ScienceRochesterMN
- Division of Nephrology and HypertensionDepartment of MedicineMayo Clinic College of Medicine and ScienceRochesterMN
| | - Amir Lerman
- Department of Cardiovascular DiseasesMayo Clinic College of Medicine and ScienceRochesterMN
| |
Collapse
|
33
|
Association of plasminogen activator inhibitor-1 and low-density lipoprotein heterogeneity as a risk factor of atherosclerotic cardiovascular disease with triglyceride metabolic disorder: a pilot cross-sectional study. Coron Artery Dis 2018; 28:577-587. [PMID: 28692480 DOI: 10.1097/mca.0000000000000521] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND We hypothesized that an increase in plasminogen activator inhibitor 1 (PAI-1) might reduce low-density lipoprotein (LDL) particle size in conjunction with triglyceride (TG) metabolism disorder, resulting in an increased risk of atherosclerotic cardiovascular disease (ASCVD). METHODS This study was carried out as a hospital-based cross-sectional study in 537 consecutive outpatients (mean age: 64 years; men: 71%) with one or more risk factors for ASCVD from April 2014 to October 2014 at the Cardiovascular Center of Nihon University Surugadai Hospital. The estimated LDL-particle size was measured as relative LDL migration using polyacrylamide gel electrophoresis with the LipoPhor system.The plasma PAI-1 level, including the tissue PA/PAI-1 complex and the active and latent forms of PAI-1, was determined using a latex photometric immunoassay method. RESULTS A multivariate regression analysis after adjustments for ASCVD risk factors showed that an elevated PAI-1 level was an independent predictor of smaller-sized LDL-particle in both the overall patients population (β=0.209, P<0.0001) and a subset of patients with a serum low-density lipoprotein cholesterol (LDL-C) level lower than 100 mg/dl (β=0.276, P<0.0001). Furthermore, an increased BMI and TG-rich lipoprotein related markers [TG, remnant-like particle cholesterol, apolipoprotein (apo) B, apo C-II, and apo C-III] were found to be independent variables associated with an increased PAI-1 level in multivariate regression models. A statistical analysis of data from nondiabetic patients with well-controlled serum LDL-C levels yielded similar findings. Furthermore, in the 310 patients followed up for at least 6 months, a multiple-logistic regression analysis after adjustments for ASCVD risk factors identified the percent changes of the plasma PAI-1 level in the third tertile compared with those in the first tertile as being independently predictive of decreased LDL-particle size [odds ratio (95% confidence interval): 2.11 (1.12/3.40), P=0.02]. CONCLUSION The plasma PAI-1 levels may be determined by the degree of obesity and TG metabolic disorders. These factors were also shown to be correlated with a decreased LDL-particle size, increasing the risk of ASCVD, even in nondiabetic patients with well-controlled serum LDL-C levels.
Collapse
|
34
|
Lalić K, Nedeljković M, Jotić A, Babić R, Rajković N, Popović L, Lukić L, Miličić T, Singh Lukač S, Stošić L, Maćešić M, Rasulić I, Gajović JS, Lalić NM. Endothelial dysfunction of coronary arteries in subjects without diabetes: An association with both insulin resistance and impaired insulin secretion response. Diabetes Res Clin Pract 2018. [PMID: 29526680 DOI: 10.1016/j.diabres.2018.03.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
AIMS This study was aimed to compare insulin sensitivity and secretion response, lipoprotein and plasminogen activator inhibitor 1 (PAI-1) levels between the subjects with and without coronary artery endothelial dysfunction (ED). METHODS ED was detected by intracoronary injection of acetylcholine (ACh) in 47 nondiabetes subjects without stenotic coronary arteries, selected from 316 consecutive patients with coronary angiography performed for suspected coronary artery disease. The subjects were divided into two groups: presence of ACh-induced coronary spasm (group ED+, N = 30) and absence of ACh-induced coronary spasm (group ED-, N = 17). Insulin sensitivity (Si) was evaluated by frequently sampled intravenous glucose tolerance test (FSIGTT) with minimal model analysis and by HOMA-IR, insulin secretion by acute insulin response (AIR) (calculated from the first 8 min of FSIGTT) and by disposition index (DI) (Si × AIR). Lipids and PAI-1 levels were determined enzymatically, and LDL particle size by gradient gel electrophoresis. RESULTS Si was significantly lower (4.22 ± 0.62 vs 6.98 ± 1.47 min-1/mU/l × 104; p < 0.05) while HOMA-IR was significantly higher in ED + group vs ED- group (2.8 ± 0.3 vs 1.7 ± 0.2; p < 0.05). Simultaneously, AIR and DI was significantly lower in ED + vs ED- groups (p < 0.05 and p < 0.01, respectively). Investigated groups did not differ in fasting lipid levels but ED+ group had significantly smaller LDL particles (p < 0.01) and higher PAI-1 levels (p < 0.05). Regression analysis shown that DI was a strong independent predictor of appearance of ED, together with PAI-1 and LDL particle size. CONCLUSIONS Both insulin resistance and impairment in insulin secretion response strongly correlate with coronary ED in subjects without diabetes.
