1
|
Cimini M, Hansmann UHE, Gonzalez C, Chesney AD, Truongcao MM, Gao E, Wang T, Roy R, Forte E, Mallaredy V, Thej C, Magadum A, Joladarashi D, Benedict C, Koch WJ, Tükel Ç, Kishore R. Podoplanin Positive Cell-derived Extracellular Vesicles Contribute to Cardiac Amyloidosis After Myocardial Infarction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.28.601297. [PMID: 39005419 PMCID: PMC11244852 DOI: 10.1101/2024.06.28.601297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Background Amyloidosis is a major long-term complication of chronic disease; however, whether it represents one of the complications of post-myocardial infarction (MI) is yet to be fully understood. Methods Using wild-type and knocked-out MI mouse models and characterizing in vitro the exosomal communication between bone marrow-derived macrophages and activated mesenchymal stromal cells (MSC) isolated after MI, we investigated the mechanism behind Serum Amyloid A 3 (SAA3) protein overproduction in injured hearts. Results Here, we show that amyloidosis occurs after MI and that amyloid fibers are composed of macrophage-derived SAA3 monomers. SAA3 overproduction in macrophages is triggered by exosomal communication from a subset of activated MSC, which, in response to MI, acquire the expression of a platelet aggregation-inducing type I transmembrane glycoprotein named Podoplanin (PDPN). Cardiac MSC PDPN+ communicate with and activate macrophages through their extracellular vesicles or exosomes. Specifically, MSC PDPN+ derived exosomes (MSC PDPN+ Exosomes) are enriched in SAA3 and exosomal SAA3 protein engages with Toll-like receptor 2 (TRL2) on macrophages, triggering an overproduction and impaired clearance of SAA3 proteins, resulting in aggregation of SAA3 monomers as rigid amyloid deposits in the extracellular space. The onset of amyloid fibers deposition alongside extra-cellular-matrix (ECM) proteins in the ischemic heart exacerbates the rigidity and stiffness of the scar, hindering the contractility of viable myocardium and overall impairing organ function. Using SAA3 and TLR2 deficient mouse models, we show that SAA3 delivered by MSC PDPN+ exosomes promotes post-MI amyloidosis. Inhibition of SAA3 aggregation via administration of a retro-inverso D-peptide, specifically designed to bind SAA3 monomers, prevents the deposition of SAA3 amyloid fibrils, positively modulates the scar formation, and improves heart function post-MI. Conclusion Overall, our findings provide mechanistic insights into post-MI amyloidosis and suggest that SAA3 may be an attractive target for effective scar reversal after ischemic injury and a potential target in multiple diseases characterized by a similar pattern of inflammation and amyloid deposition. NOVELTY AND SIGNIFICANCE What is known? Accumulation of rigid amyloid structures in the left ventricular wall impairs ventricle contractility.After myocardial infarction cardiac Mesenchymal Stromal Cells (MSC) acquire Podoplanin (PDPN) to better interact with immune cells.Amyloid structures can accumulate in the heart after chronic inflammatory conditions. What information does this article contribute? Whether accumulation of cumbersome amyloid structures in the ischemic scar impairs left ventricle contractility, and scar reversal after myocardial infarction (MI) has never been investigated.The pathophysiological relevance of PDPN acquirement by MSC and the functional role of their secreted exosomes in the context of post-MI cardiac remodeling has not been investigated.Amyloid structures are present in the scar after ischemia and are composed of macrophage-derived Serum Amyloid A (SAA) 3 monomers, although mechanisms of SAA3 overproduction is not established. SUMMARY OF NOVELTY AND SIGNIFICANCE Here, we report that amyloidosis, a secondary phenomenon of an already preexisting and prolonged chronic inflammatory condition, occurs after MI and that amyloid structures are composed of macrophage-derived SAA3 monomers. Frequently studied cardiac amyloidosis are caused by aggregation of immunoglobulin light chains, transthyretin, fibrinogen, and apolipoprotein in a healthy heart as a consequence of systemic chronic inflammation leading to congestive heart failure with various types of arrhythmias and tissue stiffness. Although chronic MI is considered a systemic inflammatory condition, studies regarding the possible accumulation of amyloidogenic proteins after MI and the mechanisms involved in that process are yet to be reported. Here, we show that SAA3 overproduction in macrophages is triggered in a Toll-like Receptor 2 (TLR2)-p38MAP Kinase-dependent manner by exosomal communication from a subset of activated MSC, which, in response to MI, express a platelet aggregation-inducing type I transmembrane glycoprotein named Podoplanin. We provide the full mechanism of this phenomenon in murine models and confirm SAA3 amyloidosis in failing human heart samples. Moreover, we developed a retro-inverso D-peptide therapeutic approach, "DRI-R5S," specifically designed to bind SAA3 monomers and prevent post-MI aggregation and deposition of SAA3 amyloid fibrils without interfering with the innate immune response.
Collapse
|
2
|
Ji H, Chen S, Hu Q, He Y, Zhou L, Xie J, Pan H, Tong X, Wu C. Investigating the Correlation between Serum Amyloid A and Infarct-Related Artery Patency Prior to Percutaneous Coronary Intervention in ST-Segment Elevation Myocardial Infarction Patients. Angiology 2024; 75:585-594. [PMID: 37402552 DOI: 10.1177/00033197231183031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/06/2023]
Abstract
Serum amyloid A (SAA) is a cardiovascular risk factor and may serve as a predictor of infarct-related artery (IRA) patency in patients with ST-segment elevation myocardial infarction (STEMI). We measured SAA levels in STEMI patients who underwent percutaneous coronary intervention (PCI) and investigated their association with IRA patency. According to the Thrombolysis in Myocardial Infarction (TIMI) flow grade, 363 STEMI patients undergoing PCI in our hospital were divided into an occlusion group (TIMI 0-2) and a patency group (TIMI 3). The SAA level before PCI was significantly higher in STEMI patients with IRA occluded than in those with patent ones. At a cutoff value of 36.9 mg/L, SAA had a sensitivity of 63.0% and a specificity of 90.6% (area under the ROC curve [AUC] = .833, 95% CI: .793-.873, P < .001). Multivariate logistic regression analysis showed that SAA was an independent predictor of IRA patency in STEMI patients before PCI (odds ratio [OR] = 1.041, 95% CI: 1.020-1.062, P < .001). SAA can be used as a potential predictor of IRA patency in STEMI patients before PCI.
Collapse
Affiliation(s)
- Hao Ji
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Senjiang Chen
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qingqing Hu
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ying He
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Liang Zhou
- Department of Cardiology, Hangzhou First People's Hospital Affiliated to Zhejiang University, School of Medicine, Hangzhou, China
| | - Jianchang Xie
- Department of Cardiology, Hangzhou First People's Hospital Affiliated to Zhejiang University, School of Medicine, Hangzhou, China
| | - Hao Pan
- Department of Cardiology, Hangzhou First People's Hospital Affiliated to Zhejiang University, School of Medicine, Hangzhou, China
| | - Xiaoshan Tong
- Catheter Room, Hangzhou First People's Hospital Affiliated to Zhejiang University, School of Medicine, Hangzhou, China
| | - Chenghao Wu
- Department of Critical Care Medicine, Hangzhou First People's Hospital Affiliated to Zhejiang University, School of Medicine, Hangzhou, China
| |
Collapse
|
3
|
Huang S, Jiang Y, Li J, Mao L, Qiu Z, Zhang S, Jiang Y, Liu Y, Liu W, Xiong Z, Zhang W, Liu X, Zhang Y, Bai X, Guo B. Osteocytes/Osteoblasts Produce SAA3 to Regulate Hepatic Metabolism of Cholesterol. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307818. [PMID: 38613835 PMCID: PMC11199997 DOI: 10.1002/advs.202307818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 03/19/2024] [Indexed: 04/15/2024]
Abstract
Hypercholesterolaemia is a systemic metabolic disease, but the role of organs other than liver in cholesterol metabolism is unappreciated. The phenotypic characterization of the Tsc1Dmp1 mice reveal that genetic depletion of tuberous sclerosis complex 1 (TSC1) in osteocytes/osteoblasts (Dmp1-Cre) triggers progressive increase in serum cholesterol level. The resulting cholesterol metabolic dysregulation is shown to be associated with upregulation and elevation of serum amyloid A3 (SAA3), a lipid metabolism related factor, in the bone and serum respectively. SAA3, elicited from the bone, bound to toll-like receptor 4 (TLR4) on hepatocytes to phosphorylate c-Jun, and caused impeded conversion of cholesterol to bile acids via suppression on cholesterol 7 α-hydroxylase (Cyp7a1) expression. Ablation of Saa3 in Tsc1Dmp1 mice prevented the CYP7A1 reduction in liver and cholesterol elevation in serum. These results expand the understanding of bone function and hepatic regulation of cholesterol metabolism and uncover a potential therapeutic use of pharmacological modulation of SAA3 in hypercholesterolaemia.