Collapse
Affiliation(s)
- Katarina Lalić
- Faculty of Medicine University of Belgrade, Dr. Subotića 8, 11000 Belgrade, Serbia; Clinic for Endocrinology, Diabetes and Metabolic Diseases, Clinical Center of Serbia, Dr. Subotića 13, 11000 Belgrade, Serbia.
| | - Milan Nedeljković
- Faculty of Medicine University of Belgrade, Dr. Subotića 8, 11000 Belgrade, Serbia; Clinic for Cardiology, Clinical Center of Serbia, Koste Todorovića 8, 11000 Belgrade, Serbia.
| | - Alekasandra Jotić
- Faculty of Medicine University of Belgrade, Dr. Subotića 8, 11000 Belgrade, Serbia; Clinic for Endocrinology, Diabetes and Metabolic Diseases, Clinical Center of Serbia, Dr. Subotića 13, 11000 Belgrade, Serbia.
| | - Rade Babić
- Faculty of Medicine University of Belgrade, Dr. Subotića 8, 11000 Belgrade, Serbia; Clinic for Cardiology, Clinical Center of Serbia, Koste Todorovića 8, 11000 Belgrade, Serbia.
| | - Nataša Rajković
- Faculty of Medicine University of Belgrade, Dr. Subotića 8, 11000 Belgrade, Serbia; Clinic for Endocrinology, Diabetes and Metabolic Diseases, Clinical Center of Serbia, Dr. Subotića 13, 11000 Belgrade, Serbia.
| | - Ljiljana Popović
- Faculty of Medicine University of Belgrade, Dr. Subotića 8, 11000 Belgrade, Serbia; Clinic for Endocrinology, Diabetes and Metabolic Diseases, Clinical Center of Serbia, Dr. Subotića 13, 11000 Belgrade, Serbia.
| | - Ljiljana Lukić
- Faculty of Medicine University of Belgrade, Dr. Subotića 8, 11000 Belgrade, Serbia; Clinic for Endocrinology, Diabetes and Metabolic Diseases, Clinical Center of Serbia, Dr. Subotića 13, 11000 Belgrade, Serbia.
| | - Tanja Miličić
- Faculty of Medicine University of Belgrade, Dr. Subotića 8, 11000 Belgrade, Serbia; Clinic for Endocrinology, Diabetes and Metabolic Diseases, Clinical Center of Serbia, Dr. Subotića 13, 11000 Belgrade, Serbia.
| | - Sandra Singh Lukač
- Clinic for Endocrinology, Diabetes and Metabolic Diseases, Clinical Center of Serbia, Dr. Subotića 13, 11000 Belgrade, Serbia.
| | - Ljubica Stošić
- Clinic for Endocrinology, Diabetes and Metabolic Diseases, Clinical Center of Serbia, Dr. Subotića 13, 11000 Belgrade, Serbia.
| | - Marija Maćešić
- Faculty of Medicine University of Belgrade, Dr. Subotića 8, 11000 Belgrade, Serbia; Clinic for Endocrinology, Diabetes and Metabolic Diseases, Clinical Center of Serbia, Dr. Subotića 13, 11000 Belgrade, Serbia.
| | - Iva Rasulić
- Clinic for Endocrinology, Diabetes and Metabolic Diseases, Clinical Center of Serbia, Dr. Subotića 13, 11000 Belgrade, Serbia.
| | - Jelena Stanarčić Gajović
- Clinic for Endocrinology, Diabetes and Metabolic Diseases, Clinical Center of Serbia, Dr. Subotića 13, 11000 Belgrade, Serbia.
| | - Nebojša M Lalić
- Faculty of Medicine University of Belgrade, Dr. Subotića 8, 11000 Belgrade, Serbia; Clinic for Endocrinology, Diabetes and Metabolic Diseases, Clinical Center of Serbia, Dr. Subotića 13, 11000 Belgrade, Serbia.
| |
Collapse
|
35
|
Maloberti A, Farina F, Carbonaro M, Piccinelli E, Bassi I, Pansera F, Grassi G, Mancia G, Palestini P, Giannattasio C. In healthy normotensive subjects age and blood pressure better predict subclinical vascular and cardiac organ damage than atherosclerosis biomarkers. Blood Press 2018; 27:262-270. [PMID: 29648468 DOI: 10.1080/08037051.2018.1461010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE Only few studies evaluated biomarkers useful for defining the cardiovascular risk of a subject in a pre-clinical condition (i.e. healthy subjects). In this context we sought to determine the relationships of Plasminogen activator inhibitor type 1 (PAI-1), P-Selectin, Tissue Inhibitors Metalloproteinases type 1 (TIMP-1) and Cystatin-C with subclinical Target Organ Damage (TOD) in normotensive and normoglycemic subjects without known cardiovascular and kidney diseases. MATERIALS AND METHODS 480 blood donors participated at the present analysis. TOD was evaluated as Pulse Wave Velocity (PWV), Left Ventricular Hypertrophy (LVH) and Intima Media Thickness (IMT) and carotid plaque presence) grouped together under carotid TOD. RESULTS 3.1% of the subjects showed a PWV higher than 10 m/sec with those subjects exerting significantly lower values of P-Selectine (0.068 ± 0.015 vs 0.08 ± 0.036 mg/L, p = .014). 8.8% of the subjects showed carotid TOD that was associated with higher Cystatin-C values (0.67 ± 0.17 vs 0.63 ± 0.14 mg/L, p = .045). Finally 23.8% of the subjects showed LVH with no significant differences regarding biomarkers. Despite some significant correlations between biomarkers and TOD, at the multivariate analysis none came out to be as significant predictor of the assessed TOD. CONCLUSIONS in normotensive and normoglycemic healthy subjects, the evaluated biomarkers of atherosclerotic process didn't show any significant association with cardiac, carotid and vascular TOD while age and BP are its principal predictors.