Collapse
Affiliation(s)
- Shijiang Huang
- State Key Laboratory of Organ Failure ResearchDepartment of Cell BiologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Yuanjun Jiang
- State Key Laboratory of Organ Failure ResearchDepartment of Cell BiologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Jing Li
- Department of Obstetrics and GynecologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Linlin Mao
- State Key Laboratory of Organ Failure ResearchDepartment of Cell BiologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Zeyou Qiu
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
- Equipment Material DepartmentWest China Xiamen Hospital of Sichuan UniversityXiamenFujian361000China
| | - Sheng Zhang
- State Key Laboratory of Organ Failure ResearchDepartment of Cell BiologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Yuhui Jiang
- State Key Laboratory of Organ Failure ResearchDepartment of Cell BiologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Yong Liu
- State Key Laboratory of Organ Failure ResearchDepartment of Cell BiologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Wen Liu
- State Key Laboratory of Organ Failure ResearchDepartment of Cell BiologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Zhi Xiong
- State Key Laboratory of Organ Failure ResearchDepartment of Cell BiologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Wuju Zhang
- State Key Laboratory of Organ Failure ResearchDepartment of Cell BiologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
- Central LaboratoryThe Fifth Affiliated HospitalSouthern Medical UniversityGuangzhouGuangdong510900China
| | - Xiaolin Liu
- State Key Laboratory of Organ Failure ResearchDepartment of Cell BiologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Yue Zhang
- State Key Laboratory of Organ Failure ResearchDepartment of Cell BiologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Xiaochun Bai
- State Key Laboratory of Organ Failure ResearchDepartment of Cell BiologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
- Guangdong Provincial Key Laboratory of Bone and Joint Degenerative DiseasesThe Third Affiliated Hospital of Southern Medical UniversityGuangzhouGuangdong510630China
| | - Bin Guo
- State Key Laboratory of Organ Failure ResearchDepartment of Cell BiologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
- The Tenth Affiliated HospitalSouthern Medical UniversityDongguanGuangdong523018China
| |
Collapse
|
4
|
Grymyr LMD, Mellgren G, McCann A, Gerdts E, Meyer K, Nadirpour S, Fernø J, Nedrebø BG, Cramariuc D. Preoperative risk factors associated with left ventricular dysfunction after bariatric surgery. Sci Rep 2024; 14:2173. [PMID: 38273044 PMCID: PMC10810803 DOI: 10.1038/s41598-024-52623-1] [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: 08/24/2023] [Accepted: 01/22/2024] [Indexed: 01/27/2024] Open
Abstract
A large proportion of patients with severe obesity remain with left ventricular (LV) dysfunction after bariatric surgery. We assessed whether preoperative evaluation by echocardiography and inflammatory proteins can identify this high-risk group. In the Bariatric Surgery on the West Coast of Norway study, 75 patients (44 ± 10 years, body mass index [BMI] 41.5 ± 4.7 kg/m2) were prospectively evaluated by echocardiography and inflammatory proteins (high-sensitivity C-reactive protein [hsCRP], serum amyloid A [SAA] and calprotectin) before and one year after Roux-en-Y gastric bypass surgery. LV mechanics was assessed by the midwall shortening (MWS) and global longitudinal strain (GLS). Bariatric surgery improved BMI and GLS, and lowered hsCRP, calprotectin and SAA (p < 0.05). MWS remained unchanged and 35% of patients had impaired MWS at 1-year follow-up. A preoperative risk index including sex, hypertension, ejection fraction (EF) and high hsCRP (index 1) or SAA (index 2) predicted low 1-year MWS with 81% sensitivity/71% specificity (index 1), and 77% sensitivity/77% specificity (index 2) in ROC analyses (AUC 0.80 and 0.79, p < 0.001). Among individuals with severe obesity, women and patients with hypertension, increased serum levels of inflammatory proteins and reduced EF are at high risk of impaired LV midwall mechanics 1 year after bariatric surgery.ClinicalTrials.gov identifier NCT01533142 February 15, 2012.
Collapse
Affiliation(s)
- Lisa M D Grymyr
- Department of Heart Disease, Haukeland University Hospital, Jonas Liesvei 65, 5021, Bergen, Norway
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Gunnar Mellgren
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
| | | | - Eva Gerdts
- Center for Research on Cardiac Disease in Women, Department of Clinical Science, University of Bergen, Bergen, Norway
| | | | - Saied Nadirpour
- Department of Medicine, Haugesund Hospital, Haugesund, Norway
| | - Johan Fernø
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
| | - Bjørn G Nedrebø
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medicine, Haugesund Hospital, Haugesund, Norway
| | - Dana Cramariuc
- Department of Heart Disease, Haukeland University Hospital, Jonas Liesvei 65, 5021, Bergen, Norway.
- Department of Clinical Science, University of Bergen, Bergen, Norway.
| |
Collapse
|
5
|
Plášek J, Dodulík J, Lazárová M, Stejskal D, Švagera Z, Chobolová N, Šulc P, Evin L, Purová D, Václavík J. Secretoneurin levels are higher in dilated cardiomyopathy than in ischaemic cardiomyopathy: preliminary results. Front Cardiovasc Med 2024; 10:1297900. [PMID: 38259301 PMCID: PMC10800804 DOI: 10.3389/fcvm.2023.1297900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 12/14/2023] [Indexed: 01/24/2024] Open
Abstract
Background Secretoneurin (SN) is a neuropeptide with potential utility as a biomarker of cardiovascular episodes. The main effect of SN is mediated through its inhibition of calmodulin-dependent kinase II (CaMKII), which influences calcium handling. We aimed to associate the levels of SN in plasma with different causes of heart failure. Methods We prospectively enrolled consecutive patients with ischaemic (ICM) and dilated (DCM) cardiomyopathy from the outpatient heart failure clinic and healthy individuals. SN was analysed from venous blood by use of the ELISA method. SN plasma levels were compared in DCM, ICM and healthy individuals with non-parametric tests. Results A total of 53 patients (81.1% male, 18.9% female; mean age 67.9 ± 12.6 years) and 34 healthy individuals (38% male, 62% female) were included in the analysis. Plasma SN levels were significantly higher in the dilated cardiomyopathy (38.8 ± 27 pmol/L) as compared with the ischaemic cardiomyopathy (19.7 ± 22.6 pmol/L) group (P = 0.006). There was no significant difference between females vs. males (27.1 ± 23 vs. 25.5 ± 26.2 pmol/L, P = NS). Plasma SN levels allowed DCM and ICM to be differentiated with 88% sensitivity and 61% specificity (P = 0.007), the cut of value is 13.3 pmol/L. Plasma SN levels differed significantly between healthy volunteers and both ICM (P < 0.0001) and DCM (P = 0.049). Plasma SN levels did not differ according to age and were not associated with comorbidities, left ventricular ejection fraction, heart failure medication, troponin, creatinine, or natriuretic peptide plasma levels. Conclusion Plasma secretoneurin levels differed significantly in DCM vs. ICM, being higher in the former. Based on plasma SN levels, discrimination between DCM and ICM might be possible. Healthy individuals produce higher SN plasma levels than stable HFrEF patients.
Collapse
Affiliation(s)
- Jiří Plášek
- Department of Internal Medicine and Cardiology, University Hospital Ostrava, Ostrava, Czechia
- Research Center for Internal and Cardiovascular Diseases Faculty of Medicine, University of Ostrava, Ostrava, Czechia
| | - Jozef Dodulík
- Department of Internal Medicine and Cardiology, University Hospital Ostrava, Ostrava, Czechia
| | - Marie Lazárová
- Department of Internal Medicine and Cardiology, University Hospital Ostrava, Ostrava, Czechia
| | - David Stejskal
- Institute of Laboratory Medicine, University Hospital Ostrava, Ostrava, Czechia
- Institute of Laboratory Medicine, University of Ostrava, Ostrava, Czechia
| | - Zdeněk Švagera
- Institute of Laboratory Medicine, University Hospital Ostrava, Ostrava, Czechia
- Institute of Laboratory Medicine, University of Ostrava, Ostrava, Czechia
| | - Nela Chobolová
- Institute of Laboratory Medicine, University Hospital Ostrava, Ostrava, Czechia
| | - Patrik Šulc
- Department of Internal Medicine and Cardiology, University Hospital Ostrava, Ostrava, Czechia
| | - Lukáš Evin
- Department of Internal Medicine and Cardiology, University Hospital Ostrava, Ostrava, Czechia
- Research Center for Internal and Cardiovascular Diseases Faculty of Medicine, University of Ostrava, Ostrava, Czechia
| | - Dana Purová
- Social Health Institute, Palacky University Olomouc, Olomouc, Czechia
| | - Jan Václavík
- Department of Internal Medicine and Cardiology, University Hospital Ostrava, Ostrava, Czechia
- Research Center for Internal and Cardiovascular Diseases Faculty of Medicine, University of Ostrava, Ostrava, Czechia
| |
Collapse
|
6
|
Abstract
Atherosclerosis is the main cause of arterial thrombosis, causing acute occlusive cardiovascular syndromes. Numerous risk prediction models have been developed, which mathematically combine multiple predictors, to estimate the risk of developing cardiovascular events. Current risk models typically do not include information from biomarkers that can potentially improve these existing prediction models especially if they are pathophysiologically relevant. Numerous cardiovascular disease biomarkers have been investigated that have focused on known pathophysiological pathways including those related to cardiac stress, inflammation, matrix remodelling, and endothelial dysfunction. Imaging biomarkers have also been studied that have yielded promising results with a potential higher degree of clinical applicability in detection of atherosclerosis and cardiovascular event prediction. To further improve therapy decision-making and guidance, there is continuing intense research on emerging biologically relevant biomarkers. As the pathogenesis of cardiovascular disease is multifactorial, improvements in discrimination and reclassification in risk prediction models will likely involve multiple biomarkers. This article will provide an overview of the literature on potential blood-based and imaging biomarkers of atherosclerosis studied so far, as well as potential future directions.
Collapse
Affiliation(s)
- Kashan Ali
- From the Division of Molecular & Clinical Medicine, School of Medicine, Ninewells Hospital & Medical School, University of Dundee, Dundee, UK
| | - Chim C Lang
- From the Division of Molecular & Clinical Medicine, School of Medicine, Ninewells Hospital & Medical School, University of Dundee, Dundee, UK
| | - Jeffrey T J Huang
- Biomarker and Drug Analysis Core Facility, Medical Research Institute, Ninewells Hospital & Medical School, University of Dundee, Dundee, UK
| | - Anna-Maria Choy
- From the Division of Molecular & Clinical Medicine, School of Medicine, Ninewells Hospital & Medical School, University of Dundee, Dundee, UK
| |
Collapse
|
7
|
Chen L, Zhao Y, Wang Z, Wang Y, Bo X, Jiang X, Hao C, Ju C, Qu Y, Dong H. Very high HDL-C (high-density lipoprotein cholesterol) is associated with increased cardiovascular risk in patients with NSTEMI (non-ST-segment elevation myocardial infarction) undergoing PCI (percutaneous coronary intervention). BMC Cardiovasc Disord 2023; 23:357. [PMID: 37461001 DOI: 10.1186/s12872-023-03383-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 07/06/2023] [Indexed: 07/20/2023] Open
Abstract
BACKGROUND Studies in populations with or without cardiovascular disease have shown that very high HDL-C levels are associated with an increased risk of cardiovascular events. However, the exact relationship between HDL-C levels and long-term prognosis remains unknown in patients with myocardial infarction (MI) undergoing percutaneous coronary intervention (PCI). METHODS This was a post hoc secondary analysis of long-term follow-up results in patients undergoing PCI open-label, observational cohort study. Patients with MI who had undergone PCI were enrolled. Restricted cubic spline (RCS) analysis and logistic regression analysis were performed to assess the relationship between HDL-C levels and the risk of cardiovascular events. RESULTS A total of 1934 patients with MI undergoing PCI were enrolled in our analysis and our population was divided in 3 groups according to the HDL-C plasma levels: HDL-C < 40 mg/dL (low HDL-C); HDL-C between 40 and 80 mg/ dL (medium HDL-C); and HDL-C > 80 mg/dL (high HDL-C). RCS analysis showed a nonlinear U-shaped association between HDL-C levels and major adverse cardiac and cerebrovascular events (MACCE) in patients with NSTEMI with adjusted variables. After adjusting for potential confounders, the follow-up analysis indicated that high risk group had elevated occurrence of MACCE than low risk group (HDL-C 35 and 55 mg/dL) (OR:1.645, P = 0.006). CONCLUSIONS Our analysis demonstrated that there is a U-shaped association between HDL-C and MACCE in patients with NSTEMI undergoing PCI.
Collapse
Affiliation(s)
- Lijuan Chen
- Department of Cardiology, Zhongda Hospital, Southeast University, 210009, Nanjing, China
- Department of Cardiology, Nanjing Lishui People's Hospital, Zhongda Hospital Lishui Branch, 211200, Nanjing, China
| | - Yuanyuan Zhao
- Department of Cardiology, Zhongda Hospital, Southeast University, 210009, Nanjing, China
- School of Medicine, Southeast University, 210009, Nanjing, China
| | - Zheng Wang
- Department of Cardiology, Zhongda Hospital, Southeast University, 210009, Nanjing, China
- School of Medicine, Southeast University, 210009, Nanjing, China
| | - Yifei Wang
- Department of Cardiology, Zhongda Hospital, Southeast University, 210009, Nanjing, China
- School of Medicine, Southeast University, 210009, Nanjing, China
| | - Xiangwei Bo
- Department of Cardiology, Zhongda Hospital, Southeast University, 210009, Nanjing, China
- School of Medicine, Southeast University, 210009, Nanjing, China
| | - Xiaoxi Jiang
- Department of Cardiology, Zhongda Hospital, Southeast University, 210009, Nanjing, China
- Department of Cardiology, Nanjing Lishui People's Hospital, Zhongda Hospital Lishui Branch, 211200, Nanjing, China
| | - Chunshu Hao
- Department of Cardiology, Zhongda Hospital, Southeast University, 210009, Nanjing, China
- School of Medicine, Southeast University, 210009, Nanjing, China
| | - Chengwei Ju
- Department of Cardiology, Zhongda Hospital, Southeast University, 210009, Nanjing, China
- Department of Cardiology, Nanjing Lishui People's Hospital, Zhongda Hospital Lishui Branch, 211200, Nanjing, China
| | - Yangyang Qu
- Department of Cardiology, Zhongda Hospital, Southeast University, 210009, Nanjing, China
- School of Medicine, Southeast University, 210009, Nanjing, China
| | - Hongjian Dong
- Department of Cardiology, Zhongda Hospital, Southeast University, 210009, Nanjing, China.
- School of Medicine, Southeast University, 210009, Nanjing, China.
| |
Collapse
|
8
|
Yazdani AN, Pletsch M, Chorbajian A, Zitser D, Rai V, Agrawal DK. Biomarkers to monitor the prognosis, disease severity, and treatment efficacy in coronary artery disease. Expert Rev Cardiovasc Ther 2023; 21:675-692. [PMID: 37772751 PMCID: PMC10615890 DOI: 10.1080/14779072.2023.2264779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 09/26/2023] [Indexed: 09/30/2023]
Abstract
INTRODUCTION Coronary Artery Disease (CAD) is a prevalent condition characterized by the presence of atherosclerotic plaques in the coronary arteries of the heart. The global burden of CAD has increased significantly over the years, resulting in millions of deaths annually and making it the leading health-care expenditure and cause of mortality in developed countries. The lack of cost-effective strategies for monitoring the prognosis of CAD warrants a pressing need for accurate and efficient markers to assess disease severity and progression for both reducing health-care costs and improving patient outcomes. AREA COVERED To effectively monitor CAD, prognostic biomarkers and imaging techniques play a vital role in risk-stratified patients during acute treatment and over time. However, with over 1,000 potential markers of interest, it is crucial to identify the key markers with substantial utility in monitoring CAD progression and evaluating therapeutic interventions. This review focuses on identifying and highlighting the most relevant markers for monitoring CAD prognosis and disease severity. We searched for relevant literature using PubMed and Google Scholar. EXPERT OPINION By utilizing the markers discussed, health-care providers can improve patient care, optimize treatment plans, and ultimately reduce health-care costs associated with CAD management.