Collapse
Affiliation(s)
- Alessandro Maloberti
- a Cardiology IV, "A.De Gasperis" Department , Ospedale Niguarda Ca' Granda , Milan , Italy.,b School of Medicine and Surgery , Milano-Bicocca University , Milan , Italy
| | - Francesca Farina
- b School of Medicine and Surgery , Milano-Bicocca University , Milan , Italy
| | - Marco Carbonaro
- b School of Medicine and Surgery , Milano-Bicocca University , Milan , Italy
| | - Enrico Piccinelli
- a Cardiology IV, "A.De Gasperis" Department , Ospedale Niguarda Ca' Granda , Milan , Italy.,b School of Medicine and Surgery , Milano-Bicocca University , Milan , Italy
| | - Ilaria Bassi
- a Cardiology IV, "A.De Gasperis" Department , Ospedale Niguarda Ca' Granda , Milan , Italy.,b School of Medicine and Surgery , Milano-Bicocca University , Milan , Italy
| | - Francesco Pansera
- a Cardiology IV, "A.De Gasperis" Department , Ospedale Niguarda Ca' Granda , Milan , Italy.,b School of Medicine and Surgery , Milano-Bicocca University , Milan , Italy
| | - Guido Grassi
- b School of Medicine and Surgery , Milano-Bicocca University , Milan , Italy.,c IRCCS Multimedica , Milan , Italy
| | - Giuseppe Mancia
- b School of Medicine and Surgery , Milano-Bicocca University , Milan , Italy
| | - Paola Palestini
- b School of Medicine and Surgery , Milano-Bicocca University , Milan , Italy
| | - Cristina Giannattasio
- a Cardiology IV, "A.De Gasperis" Department , Ospedale Niguarda Ca' Granda , Milan , Italy.,b School of Medicine and Surgery , Milano-Bicocca University , Milan , Italy
| |
Collapse
|
36
|
Li B, Hu J, Chen X. MicroRNA-30b protects myocardial cell function in patients with acute myocardial ischemia by targeting plasminogen activator inhibitor-1. Exp Ther Med 2018; 15:5125-5132. [PMID: 29805539 DOI: 10.3892/etm.2018.6039] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Accepted: 01/18/2018] [Indexed: 12/26/2022] Open
Abstract
The aim of the present study was to determine the expression of plasminogen activator inhibitor-1 (PAI-1) and microRNA (miR)-30b in the blood of patients with acute myocardial ischemia (AMI) and in the blood and myocardial tissue of mice with AMI. In addition, the present study aimed to identify the mechanism of action of miR-30b in AMI. A total of 36 patients with AMI were included in the present study and 28 healthy subjects were included as a control. Peripheral blood was collected from all subjects. For animal experiments, mice in the AMI group received an intraperitoneal injection of pituitrin (20 U/kg), whereas mice in the negative control group received an intraperitoneal injection of the same volume of saline. Blood and myocardial tissue was collected from all mice for analysis. Reverse transcription-quantitative polymerase chain reaction was performed to determine the expression of PAI-1 mRNA and miR-30b in the serum and myocardial tissue. An enzyme-linked immunosorbent assay was performed to measure the expression of PAI-1 protein in the serum of humans and mice, whereas western blotting was performed to determine the expression of PAI-1 protein in mouse myocardial tissue. Catalase, glutathione peroxidase and superoxide dismutase activity was measured using an automatic biochemical analyzer. A dual luciferase assay was performed to identify the interactions between PAI-1 mRNA and miR-30b. The results indicated that patients with AMI have higher PAI-1 levels and lower miR-30b expression in the peripheral blood compared with healthy subjects. AMI damaged the myocardium tissue of mice and reduced catalase, glutathione peroxidase and superoxide dismutase activity. Mice that have undergone AMI exhibit increased PAI-1 levels but decreased miR-30b expression in the peripheral blood and myocardial tissues. It was also demonstrated that miR-30b is able to bind to the 3'-untranslated region of PAI-1 mRNA to regulate its expression. The present study demonstrates that patients with AMI exhibit decreased miR-30b expression and elevated PAI-1 expression in the peripheral blood. miR-30b may therefore inhibit the damage to myocardial cells that occurs following AMI and protect myocardial cell function by targeting PAI-1 expression.
Collapse
Affiliation(s)
- Bin Li
- Department of Cardiac Surgery, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan 471009, P.R. China
| | - Jie Hu
- Department of Cardiac Surgery, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan 471009, P.R. China
| | - Xingpeng Chen
- Department of Cardiac Surgery, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan 471009, P.R. China
| |
Collapse
|
37
|
Mice with diet-induced obesity demonstrate a relative prothrombotic factor profile and a thicker aorta with reduced ex-vivo function. Blood Coagul Fibrinolysis 2018; 29:257-266. [DOI: 10.1097/mbc.0000000000000713] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
38
|
Hmimech W, Idrissi HH, Diakite B, Korchi F, Baghdadi D, Tahri Joutey Hassani Idrissi H, Haboub M, Habbal R, Nadifi S. Impact of I/D polymorphism of angiotensin-converting enzyme (ACE) gene on myocardial infarction susceptibility among young Moroccan patients. BMC Res Notes 2017; 10:763. [PMID: 29268798 PMCID: PMC5740925 DOI: 10.1186/s13104-017-3039-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 11/30/2017] [Indexed: 11/13/2022] Open
Abstract
Objective Our case–control study aimed to access the potential association of insertion/deletion (I/D) ACE (angiotensin converting enzyme) gene polymorphism with myocardial infarction (MI) risk of occurrence among a sample of Moroccan patients, especially young ones. Results Distribution of I/D ACE gene variant among cases vs controls, showed that healthy controls carried out higher frequency of wild type allele I compared to cases (23.5% vs 21.79% respectively), when cases were carrying higher frequency of mutant allele D (78.21% vs 76.5% for controls). Patients were-after this- divided into two groups of < 45 and > 55 years of age, to investigate whether or not younger patients carried out higher frequency of the mutant allele D, than older ones. As expected, < 45 years old patients carried out more DD genotype than older ones (68.9% vs 54.6% respectively), and higher frequency of mutant allele D (81.08% vs 75% respectively). Besides, a tendency to a positive association was found under the recessive genetic transmission model (OR [95% CI] = 1.85 [0.93–3.69], P = 0.08), suggesting that the I/D ACE polymorphism may be associated with MI occurrence among younger patients (< 45 years of age). Electronic supplementary material The online version of this article (10.1186/s13104-017-3039-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wiam Hmimech
- Laboratory of Genetics and Molecular Pathology, Medical School, University Hassan II, 19, Street TarikIbnouZiad, B. P: 9154, Casablanca, Morocco
| | - Hind Hassani Idrissi
- Laboratory of Genetics and Molecular Pathology, Medical School, University Hassan II, 19, Street TarikIbnouZiad, B. P: 9154, Casablanca, Morocco.