Collapse
Affiliation(s)
- Armand N. Yazdani
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766
| | - Michaela Pletsch
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766
| | - Abraham Chorbajian
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766
| | - David Zitser
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766
| | - Vikrant Rai
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766
| | - Devendra K. Agrawal
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766
| |
Collapse
|
9
|
den Hartigh LJ, May KS, Zhang XS, Chait A, Blaser MJ. Serum amyloid A and metabolic disease: evidence for a critical role in chronic inflammatory conditions. Front Cardiovasc Med 2023; 10:1197432. [PMID: 37396595 PMCID: PMC10311072 DOI: 10.3389/fcvm.2023.1197432] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 05/15/2023] [Indexed: 07/04/2023] Open
Abstract
Serum amyloid A (SAA) subtypes 1-3 are well-described acute phase reactants that are elevated in acute inflammatory conditions such as infection, tissue injury, and trauma, while SAA4 is constitutively expressed. SAA subtypes also have been implicated as playing roles in chronic metabolic diseases including obesity, diabetes, and cardiovascular disease, and possibly in autoimmune diseases such as systemic lupus erythematosis, rheumatoid arthritis, and inflammatory bowel disease. Distinctions between the expression kinetics of SAA in acute inflammatory responses and chronic disease states suggest the potential for differentiating SAA functions. Although circulating SAA levels can rise up to 1,000-fold during an acute inflammatory event, elevations are more modest (∼5-fold) in chronic metabolic conditions. The majority of acute-phase SAA derives from the liver, while in chronic inflammatory conditions SAA also derives from adipose tissue, the intestine, and elsewhere. In this review, roles for SAA subtypes in chronic metabolic disease states are contrasted to current knowledge about acute phase SAA. Investigations show distinct differences between SAA expression and function in human and animal models of metabolic disease, as well as sexual dimorphism of SAA subtype responses.
Collapse
Affiliation(s)
- Laura J. den Hartigh
- Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition, University of Washington, Seattle, WA, United States
- Diabetes Institute, University of Washington, Seattle, WA, United States
| | - Karolline S. May
- Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition, University of Washington, Seattle, WA, United States
- Diabetes Institute, University of Washington, Seattle, WA, United States
| | - Xue-Song Zhang
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, United States
| | - Alan Chait
- Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition, University of Washington, Seattle, WA, United States
- Diabetes Institute, University of Washington, Seattle, WA, United States
| | - Martin J. Blaser
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, United States
| |
Collapse
|
10
|
Huang H, Ye F, Huang Y, Ye G, Zhu J, Chi X, Zhang G. Coronary CT angiography and serum biomarkers are potential biomarkers for predicting MACE at three-months and one-year follow-up. Int J Cardiovasc Imaging 2022; 38:2763-2770. [PMID: 36445669 PMCID: PMC9708748 DOI: 10.1007/s10554-022-02646-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 05/07/2022] [Indexed: 11/24/2022]
Abstract
AIMS To assess the prognostic value of coronary computed tomography angiography (CTA) and serum biomarkers for the prediction of major adverse cardiac events (MACE) at three-month and one-year follow-ups. METHODS AND RESULTS A total of 720 patients with acute chest pain and normal electrocardiography (ECG) were included in the prospective cohort study. These patients received both coronary CTA screening and serum biomarkers testing, followed by three-month and one-year follow-ups for the occurrence of major adverse cardiac events (MACE). The primary outcome was the occurrence of MACE, which is defined as acute coronary syndrome (ACS), nonfatal MI, and all-cause mortality. The MACE rate was 17.8% (128 cases) and 25.2% (182 cases) at three-months and one-year follow-up. ApoB/apoA1(OR = 7.45, P < 0.001) and the number of atherosclerotic vessels (OR = 2.86, P < 0.001) were independent predictors for MACE at the three-month follow-up, so were apoB/apoA1 (OR = 5.23, P = 0.003), Serum amyloid protein A (SAA, OR = 1.04, P < 0.001) and the number of atherosclerotic vessels (OR = 2.54, P < 0.001) at the one-year follow-up. While apoB/apoA1 suggested its sensitivities of 84% for predicting MACE at three-month follow-ups, the number of atherosclerotic vessels had 81% specificity at one-year follow-up. CONCLUSIONS Among patients with acute chest pain and normal ECG, apoB/apoA1, SAA and the number of atherosclerotic vessels are the most powerful predictors of MACE at three-month and one-year follow-ups.
Collapse
Affiliation(s)
- Honglei Huang
- Department of Radiology, The Affiliated Nanping First Hospital of Fujian Medical University, Nanping, China
| | - Fei Ye
- Department of Radiology, The Affiliated Nanping First Hospital of Fujian Medical University, Nanping, China
| | - Yuanmao Huang
- Department of Laboratory, The Affiliated Nanping First Hospital of Fujian Medical University, Nanping, China
| | - Guiyun Ye
- Department of Laboratory, The Affiliated Nanping First Hospital of Fujian Medical University, Nanping, China.
| | - Jiarui Zhu
- Department of Laboratory, The Affiliated Nanping First Hospital of Fujian Medical University, Nanping, China
| | - Xidi Chi
- Department of Laboratory, The Affiliated Nanping First Hospital of Fujian Medical University, Nanping, China
| | - Gaofeng Zhang
- Department of Radiology, Affiliated Hospital of Zunyi Medical University, Zunyi City, China.
| |
Collapse
|
11
|
Vyletelová V, Nováková M, Pašková Ľ. Alterations of HDL's to piHDL's Proteome in Patients with Chronic Inflammatory Diseases, and HDL-Targeted Therapies. Pharmaceuticals (Basel) 2022; 15:1278. [PMID: 36297390 PMCID: PMC9611871 DOI: 10.3390/ph15101278] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/03/2022] [Accepted: 10/14/2022] [Indexed: 09/10/2023] Open
Abstract
Chronic inflammatory diseases, such as rheumatoid arthritis, steatohepatitis, periodontitis, chronic kidney disease, and others are associated with an increased risk of atherosclerotic cardiovascular disease, which persists even after accounting for traditional cardiac risk factors. The common factor linking these diseases to accelerated atherosclerosis is chronic systemic low-grade inflammation triggering changes in lipoprotein structure and metabolism. HDL, an independent marker of cardiovascular risk, is a lipoprotein particle with numerous important anti-atherogenic properties. Besides the essential role in reverse cholesterol transport, HDL possesses antioxidative, anti-inflammatory, antiapoptotic, and antithrombotic properties. Inflammation and inflammation-associated pathologies can cause modifications in HDL's proteome and lipidome, transforming HDL from atheroprotective into a pro-atherosclerotic lipoprotein. Therefore, a simple increase in HDL concentration in patients with inflammatory diseases has not led to the desired anti-atherogenic outcome. In this review, the functions of individual protein components of HDL, rendering them either anti-inflammatory or pro-inflammatory are described in detail. Alterations of HDL proteome (such as replacing atheroprotective proteins by pro-inflammatory proteins, or posttranslational modifications) in patients with chronic inflammatory diseases and their impact on cardiovascular health are discussed. Finally, molecular, and clinical aspects of HDL-targeted therapies, including those used in therapeutical practice, drugs in clinical trials, and experimental drugs are comprehensively summarised.
Collapse
Affiliation(s)
| | | | - Ľudmila Pašková
- Department of Cell and Molecular Biology of Drugs, Faculty of Pharmacy, Comenius University, 83232 Bratislava, Slovakia
| |
Collapse
|
12
|
Shridas P, Ji A, Trumbauer AC, Noffsinger VP, Leung SW, Dugan AJ, Thatcher SE, Cassis LA, de Beer FC, Webb NR, Tannock LR. Adipocyte-Derived Serum Amyloid A Promotes Angiotensin II-Induced Abdominal Aortic Aneurysms in Obese C57BL/6J Mice. Arterioscler Thromb Vasc Biol 2022; 42:632-643. [PMID: 35344382 PMCID: PMC9050948 DOI: 10.1161/atvbaha.121.317225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 03/09/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND Obesity increases the risk for human abdominal aortic aneurysms (AAAs) and enhances Ang II (angiotensin II)-induced AAA formation in C57BL/6J mice. Obesity is also associated with increases in perivascular fat that expresses proinflammatory markers including SAA (serum amyloid A). We previously reported that deficiency of SAA significantly reduces Ang II-induced inflammation and AAA in hyperlipidemic apoE-deficient mice. In this study. we investigated whether adipose tissue-derived SAA plays a role in Ang II-induced AAA in obese C57BL/6J mice. METHODS The development of AAA was compared between male C57BL/6J mice (wild type), C57BL/6J mice lacking SAA1.1, SAA2.1, and SAA3 (TKO); and TKO mice harboring a doxycycline-inducible, adipocyte-specific SAA1.1 transgene (TKO-Tgfat; SAA expressed only in fat). All mice were fed an obesogenic diet and doxycycline to induce SAA transgene expression and infused with Ang II to induce AAA. RESULTS In response to Ang II infusion, SAA expression was significantly increased in perivascular fat of obese C57BL/6J mice. Maximal luminal diameters of the abdominal aorta were determined by ultrasound before and after Ang II infusion, which indicated a significant increase in aortic luminal diameters in wild type and TKO-TGfat mice but not in TKO mice. Adipocyte-specific SAA expression was associated with MMP (matrix metalloproteinase) activity and macrophage infiltration in abdominal aortas of Ang II-infused obese mice. CONCLUSIONS We demonstrate for the first time that SAA deficiency protects obese C57BL/6J mice from Ang II-induced AAA. SAA expression only in adipocytes is sufficient to cause AAA in obese mice infused with Ang II.
Collapse
Affiliation(s)
- Preetha Shridas
- Departments of Internal Medicine (P.S., A.J., V.P.N., S.W.L., F.C.d.B., L.R.T.), University of Kentucky, Lexington
- Saha Cardiovascular Research Center (P.S., A.C.T., S.W.L., F.C.d.B., N.R.W., L.R.T.), University of Kentucky, Lexington
- Barnstable Brown Diabetes Center (P.S., F.C.d.B., N.R.W., L.R.T.), University of Kentucky, Lexington
| | - Ailing Ji
- Departments of Internal Medicine (P.S., A.J., V.P.N., S.W.L., F.C.d.B., L.R.T.), University of Kentucky, Lexington
| | - Andrea C Trumbauer
- Saha Cardiovascular Research Center (P.S., A.C.T., S.W.L., F.C.d.B., N.R.W., L.R.T.), University of Kentucky, Lexington
| | - Victoria P Noffsinger
- Departments of Internal Medicine (P.S., A.J., V.P.N., S.W.L., F.C.d.B., L.R.T.), University of Kentucky, Lexington
| | - Steve W Leung
- Departments of Internal Medicine (P.S., A.J., V.P.N., S.W.L., F.C.d.B., L.R.T.), University of Kentucky, Lexington
- Saha Cardiovascular Research Center (P.S., A.C.T., S.W.L., F.C.d.B., N.R.W., L.R.T.), University of Kentucky, Lexington
| | - Adam J Dugan
- Biostatistics (A.J.D.), University of Kentucky, Lexington
| | - Sean E Thatcher
- Department of Pharmacology, Temple University, Philadelphia, PA (S.E.T.)
| | - Lisa A Cassis
- Pharmacology and Nutritional Sciences (L.A.C., N.R.W.), University of Kentucky, Lexington
| | - Frederick C de Beer
- Departments of Internal Medicine (P.S., A.J., V.P.N., S.W.L., F.C.d.B., L.R.T.), University of Kentucky, Lexington
- Saha Cardiovascular Research Center (P.S., A.C.T., S.W.L., F.C.d.B., N.R.W., L.R.T.), University of Kentucky, Lexington
- Barnstable Brown Diabetes Center (P.S., F.C.d.B., N.R.W., L.R.T.), University of Kentucky, Lexington
| | - Nancy R Webb
- Pharmacology and Nutritional Sciences (L.A.C., N.R.W.), University of Kentucky, Lexington
- Saha Cardiovascular Research Center (P.S., A.C.T., S.W.L., F.C.d.B., N.R.W., L.R.T.), University of Kentucky, Lexington
- Barnstable Brown Diabetes Center (P.S., F.C.d.B., N.R.W., L.R.T.), University of Kentucky, Lexington
| | - Lisa R Tannock
- Departments of Internal Medicine (P.S., A.J., V.P.N., S.W.L., F.C.d.B., L.R.T.), University of Kentucky, Lexington
- Saha Cardiovascular Research Center (P.S., A.C.T., S.W.L., F.C.d.B., N.R.W., L.R.T.), University of Kentucky, Lexington
- Barnstable Brown Diabetes Center (P.S., F.C.d.B., N.R.W., L.R.T.), University of Kentucky, Lexington
- Department of Veterans Affairs, Lexington, KY (L.R.T.)
| |
Collapse
|
13
|
Thioredoxin Domain Containing 5 Suppression Elicits Serum Amyloid A-Containing High-Density Lipoproteins. Biomedicines 2022; 10:biomedicines10030709. [PMID: 35327511 PMCID: PMC8945230 DOI: 10.3390/biomedicines10030709] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/15/2022] [Accepted: 03/16/2022] [Indexed: 11/29/2022] Open
Abstract
Thioredoxin domain containing 5 (TXNDC5) is a protein disulfide isomerase involved in several diseases related to oxidative stress, energy metabolism and cellular inflammation. In a previous manuscript, a negative association between fatty liver development and hepatic Txndc5 expression was observed. To study the role of TXNDC5 in the liver, we generated Txndc5-deficient mice. The absence of the protein caused an increased metabolic need to gain weight along with a bigger and fatter liver. RNAseq was performed to elucidate the putative mechanisms, showing a substantial liver overexpression of serum amyloid genes (Saa1, Saa2) with no changes in hepatic protein, but discrete plasma augmentation by the gene inactivation. Higher levels of malonyldialdehyde, apolipoprotein A1 and platelet activating factor-aryl esterase activity were also found in serum from Txndc5-deficient mice. However, no difference in the distribution of high-density lipoproteins (HDL)-mayor components and SAA was found between groups, and even the reactive oxygen species decreased in HDL coming from Txndc5-deficient mice. These results confirm the relation of this gene with hepatic steatosis and with a fasting metabolic derive remedying an acute phase response. Likewise, they pose a new role in modulating the nature of HDL particles, and SAA-containing HDL particles are not particularly oxidized.
Collapse
|
14
|
Zhao Y, Godier-Furnemont A, Bax NA, Bouten CV, Brown LM, Fine B, Vunjak-Novakovic G. Changes in extracellular matrix in failing human non-ischemic and ischemic hearts with mechanical unloading. J Mol Cell Cardiol 2022; 166:137-151. [PMID: 35219725 PMCID: PMC9035113 DOI: 10.1016/j.yjmcc.2022.02.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 01/28/2022] [Accepted: 02/11/2022] [Indexed: 10/19/2022]
Abstract
Ischemic and non-ischemic cardiomyopathies have distinct etiologies and underlying disease mechanisms, which require in-depth investigation for improved therapeutic interventions. The goal of this study was to use clinically obtained myocardium from healthy and heart failure patients, and characterize the changes in extracellular matrix (ECM) in ischemic and non-ischemic failing hearts, with and without mechanical unloading. Using tissue engineering methodologies, we also investigated how diseased human ECM, in the absence of systemic factors, can influence cardiomyocyte function. Heart tissues from heart failure patients with ischemic and non-ischemic cardiomyopathy were compared to explore differential disease phenotypes and reverse remodeling potential of left ventricular assisted device (LVAD) support at transcriptomic, proteomic and structural levels. The collected data demonstrated that the differential ECM compositions recapitulated the disease microenvironment and induced cardiomyocytes to undergo disease-like functional alterations. In addition, our study also revealed molecular profiles of non-ischemic and ischemic heart failure patients and explored the underlying mechanisms of etiology-specific impact on clinical outcome of LVAD support and tendency towards reverse remodeling.