| | - Brehima Diakite
- Laboratory of Genetics and Molecular Pathology, Medical School, University Hassan II, 19, Street TarikIbnouZiad, B. P: 9154, Casablanca, Morocco
| | - Farah Korchi
- Department of Cardiology, University Hospital Center IbnRochd, Casablanca, Morocco
| | - Dalila Baghdadi
- Department of Cardiology, University Hospital Center IbnRochd, Casablanca, Morocco
| | | | - Meriem Haboub
- Department of Cardiology, University Hospital Center IbnRochd, Casablanca, Morocco
| | - Rachida Habbal
- Department of Cardiology, University Hospital Center IbnRochd, Casablanca, Morocco
| | - Sellama Nadifi
- Laboratory of Genetics and Molecular Pathology, Medical School, University Hassan II, 19, Street TarikIbnouZiad, B. P: 9154, Casablanca, Morocco
| |
Collapse
|
39
|
Olatunji LA, Usman TO, Akinade AI, Adeyanju OA, Kim I, Soladoye AO. Low-dose spironolactone ameliorates insulin resistance and suppresses elevated plasminogen activator inhibitor-1 during gestational testosterone exposure. Arch Physiol Biochem 2017; 123:286-292. [PMID: 28480754 DOI: 10.1080/13813455.2017.1320681] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
CONTEXT Elevated gestational circulating testosterone has been associated with pathological pregnancies that increase the risk of development of cardiometabolic disorder in later life. OBJECTIVE We hypothesised that gestational testosterone exposure, in late pregnancy, causes glucose deregulation and atherogenic dyslipidaemia that would be accompanied by high plasminogen activator inhibitor-1 (PAI-1). The study also hypothesise that low-dose spironolactone treatment would ameliorate these effects. METHODS Pregnant Wistar rats received vehicle, testosterone (0.5 mg/kg; sc), spironolactone (0.5 mg/kg, po) or testosterone and spironolactone daily between gestational days 15 and 19. RESULTS Gestational testosterone exposure led to increased HOMA-IR, circulating insulin, testosterone, 1-h post-load glucose, atherogenic dyslipidaemia, PLR, PAI-1 and MDA. However, all these effects, except that of circulating testosterone, were ameliorated by spironolactone. CONCLUSIONS These results demonstrate that low-dose spironolactone ameliorates glucose deregulation and atherogenic dyslipidaemia during elevated gestational testosterone exposure, at least in part, by suppressing elevated PAI-1.
Collapse
Affiliation(s)
- Lawrence A Olatunji
- a Department of Physiology, Cardiovascular Research Laboratory , College of Health Sciences, University of Ilorin , Ilorin , Nigeria
| | - Taofeek O Usman
- a Department of Physiology, Cardiovascular Research Laboratory , College of Health Sciences, University of Ilorin , Ilorin , Nigeria
- b Department of Physiology, Cardiovascular Unit, College of Health sciences , Osun State University , Osogbo , Nigeria
| | - Aminat I Akinade
- a Department of Physiology, Cardiovascular Research Laboratory , College of Health Sciences, University of Ilorin , Ilorin , Nigeria
| | - Oluwaseun A Adeyanju
- a Department of Physiology, Cardiovascular Research Laboratory , College of Health Sciences, University of Ilorin , Ilorin , Nigeria
| | - InKyeom Kim
- c Department of Pharmacology, Cardiovascular Research Institute , Kyungpook National University School of Medicine , Daegu , Republic of Korea
| | - Ayodele O Soladoye
- a Department of Physiology, Cardiovascular Research Laboratory , College of Health Sciences, University of Ilorin , Ilorin , Nigeria
| |
Collapse
|
40
|
Peng S, Xue G, Gong L, Fang C, Chen J, Yuan C, Chen Z, Yao L, Furie B, Huang M. A long-acting PAI-1 inhibitor reduces thrombus formation. Thromb Haemost 2017; 117:1338-1347. [DOI: 10.1160/th16-11-0891] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 03/25/2017] [Indexed: 01/05/2023]
Abstract
SummaryPlasminogen activator inhibitor 1 (PAI-1) is the main inhibitor of tissue-type and urokinase-type plasminogen activators (t/uPA) and plays an important role in fibrinolysis. Inhibition of PAI-1 activity prevents thrombosis and accelerates fibrinolysis, indicating that PAI-1 inhibitors may be used as effective antithrombotic agents. We previously designed a PAI-1 inhibitor (PAItrap) which is a variant of inactivated urokinase protease domain. In the present study, we fused PAItrap with human serum albumin (HSA) to develop a long-acting PAI-1 inhibitor. Unfortunately, the fusion protein PAItrap-HSA lost some potency compared to PAItrap (33 nM vs 10 nM). Guided by computational method, we carried out further optimisation to enhance inhibitory potency for PAI-1. The new PAItrap, denominated PAItrap(H37R)-HSA, which was the H37R variant of PAItrap fused to HSA, gave a six-fold improvement of IC50 (5 nM) for human active PAI-1 compared to PAItrap-HSA, and showed much longer plasma half-life (200-fold) compared to PAItrap. We further demonstrated that the PAItrap(H37R)-HSA inhibited exogenous or endogenous PAI-1 to promote fibrinolysis in fibrin-clot lysis assay. PAItrap(H37R)-HSA inhibits murine PAI-1 with IC50 value of 12 nM, allowing the inhibitor to be evaluated in murine models. Using an intravital microscopy, we demonstrated that PAItrap(H37R)-HSA blocks thrombus formation and platelet accumulation in vivo in a laser-induced vascular injury mouse model. Additionally, mouse tail bleeding assay showed that PAItrap(H37R)-HSA did not affect the global haemostasis. These results suggest that PAItrap(H37R)-HSA have the potential benefit to prevent thrombosis and accelerates fibrinolysis.