Collapse
|
15
|
Shridas P, Patrick AC, Tannock LR. Role of Serum Amyloid A in Abdominal Aortic Aneurysm and Related Cardiovascular Diseases. Biomolecules 2021; 11:biom11121883. [PMID: 34944527 PMCID: PMC8699432 DOI: 10.3390/biom11121883] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/07/2021] [Accepted: 12/13/2021] [Indexed: 01/02/2023] Open
Abstract
Epidemiological data positively correlate plasma serum amyloid A (SAA) levels with cardiovascular disease severity and mortality. Studies by several investigators have indicated a causal role for SAA in the development of atherosclerosis in animal models. Suppression of SAA attenuates the development of angiotensin II (AngII)-induced abdominal aortic aneurysm (AAA) formation in mice. Thus, SAA is not just a marker for cardiovascular disease (CVD) development, but it is a key player. However, to consider SAA as a therapeutic target for these diseases, the pathway leading to its involvement needs to be understood. This review provides a brief description of the pathobiological significance of this enigmatic molecule. The purpose of this review is to summarize the data relevant to its role in the development of CVD, the pitfalls in SAA research, and unanswered questions in the field.
Collapse
Affiliation(s)
- Preetha Shridas
- Department of Internal Medicine, University of Kentucky, Lexington, KY 40536, USA
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY 40536, USA
- Barnstable Brown Diabetes Center, University of Kentucky, Lexington, KY 40536, USA
| | - Avery C Patrick
- Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Lisa R Tannock
- Department of Internal Medicine, University of Kentucky, Lexington, KY 40536, USA
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY 40536, USA
- Barnstable Brown Diabetes Center, University of Kentucky, Lexington, KY 40536, USA
- Veterans Affairs Lexington, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
16
|
Saiki O, Uda H. Ratio of serum amyloid A to C-reactive protein is constant in the same patients but differs greatly between patients with inflammatory diseases. Scand J Immunol 2021; 95:e13121. [PMID: 34796986 DOI: 10.1111/sji.13121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 11/13/2021] [Accepted: 11/17/2021] [Indexed: 11/28/2022]
Abstract
C-reactive protein (CRP) is commonly monitored to track the activity of inflammation and has become the gold standard in the management of all inflammatory diseases. Indeed, serum amyloid A (SAA) have seemed to correlate moderately with CRP, but the discrepancy of CRP and SAA levels has often been reported, especially in rheumatoid arthritis. Then, we examined CRP reflects a real magnitude of inflammation in patients with rheumatic and infectious inflammatory diseases. A total of 414 patients with infectious and non-infectious inflammatory diseases were enrolled. At initial visit, each patient underwent a clinical assessment and had also laboratory tests such as SAA and CRP. In each patient, we carried out a longitudinal analysis of CRP and SAA levels. We determined the inter-individual correlation between SAA and CRP and also clarified intra-individual changes of SAA/CRP ratio. SAA and CRP levels changed approximately linearly over time within individuals irrespective of rheumatic and infectious inflammatory diseases. However, SAA/CRP ratios differed dramatically between patients (from 0.117 to 50.8, median 5.71). In patients with high SAA/CRP ratio (>8.44), SAA is a better predictor of inflammation than CRP. In contrast, CRP is a better predictor in patients with low ratio (<3.52). Our results suggest that the SAA/CRP ratio differed greatly between individuals but was constant in intra-individuals. Low CRP levels could be accompanied by SAA levels predicting any degree of inflammation, implying that CRP is not reflecting a real magnitude of inflammation. To evaluate the real magnitude of inflammation, to access the SAA/CRP ratio in advance is essential.
Collapse
Affiliation(s)
- Osamu Saiki
- Department of Rheumatology, Higashiosaka City Medical Center, Higashiosaka City, Japan
| | - Hiroshi Uda
- Department of Rheumatology, Higashiosaka City Medical Center, Higashiosaka City, Japan
| |
Collapse
|
17
|
Uda H, Ebisu R, Maeda K, Saiki O. Discrepancy Between SAA and CRP levels Linked to the Difference of SAA/CRP ratio in Early Rheumatoid Arthritis. Mod Rheumatol 2021; 32:1035-1040. [PMID: 34792605 DOI: 10.1093/mr/roab097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/24/2021] [Accepted: 10/28/2021] [Indexed: 01/05/2023]
Abstract
OBJECTIVES Indeed, serum amyloid A (SAA) and C-reactive protein (CRP) reportedly seem to have moderate correlation, but discrepancies between CRP and SAA levels have often been reported in patients with early rheumatoid arthritis (ERA). This study aimed to determine the reasons for this discrepancy. METHODS ERA patients (n = 206) were enrolled and treated with anti-RA drugs. Clinical features and disease activities were estimated. CRP and SAA levels were monitored, and the SAA/CRP ratio was compared. Correlations between CRP and SAA levels in individuals and between individuals, and disease activity scores were examined. RESULTS In a follow-up study, the SAA/CRP ratio remained almost constant over time in the same patients. However, SAA/CRP ratios differed widely between patients (0.233-106.3). In patients with high SAA/CRP ratio (>6.52), many (26.2%) had abnormal SAA values only. In patients with low SAA/CRP ratio (<6.52), not a few (6.8%) exhibited abnormal CRP values only. CONCLUSIONS The SAA/CRP ratio remained virtually constant in the same patients but differed dramatically between patients, which clarifies the discrepancy between CRP and SAA levels. CRP is the better marker in low-ratio patients, but not in high-ratio patients; the SAA/CRP ratio is critical for its interpretation.
Collapse
Affiliation(s)
- Hiroshi Uda
- Department of Rheumatology, Higashiosaka City Medical Center, Osaka, Japan
| | - Rika Ebisu
- Department of Rheumatology, Higashiosaka City Medical Center, Osaka, Japan
| | - Keiji Maeda
- Department of Rheumatology, Higashiosaka City Medical Center, Osaka, Japan
| | - Osamu Saiki
- Department of Rheumatology, Higashiosaka City Medical Center, Osaka, Japan
| |
Collapse
|
18
|
Para I, Albu A, Porojan MD. Adipokines and Arterial Stiffness in Obesity. ACTA ACUST UNITED AC 2021; 57:medicina57070653. [PMID: 34202323 PMCID: PMC8305474 DOI: 10.3390/medicina57070653] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 06/17/2021] [Accepted: 06/21/2021] [Indexed: 12/11/2022]
Abstract
Adipokines are active molecules with pleiotropic effects produced by adipose tissue and involved in obesity-related metabolic and cardiovascular diseases. Arterial stiffness, which is a consequence of arteriosclerosis, has been shown to be an independent predictor of cardiovascular morbidity and mortality. The pathogenesis of arterial stiffness is complex but incompletely understood. Adipokines dysregulation may induce, by various mechanisms, vascular inflammation, endothelial dysfunction, and vascular remodeling, leading to increased arterial stiffness. This article summarizes literature data regarding adipokine-related pathogenetic mechanisms involved in the development of arterial stiffness, particularly in obesity, as well as the results of clinical and epidemiological studies which investigated the relationship between adipokines and arterial stiffness.