Collapse
|
41
|
Rautio A, Boman K, Gerstein HC, Hernestål-Boman J, Lee SF, Olofsson M, Mellbin LG. The effect of basal insulin glargine on the fibrinolytic system and von Willebrand factor in people with dysglycaemia and high risk for cardiovascular events: Swedish substudy of the Outcome Reduction with an Initial Glargine Intervention trial. Diab Vasc Dis Res 2017; 14:345-352. [PMID: 28403644 DOI: 10.1177/1479164117703034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
INTRODUCTION Fibrinolytic factors, plasminogen activator inhibitor-1, tissue plasminogen activator, tissue plasminogen activator/plasminogen activator-complex and the haemostatic factor von Willebrand factor are known markers of cardiovascular disease. Their plasma levels are adversely affected in patients with dysglycaemia, and glucose normalization with insulin glargine might improve the levels of these factors. METHODS Prespecified Swedish substudy of the Outcome Reduction with an Initial Glargine Intervention trial (ClinicalTrials.gov number, NCT00069784). Tissue plasminogen activator activity, tissue plasminogen activator antigen, plasminogen activator inhibitor-1 antigen, tissue plasminogen activator/plasminogen activator inhibitor-1 complex and von Willebrand factor were analysed at study start, after 2 years and at the end of the study (median follow-up of 6.2 years). RESULTS Of 129 patients (mean age of 64 ± 7 years, females: 19%), 68 (53%) and 61 (47%) were randomized to the insulin glargine and standard care group, respectively. Allocation to insulin glargine did not significantly affect the studied fibrinolytic markers or von Willebrand factor compared to standard care. Likewise, there were no significant differences in plasminogen activator inhibitor-1, tissue plasminogen activator antigen and von Willebrand factor. During the whole study period, the within-group analysis revealed a curvilinear pattern and significant changes for tissue plasminogen activator/plasminogen activator inhibitor-1 complex, tissue plasminogen activator antigen and von Willebrand factor in the insulin glargine but not in the standard care group. CONCLUSION In people with dysglycaemia and other cardiovascular risk factors, basal insulin does not improve the levels of markers of fibrinolysis or von Willebrand factor compared to standard glucose-lowering treatments.
Collapse
Affiliation(s)
- Aslak Rautio
- 1 Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
- 2 Department of Medicine, Sunderby Hospital, Luleå, Sweden
| | - Kurt Boman
- 1 Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
- 3 Research Unit, Skellefteå Hospital, Skellefteå, Sweden
| | - Hertzel C Gerstein
- 4 Population Health Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, ON, Canada
| | - Jenny Hernestål-Boman
- 1 Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
- 3 Research Unit, Skellefteå Hospital, Skellefteå, Sweden
| | - Shun Fu Lee
- 4 Population Health Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, ON, Canada
| | - Mona Olofsson
- 1 Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
- 3 Research Unit, Skellefteå Hospital, Skellefteå, Sweden
| | - Linda Garcia Mellbin
- 5 Unit of Cardiology, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
42
|
Hendrix P, Foreman PM, Harrigan MR, Fisher WS, Vyas NA, Lipsky RH, Lin M, Walters BC, Tubbs RS, Shoja MM, Pittet JF, Mathru M, Griessenauer CJ. Association of Plasminogen Activator Inhibitor 1 (SERPINE1) Polymorphisms and Aneurysmal Subarachnoid Hemorrhage. World Neurosurg 2017; 105:672-677. [PMID: 28599907 DOI: 10.1016/j.wneu.2017.05.175] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 05/30/2017] [Accepted: 05/31/2017] [Indexed: 11/19/2022]
Abstract
BACKGROUND Genetic variations of the serine proteinase inhibitor family E member 1 (SERPINE1) gene, which encodes plasminogen activator inhibitor 1, correlate with serum levels of its product and are associated with thrombophilia and coronary atherosclerosis. Various SERPINE1 ;gene polymorphisms have been identified. However, only the functional 5G/4G polymorphism has been assessed in the context of aneurysmal subarachnoid hemorrhage (aSAH). We assessed associations of 6 SERPINE1 polymorphisms with the clinical sequelae of aSAH. METHODS From 2012 to 2015, patients with aSAH were prospectively enrolled into the CARAS (Cerebral Aneurysm Renin Angiotensin System) study at 2 major academic institutions. Blood samples were used to evaluate 6 common SERPINE1 single nucleotide polymorphisms via 5' exonuclease (Taqman) genotyping assays. RESULTS There was an association of the AA genotype of rs2227631 with the 4G/4G genotype and of the GG genotype of rs7242 with the AA genotype of rs2227684. In multivariable analysis, patients with the AA genotype of rs2227631 and 4G/4G genotype had an increased risk for developing delayed cerebral ischemia. Patients with the GG genotype of rs7242 and AA genotype of rs2227684 had a decreased risk for a poor functional outcome. CONCLUSIONS SERPINE1 gene polymorphisms were associated with delayed cerebral ischemia and functional outcome after aSAH. These associations may arise from alterations of plasminogen activator inhibitor 1 levels.