Collapse
Affiliation(s)
- Ioana Para
- 4th Department of Internal Medicine, University of Medicine and Pharmacy “Iuliu Hatieganu”, 400012 Cluj-Napoca, Romania;
| | - Adriana Albu
- 2nd Department of Internal Medicine, University of Medicine and Pharmacy “Iuliu Hatieganu”, 400012 Cluj-Napoca, Romania;
- Correspondence:
| | - Mihai D. Porojan
- 2nd Department of Internal Medicine, University of Medicine and Pharmacy “Iuliu Hatieganu”, 400012 Cluj-Napoca, Romania;
| |
Collapse
|
19
|
Chait A, Wang S, Goodspeed L, Gomes D, Turk KE, Wietecha T, Tang J, Storey C, O'Brien KD, Rubinow KB, Tang C, Vaisar T, Gharib SA, Lusis AJ, Den Hartigh LJ. Sexually Dimorphic Relationships Among Saa3 (Serum Amyloid A3), Inflammation, and Cholesterol Metabolism Modulate Atherosclerosis in Mice. Arterioscler Thromb Vasc Biol 2021; 41:e299-e313. [PMID: 33761762 PMCID: PMC8159856 DOI: 10.1161/atvbaha.121.316066] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Alan Chait
- Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition (A.C., S.W., L.G., D.G., K.E.T., J.T., C.S., K.B.R., C.T., T.V., L.J.D.H.), University of Washington, Seattle
- Diabetes Institute (A.C., S.W., L.G., D.G., K.E.T., T.W., J.T., C.S., K.D.O., K.B.R., C.T., T.V., L.J.D.H.), University of Washington, Seattle
| | - Shari Wang
- Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition (A.C., S.W., L.G., D.G., K.E.T., J.T., C.S., K.B.R., C.T., T.V., L.J.D.H.), University of Washington, Seattle
- Diabetes Institute (A.C., S.W., L.G., D.G., K.E.T., T.W., J.T., C.S., K.D.O., K.B.R., C.T., T.V., L.J.D.H.), University of Washington, Seattle
| | - Leela Goodspeed
- Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition (A.C., S.W., L.G., D.G., K.E.T., J.T., C.S., K.B.R., C.T., T.V., L.J.D.H.), University of Washington, Seattle
- Diabetes Institute (A.C., S.W., L.G., D.G., K.E.T., T.W., J.T., C.S., K.D.O., K.B.R., C.T., T.V., L.J.D.H.), University of Washington, Seattle
| | - Diego Gomes
- Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition (A.C., S.W., L.G., D.G., K.E.T., J.T., C.S., K.B.R., C.T., T.V., L.J.D.H.), University of Washington, Seattle
- Diabetes Institute (A.C., S.W., L.G., D.G., K.E.T., T.W., J.T., C.S., K.D.O., K.B.R., C.T., T.V., L.J.D.H.), University of Washington, Seattle
| | - Katherine E Turk
- Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition (A.C., S.W., L.G., D.G., K.E.T., J.T., C.S., K.B.R., C.T., T.V., L.J.D.H.), University of Washington, Seattle
- Diabetes Institute (A.C., S.W., L.G., D.G., K.E.T., T.W., J.T., C.S., K.D.O., K.B.R., C.T., T.V., L.J.D.H.), University of Washington, Seattle
| | - Tomasz Wietecha
- Diabetes Institute (A.C., S.W., L.G., D.G., K.E.T., T.W., J.T., C.S., K.D.O., K.B.R., C.T., T.V., L.J.D.H.), University of Washington, Seattle
- Department of Medicine, Division of Cardiology (T.W., K.D.O.), University of Washington, Seattle
| | - Jingjing Tang
- Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition (A.C., S.W., L.G., D.G., K.E.T., J.T., C.S., K.B.R., C.T., T.V., L.J.D.H.), University of Washington, Seattle
- Diabetes Institute (A.C., S.W., L.G., D.G., K.E.T., T.W., J.T., C.S., K.D.O., K.B.R., C.T., T.V., L.J.D.H.), University of Washington, Seattle
| | - Carl Storey
- Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition (A.C., S.W., L.G., D.G., K.E.T., J.T., C.S., K.B.R., C.T., T.V., L.J.D.H.), University of Washington, Seattle
- Diabetes Institute (A.C., S.W., L.G., D.G., K.E.T., T.W., J.T., C.S., K.D.O., K.B.R., C.T., T.V., L.J.D.H.), University of Washington, Seattle
| | - Kevin D O'Brien
- Diabetes Institute (A.C., S.W., L.G., D.G., K.E.T., T.W., J.T., C.S., K.D.O., K.B.R., C.T., T.V., L.J.D.H.), University of Washington, Seattle
- Department of Medicine, Division of Cardiology (T.W., K.D.O.), University of Washington, Seattle
| | - Katya B Rubinow
- Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition (A.C., S.W., L.G., D.G., K.E.T., J.T., C.S., K.B.R., C.T., T.V., L.J.D.H.), University of Washington, Seattle
- Diabetes Institute (A.C., S.W., L.G., D.G., K.E.T., T.W., J.T., C.S., K.D.O., K.B.R., C.T., T.V., L.J.D.H.), University of Washington, Seattle
| | - Chongren Tang
- Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition (A.C., S.W., L.G., D.G., K.E.T., J.T., C.S., K.B.R., C.T., T.V., L.J.D.H.), University of Washington, Seattle
- Diabetes Institute (A.C., S.W., L.G., D.G., K.E.T., T.W., J.T., C.S., K.D.O., K.B.R., C.T., T.V., L.J.D.H.), University of Washington, Seattle
| | - Tomas Vaisar
- Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition (A.C., S.W., L.G., D.G., K.E.T., J.T., C.S., K.B.R., C.T., T.V., L.J.D.H.), University of Washington, Seattle
- Diabetes Institute (A.C., S.W., L.G., D.G., K.E.T., T.W., J.T., C.S., K.D.O., K.B.R., C.T., T.V., L.J.D.H.), University of Washington, Seattle
| | - Sina A Gharib
- Division of Pulmonary, Critical Care and Sleep Medicine, Computational Medicine Core, Department of Medicine, Center for Lung Biology (S.A.G.), University of Washington, Seattle
| | - Aldons J Lusis
- Department of Human Genetics, University of California, Los Angeles (A.J.L.)
| | - Laura J Den Hartigh
- Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition (A.C., S.W., L.G., D.G., K.E.T., J.T., C.S., K.B.R., C.T., T.V., L.J.D.H.), University of Washington, Seattle
- Diabetes Institute (A.C., S.W., L.G., D.G., K.E.T., T.W., J.T., C.S., K.D.O., K.B.R., C.T., T.V., L.J.D.H.), University of Washington, Seattle
| |
Collapse
|
20
|
Abstract
PURPOSE OF REVIEW Serum amyloid A (SAA) is a highly sensitive acute phase reactant that has been linked to a number of chronic inflammatory diseases. During a systemic inflammatory response, liver-derived SAA is primarily found on high-density lipoprotein (HDL). The purpose of this review is to discuss recent literature addressing the pathophysiological functions of SAA and the significance of its association with HDL. RECENT FINDINGS Studies in gene-targeted mice establish that SAA contributes to atherosclerosis and some metastatic cancers. Accumulating evidence indicates that the lipidation state of SAA profoundly affects its bioactivities, with lipid-poor, but not HDL-associated, SAA capable of inducing inflammatory responses in vitro and in vivo. Factors that modulate the equilibrium between lipid-free and HDL-associated SAA have been identified. HDL may serve to limit SAA's bioactivities in vivo. Understanding the factors leading to the release of systemic SAA from HDL may provide insights into chronic disease mechanisms.
Collapse
Affiliation(s)
- Nancy R Webb
- Department of Pharmacology and Nutritional Sciences, Saha Cardiovascular Research Center, and Barnstable Brown Diabetes Center, University of Kentucky, 553 Wethington Building, 900 South Limestone, Lexington, KY, 40536-0200, USA.
| |
Collapse
|
21
|
Chang C, Pan Y, Du H, Wang X, Li X. Serum amyloid A1 can be a novel biomarker for evaluating the presence and severity of acute coronary syndrome. Clin Biochem 2020; 85:27-32. [PMID: 32805223 DOI: 10.1016/j.clinbiochem.2020.08.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/03/2020] [Accepted: 08/12/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Serum amyloid A (SAA) is an acute phase protein and a novel inflammatory biomarker of cardiovascular diseases. Of the four subtypes, SAA1 is the most representative biomarker. In this study, we aimed to assess the value of SAA1 as a novel biomarker for evaluating the presence and severity of acute coronary syndrome (ACS) in Chinese patients. METHODS AND RESULTS A total of 140 ACS patients and 88 non-ACS patients (including 36 stable coronary artery disease (SCAD) patients and 52 healthy controls) who underwent coronary angiography were enrolled. The SAA1 level was significantly higher in ACS patients compared with the SCAD and healthy control subgroups (P < 0.001, respectively), and was significantly higher in the high SYNTAX Score II (SS II) group compared with the medium SS II group and low SS II group (P < 0.001, respectively) in ACS patients. The cutoff level of SAA1 for indicating the presence of ACS was 324.65 ng/mL (sensitivity of 77.9%, specificity of 60.2% and an area under the curve of 0.717). The increased SAA1 levels were positively associated with the presence (OR = 1.013, P < 0.001) and severity (OR = 1.023, P < 0.001) of ACS. Furthermore, there was a positive correlation between SAA1 levels and SS II (r = 0.467, P < 0.001). CONCLUSIONS Our results suggest that elevated SAA1 levels may be a novel biomarker for evaluating the presence of ACS and the severity of CAD in ACS patients. Measuring SAA1 levels makes it possible to evaluate the presence of ACS and severity of CAD in ACS patients.