Collapse
Affiliation(s)
- Philipp Hendrix
- Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg/Saar, Germany.
| | - Paul M Foreman
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Mark R Harrigan
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Winfield S Fisher
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Nilesh A Vyas
- Department of Neurosciences, Inova Health System, Falls Church, Virginia, USA
| | - Robert H Lipsky
- Department of Neurosciences, Inova Health System, Falls Church, Virginia, USA; Department of Molecular Neuroscience, George Mason University, Fairfax, Virginia, USA
| | - Minkuan Lin
- Department of Molecular Neuroscience, George Mason University, Fairfax, Virginia, USA
| | - Beverly C Walters
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, Alabama, USA; Department of Neurosciences, Inova Health System, Falls Church, Virginia, USA; Department of Molecular Neuroscience, George Mason University, Fairfax, Virginia, USA
| | - R Shane Tubbs
- Seattle Science Foundation, Seattle, Washington, USA
| | - Mohammadali M Shoja
- Neuroscience Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jean-Francois Pittet
- Department of Anesthesiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Mali Mathru
- Department of Anesthesiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Christoph J Griessenauer
- Department of Neurosurgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA; Department of Neurosurgery, Geisinger Health System, Danville, Pennsylvania, USA
| |
Collapse
|
43
|
Song C, Burgess S, Eicher JD, O'Donnell CJ, Johnson AD. Causal Effect of Plasminogen Activator Inhibitor Type 1 on Coronary Heart Disease. J Am Heart Assoc 2017; 6:JAHA.116.004918. [PMID: 28550093 PMCID: PMC5669150 DOI: 10.1161/jaha.116.004918] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Background Plasminogen activator inhibitor type 1 (PAI‐1) plays an essential role in the fibrinolysis system and thrombosis. Population studies have reported that blood PAI‐1 levels are associated with increased risk of coronary heart disease (CHD). However, it is unclear whether the association reflects a causal influence of PAI‐1 on CHD risk. Methods and Results To evaluate the association between PAI‐1 and CHD, we applied a 3‐step strategy. First, we investigated the observational association between PAI‐1 and CHD incidence using a systematic review based on a literature search for PAI‐1 and CHD studies. Second, we explored the causal association between PAI‐1 and CHD using a Mendelian randomization approach using summary statistics from large genome‐wide association studies. Finally, we explored the causal effect of PAI‐1 on cardiovascular risk factors including metabolic and subclinical atherosclerosis measures. In the systematic meta‐analysis, the highest quantile of blood PAI‐1 level was associated with higher CHD risk comparing with the lowest quantile (odds ratio=2.17; 95% CI: 1.53, 3.07) in an age‐ and sex‐adjusted model. The effect size was reduced in studies using a multivariable‐adjusted model (odds ratio=1.46; 95% CI: 1.13, 1.88). The Mendelian randomization analyses suggested a causal effect of increased PAI‐1 level on CHD risk (odds ratio=1.22 per unit increase of log‐transformed PAI‐1; 95% CI: 1.01, 1.47). In addition, we also detected a causal effect of PAI‐1 on elevating blood glucose and high‐density lipoprotein cholesterol. Conclusions Our study indicates a causal effect of elevated PAI‐1 level on CHD risk, which may be mediated by glucose dysfunction.
Collapse
Affiliation(s)
- Ci Song
- Framingham Heart Study, Framingham, MA .,The Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD
| | - Stephen Burgess
- Department of Public Health and Primary Care, University of Cambridge, United Kingdom
| | - John D Eicher
- Framingham Heart Study, Framingham, MA.,The Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD
| | - Christopher J O'Donnell
- Framingham Heart Study, Framingham, MA.,Cardiology Section and Center for Population Genomics, Boston Veteran's Administration (VA) Healthcare, Boston, MA
| | - Andrew D Johnson
- Framingham Heart Study, Framingham, MA.,The Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD
| |
Collapse
|
44
|
Double-stranded DNA induces a prothrombotic phenotype in the vascular endothelium. Sci Rep 2017; 7:1112. [PMID: 28442771 PMCID: PMC5430798 DOI: 10.1038/s41598-017-01148-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 03/23/2017] [Indexed: 12/11/2022] Open
Abstract
Double-stranded DNA (dsDNA) constitutes a potent activator of innate immunity, given its ability to bind intracellular pattern recognition receptors during viral infections or sterile tissue damage. While effects of dsDNA in immune cells have been extensively studied, dsDNA signalling and its pathophysiological implications in non-immune cells, such as the vascular endothelium, remain poorly understood. The aim of this study was to characterize prothrombotic effects of dsDNA in vascular endothelial cells. Transfection of cultured human endothelial cells with the synthetic dsDNA poly(dA:dT) induced upregulation of the prothrombotic molecules tissue factor and PAI-1, resulting in accelerated blood clotting in vitro, which was partly dependent on RIG-I signalling. Prothrombotic effects were also observed upon transfection of endothelial cells with hepatitis B virus DNA-containing immunoprecipitates as well human genomic DNA. In addition, dsDNA led to surface expression of von Willebrand factor resulting in increased platelet-endothelium-interactions under flow. Eventually, intrascrotal injection of dsDNA resulted in accelerated thrombus formation upon light/dye-induced endothelial injury in mouse cremaster arterioles and venules in vivo. In conclusion, we show that viral or endogenous dsDNA induces a prothrombotic phenotype in the vascular endothelium. These findings represent a novel link between pathogen- and danger-associated patterns within innate immunity and thrombosis.
Collapse
|
45
|
Kearney K, Tomlinson D, Smith K, Ajjan R. Hypofibrinolysis in diabetes: a therapeutic target for the reduction of cardiovascular risk. Cardiovasc Diabetol 2017; 16:34. [PMID: 28279217 PMCID: PMC5345237 DOI: 10.1186/s12933-017-0515-9] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 02/27/2017] [Indexed: 12/11/2022] Open
Abstract
An enhanced thrombotic environment and premature atherosclerosis are key factors for the increased cardiovascular risk in diabetes. The occlusive vascular thrombus, formed secondary to interactions between platelets and coagulation proteins, is composed of a skeleton of fibrin fibres with cellular elements embedded in this network. Diabetes is characterised by quantitative and qualitative changes in coagulation proteins, which collectively increase resistance to fibrinolysis, consequently augmenting thrombosis risk. Current long-term therapies to prevent arterial occlusion in diabetes are focussed on anti-platelet agents, a strategy that fails to address the contribution of coagulation proteins to the enhanced thrombotic milieu. Moreover, antiplatelet treatment is associated with bleeding complications, particularly with newer agents and more aggressive combination therapies, questioning the safety of this approach. Therefore, to safely control thrombosis risk in diabetes, an alternative approach is required with the fibrin network representing a credible therapeutic target. In the current review, we address diabetes-specific mechanistic pathways responsible for hypofibrinolysis including the role of clot structure, defects in the fibrinolytic system and increased incorporation of anti-fibrinolytic proteins into the clot. Future anti-thrombotic therapeutic options are discussed with special emphasis on the potential advantages of modulating incorporation of the anti-fibrinolytic proteins into fibrin networks. This latter approach carries theoretical advantages, including specificity for diabetes, ability to target a particular protein with a possible favourable risk of bleeding. The development of alternative treatment strategies to better control residual thrombosis risk in diabetes will help to reduce vascular events, which remain the main cause of mortality in this condition.
Collapse
Affiliation(s)
- Katherine Kearney
- Division of Cardiovascular & Diabetes Research, Leeds Institute of Cardiovascular and Metabolic Medicine (LICAMM), University of Leeds, Leeds, LS2 9JT, UK
| | - Darren Tomlinson
- Biomedical Health Research Centre, Astbury Building, University of Leeds, Leeds, LS2 9JT, UK
| | - Kerrie Smith
- Division of Cardiovascular & Diabetes Research, Leeds Institute of Cardiovascular and Metabolic Medicine (LICAMM), University of Leeds, Leeds, LS2 9JT, UK
| | - Ramzi Ajjan
- Division of Cardiovascular & Diabetes Research, Leeds Institute of Cardiovascular and Metabolic Medicine (LICAMM), University of Leeds, Leeds, LS2 9JT, UK.
| |
Collapse
|
46
|
Chang ML, Lin YS, Pao LH, Huang HC, Chiu CT. Link between plasminogen activator inhibitor-1 and cardiovascular risk in chronic hepatitis C after viral clearance. Sci Rep 2017; 7:42503. [PMID: 28211910 PMCID: PMC5304196 DOI: 10.1038/srep42503] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 01/11/2017] [Indexed: 12/15/2022] Open
Abstract
The pathophysiological implications of plasminogen activator inhibitor-1 (PAI-1) in HCV infection remain obscure. This prospective study evaluated 669 HCV patients, of whom 536 had completed a course of anti-HCV therapy and had pre-, peri- and post-therapy measurements of various profiles, including PAI-1 levels. Multivariate analysis demonstrated, before anti-HCV-therapy, platelet count and PAI-1-rs1799889 genotype were associated with PAI-1 levels. Among patients with a sustained virological response (SVR, n = 445), platelet count was associated with PAI-1 level at 24 weeks post-therapy. GEE analysis showed that PAI-1-rs-1799889 and interferon-λ3-rs12979860 genotypes affected PAI-1 levels early and late in therapy, respectively. At 24 weeks post-therapy, higher lipid, brain natriuretic peptide, homocysteine and PAI-1 levels and PAI-1 activity were noted only in SVR patients compared with pre-therapy levels. Within 24 weeks post-therapy, 2.2% of the SVR (mean age: 57.8 yr; 8 smoking males; the 2 females had pre-therapy hypercholesteremia or cardiovascular family history of disease) and 0% of the non-SVR patients experienced a new cardiovascular event. Platelet counts consistently correlated with PAI-1 levels regardless of HCV infection. PAI-1-rs-1799889 and interferon-λ3-rs12979860 genotypes mainly affected PAI-1 levels longitudinally. Within 24 weeks post-anti-HCV therapy, the SVR patients showed increasing PAI-1 levels with accelerating cardiovascular risk, especially the vulnerable cases.
Collapse
Affiliation(s)
- Ming-Ling Chang
- Liver Research Center, Division of Hepatology, Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,Department of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yu-Sheng Lin
- Department of Cardiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,Healthcare center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Li-Heng Pao
- Graduate Institute of Health-Industry Technology, Chang Gung University of Science and Technology, Taoyuan, Taiwan.,Research Center for Industry of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan
| | - Hsin-Chih Huang
- Liver Research Center, Division of Hepatology, Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Cheng-Tang Chiu
- Liver Research Center, Division of Hepatology, Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| |
Collapse
|
47
|
Olatunji LA, Usman TO, Seok YM, Kim IK. Activation of cardiac renin-angiotensin system and plasminogen activator inhibitor-1 gene expressions in oral contraceptive-induced cardiometabolic disorder. Arch Physiol Biochem 2017; 123:1-8. [PMID: 26934364 DOI: 10.3109/13813455.2016.1160935] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
CONTEXT Clinical studies have shown that combined oral contraceptive (COC) use is associated with cardiometabolic disturbances. Elevated renin-angiotensin system (RAS) and plasminogen activator inhibitor-1 (PAI-1) have also been implicated in the development of cardiometabolic events. OBJECTIVE To determine the effect of COC treatment on cardiac RAS and PAI-1 gene expressions, and whether the effect is circulating aldosterone or corticosterone dependent. METHODS Female rats were treated (p.o.) with olive oil (vehicle) or COC (1.0 µg ethinylestradiol and 10.0 µg norgestrel) daily for six weeks. RESULTS COC treatment led to increases in blood pressure, HOMA-IR, Ace1 mRNA, Atr1 mRNA, Pai1 mRNA, cardiac PAI-1, plasma PAI-1, C-reactive protein, uric acid, insulin and corticosterone. COC treatment also led to dyslipidemia, decreased glucose tolerance and plasma 17β-estradiol. CONCLUSION These results demonstrates that hypertension and insulin resistance induced by COC is associated with increased cardiac RAS and PAI-1 gene expression, which is likely to be through corticosterone-dependent but not aldosterone-dependent mechanism.
Collapse
Affiliation(s)
- Lawrence A Olatunji
- a Department of Physiology , Cardiovascular and Molecular Physiology Unit, College of Health Sciences University of Ilorin , P.M.B. 1515 , Ilorin , Nigeria
- b Cardiovascular Research Institute, Kyungpook National University School of Medicine , Daegu , Republic of Korea
| | - Taofeek O Usman
- a Department of Physiology , Cardiovascular and Molecular Physiology Unit, College of Health Sciences University of Ilorin , P.M.B. 1515 , Ilorin , Nigeria
| | - Young-Mi Seok
- b Cardiovascular Research Institute, Kyungpook National University School of Medicine , Daegu , Republic of Korea
- c Korea Promotion Institute for Traditional Medicine Industry , Gyeongsan , Gyeongbuk , Republic of Korea , and
| | - In-Kyeom Kim
- b Cardiovascular Research Institute, Kyungpook National University School of Medicine , Daegu , Republic of Korea
- d Department of Pharmacology , Kyungpook National University School of Medicine , Daegu , Republic of Korea
| |
Collapse
|
48
|
Anti-inflammatory and antithrombotic effects of nicotine exposure in oral contraceptive-induced insulin resistance are glucocorticoid-independent. Pharmacol Rep 2016; 69:512-519. [PMID: 28349880 DOI: 10.1016/j.pharep.2016.12.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 11/30/2016] [Accepted: 12/16/2016] [Indexed: 12/27/2022]
Abstract
BACKGROUND Reports showed that estrogen-progestin oral contraceptive (COC) or tobacco smoking causes increased risk of cardiovascular diseases (CVD) in premenopausal women. Studies also suggest that nicotine, a major tobacco alkaloid, may worsen or improve atherothrombotic CVD. Altered hemorheology, prothrombotic and pro-inflammatory biomarkers, have been implicated in the development of atherothrombotic CVD events. However, the effect of non-smoking nicotine exposure on these biomarkers during COC treatment is not yet established. We therefore sought to determine the effects of nicotine exposure during COC treatment on these biomarkers, and also tested the hypothesis that the nicotine effects would be glucocorticoid-dependent. METHODS Female Sprague-Dawley rats aged 10 weeks were given (po) vehicle, low-dose nicotine (0.1mg/kg) or high-dose nicotine (1.0mg/kg) with or without COC steroids (5.0μg/kg ethinylestradiol and 25.0μg/kg levonorgestrel) daily for 6 weeks. RESULTS COC treatment or nicotine exposure led to increased insulin resistance (IR), hemorheological (blood viscosity, hematocrit and plasma viscosity), prothrombotic (plasminogen activator inhibitor-1), pro-inflammatory (uric acid, C-reactive protein, neutrophil/lymphocyte and platelet/lymphocyte ratios) biomarkers and corticosterone. However, these effects except that on corticosterone were abrogated by nicotine exposure during COC treatment. CONCLUSIONS Our study indicates that nicotine- or COC-induced IR may be mediated via inflammatory/thrombotic pathway. The results imply that nicotine exposure could impact negatively on atherothrombotic biomarkers in COC non-users, whereas the impact in COC users could be positive. The results also suggest that the anti-inflammatory, antithrombotic and blood viscosity-lowering effects of nicotine exposure during COC use is circulating glucocorticoid-independent.
Collapse
|
49
|
Taylor BA, Tofler GH, Carey HMR, Morel-Kopp MC, Philcox S, Carter TR, Elliott MJ, Kull AD, Ward C, Schenck K. Full-mouth Tooth Extraction Lowers Systemic Inflammatory and Thrombotic Markers of Cardiovascular Risk. J Dent Res 2016; 85:74-8. [PMID: 16373685 DOI: 10.1177/154405910608500113] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Prior studies of a link between periodontal and cardiovascular disease have been limited by being predominantly observational. We used a treatment intervention model to study the relationship between periodontitis and systemic inflammatory and thrombotic cardiovascular indicators of risk. We studied 67 adults with advanced periodontitis requiring full-mouth tooth extraction. Blood samples were obtained: (1) at initial presentation, immediately prior to treatment of presenting symptoms; (2) one to two weeks later, before all teeth were removed; and (3) 12 weeks after full-mouth tooth extraction. After full-mouth tooth extraction, there was a significant decrease in C-reactive protein, plasminogen activator inhibitor-1 and fibrinogen, and white cell and platelet counts. This study shows that elimination of advanced periodontitis by full-mouth tooth extraction reduces systemic inflammatory and thrombotic markers of cardiovascular risk. Analysis of the data supports the hypothesis that treatment of periodontal disease may lower cardiovascular risk, and provides a rationale for further randomized studies.
Collapse
Affiliation(s)
- B A Taylor
- Sydney Dental Hospital, 2 Chalmers Street, Surry Hills NSW 2010, Australia.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Pinkney JH, Nagi DK, Yudkin JS. From ‘Syndrome X’ to the Thrifty Phenotype: A Reappraisal of the Insulin Resistance Theory of Atherogenesis. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/1358863x9300400103] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Jonathan H Pinkney
- Department of Medicine, University College London Medical School, London, UK
| | - Dinesh K Nagi
- Department of Medicine, University College London Medical School, London, UK
| | - John S Yudkin
- Department of Medicine, University College London Medical School, London, UK
| |
Collapse
|