Collapse
Affiliation(s)
- Cheng Chang
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China
| | - Yilong Pan
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China
| | - Hongjiao Du
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China
| | - Xiao Wang
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China
| | - Xiaodong Li
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China.
| |
Collapse
|
22
|
Soppert J, Lehrke M, Marx N, Jankowski J, Noels H. Lipoproteins and lipids in cardiovascular disease: from mechanistic insights to therapeutic targeting. Adv Drug Deliv Rev 2020; 159:4-33. [PMID: 32730849 DOI: 10.1016/j.addr.2020.07.019] [Citation(s) in RCA: 141] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 07/20/2020] [Accepted: 07/22/2020] [Indexed: 12/12/2022]
Abstract
With cardiovascular disease being the leading cause of morbidity and mortality worldwide, effective and cost-efficient therapies to reduce cardiovascular risk are highly needed. Lipids and lipoprotein particles crucially contribute to atherosclerosis as underlying pathology of cardiovascular disease and influence inflammatory processes as well as function of leukocytes, vascular and cardiac cells, thereby impacting on vessels and heart. Statins form the first-line therapy with the aim to block cholesterol synthesis, but additional lipid-lowering drugs are sometimes needed to achieve low-density lipoprotein (LDL) cholesterol target values. Furthermore, beyond LDL cholesterol, also other lipid mediators contribute to cardiovascular risk. This review comprehensively discusses low- and high-density lipoprotein cholesterol, lipoprotein (a), triglycerides as well as fatty acids and derivatives in the context of cardiovascular disease, providing mechanistic insights into their role in pathological processes impacting on cardiovascular disease. Also, an overview of applied as well as emerging therapeutic strategies to reduce lipid-induced cardiovascular burden is provided.
Collapse
Affiliation(s)
- Josefin Soppert
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital Aachen, Aachen, Germany
| | - Michael Lehrke
- Medical Clinic I, University Hospital Aachen, Aachen, Germany
| | - Nikolaus Marx
- Medical Clinic I, University Hospital Aachen, Aachen, Germany
| | - Joachim Jankowski
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital Aachen, Aachen, Germany; Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, Maastricht University, the Netherlands
| | - Heidi Noels
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital Aachen, Aachen, Germany; Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands.
| |
Collapse
|
23
|
Abstract
PURPOSE OF REVIEW Acute phase serum amyloid A (SAA) is persistently elevated in chronic inflammatory conditions, and elevated levels predict cardiovascular risk in humans. More recently, murine studies have demonstrated that over-expression of SAA increases and deficiency/suppression of SAA attenuates atherosclerosis. Thus, beyond being a biomarker, SAA appears to play a causal role in atherogenesis. The purpose of this review is to summarize the data supporting SAA as a key player in atherosclerosis development. RECENT FINDINGS A number of pro-inflammatory and pro-atherogenic activities have been ascribed to SAA. However, the literature is conflicted, as recombinant SAA, and/or lipid-free SAA, used in many of the earlier studies, do not reflect the activity of native human or murine SAA, which exists largely lipid-associated. Recent literatures demonstrate that SAA activates the NLRP3 inflammasome, alters vascular function, affects HDL function, and increases thrombosis. Importantly, SAA activity appears to be regulated by its lipid association, and HDL may serve to sequester and limit SAA activity. SUMMARY SAA has many pro-inflammatory and pro-atherogenic activities, is clearly demonstrated to affect atherosclerosis development, and may be a candidate target for clinical trials in cardiovascular diseases.
Collapse
Affiliation(s)
- Preetha Shridas
- Department of Internal Medicine
- Saha Cardiovascular Research Center
- Barnstable Brown Diabetes Center and University of Kentucky
| | - Lisa R Tannock
- Department of Internal Medicine
- Saha Cardiovascular Research Center
- Barnstable Brown Diabetes Center and University of Kentucky
- Veterans Affairs Lexington, Lexington, Kentucky, USA
| |
Collapse
|
24
|
Carabias CS, Castaño-León AM, Blanca Navarro B, Panero I, Eiriz C, Gómez PA, Egea J, Lagares A. Serum Amyloid A1 as a Potential Intracranial and Extracranial Clinical Severity Biomarker in Traumatic Brain Injury. J Intensive Care Med 2019; 35:1180-1195. [PMID: 30961443 DOI: 10.1177/0885066619837913] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Extracranial injury is frequently present in patients with traumatic brain injury (TBI). However, no reliable biomarker exists nowadays to evaluate the magnitude and extension of extracranial injury as well as the identification of patients who are at risk of developing secondary injuries. The purpose of this study was to identify new possible peptide biomarkers by mass spectrometry analysis in patients with TBI and ascertain whether the novel biomarker discovered by peptide mass fingerprinting, serum amyloid A1 (SAA1), is capable of reflecting the condition of the patient and both intracranial and extracranial injury extension. Demographic characteristics, clinical data, and serum samples were prospectively collected from 120 patients with TBI (Glasgow Coma Scale [GCS] score 3-15) on admission. Biomarkers were quantified by enzyme-linked immunosorbent assay. Intracranial lesion volume was measured from the semiautomatic segmentation of hematoma on computed tomography (CT) using Analyze software. Functional outcome was evaluated using the Glasgow Outcome Scale (GOS) at hospital discharge and GOS extended scores at 6 months. The SAA1 levels were significantly associated with intracranial (GCS score at admission, lesion load measured with cranial CT, and pupil responsiveness) and extracranial clinical severity (all Abbreviated Injury Scale regions, Injury Severity Score, major extracranial injury, polytrauma, and orthopedic fractures presence), along with systemic secondary insults and functional outcome. SAA1 was is associated with the volume of traumatic intracranial lesions. The SAA1 levels were correlated with astroglial S100β and glial fibrillary acidic protein (GFAP), neuronal neuron-specific enolase (NSE), and axonal total tau (T-tau) and phosphorylated neurofilament heavy chain (pNF-H) injury markers. SAA1 predicts unfavorable outcome and mortality at hospital discharge (area under the curve [AUC] = 0.90, 0.82) and 6 months (AUC = 0.89). SAA1 can be established as a marker for the overall patient condition due to its involvement in the neuroendocrine axis of the systemic response to craniocerebral trauma.
Collapse
Affiliation(s)
- Cristina Sánchez Carabias
- Department of Neurosurgery, Neurotraumatology and Subarachnoid Hemorrhage Research Unit, Instituto de Investigación 16473Hospital 12 de Octubre (i+12), Madrid, Spain
| | | | - B Blanca Navarro
- Department of Neurosurgery, 16473Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Irene Panero
- Department of Neurosurgery, 16473Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Carla Eiriz
- Department of Neurosurgery, 16473Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Pedro A Gómez
- Department of Neurosurgery, 16473Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Javier Egea
- Molecular Neuroinflammation and Neuronal Plasticity Research Unit, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria Hospital Universitario La Princesa, Madrid, Spain
| | - Alfonso Lagares
- Department of Neurosurgery, Neurotraumatology and Subarachnoid Hemorrhage Research Unit, Instituto de Investigación 16473Hospital 12 de Octubre (i+12), Madrid, Spain.,Department of Neurosurgery, 16473Hospital Universitario 12 de Octubre, Madrid, Spain
| |
Collapse
|
25
|
Wilson PG, Thompson JC, Shridas P, McNamara PJ, de Beer MC, de Beer FC, Webb NR, Tannock LR. Serum Amyloid A Is an Exchangeable Apolipoprotein. Arterioscler Thromb Vasc Biol 2018; 38:1890-1900. [PMID: 29976766 PMCID: PMC6202200 DOI: 10.1161/atvbaha.118.310979] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Objective- SAA (serum amyloid A) is a family of acute-phase reactants that have proinflammatory and proatherogenic activities. SAA is more lipophilic than apoA-I (apolipoprotein A-I), and during an acute-phase response, <10% of plasma SAA is found lipid-free. In most reports, SAA is found exclusively associated with high-density lipoprotein; however, we and others have reported SAA on apoB (apolipoprotein B)-containing lipoproteins in both mice and humans. The goal of this study was to determine whether SAA is an exchangeable apolipoprotein. Approach and Results- Delipidated human SAA was incubated with SAA-free human lipoproteins; then, samples were reisolated by fast protein liquid chromatography, and SAA analyzed by ELISA and immunoblot. Both in vitro and in vivo, we show that SAA associates with any lipoprotein and does not remain in a lipid-free form. Although SAA is preferentially found on high-density lipoprotein, it can exchange between lipoproteins. In the presence of CETP (cholesterol ester transfer protein), there is greater exchange of SAA between lipoproteins. Subjects with diabetes mellitus, but not those with metabolic syndrome, showed altered SAA lipoprotein distribution postprandially. Proteoglycan-mediated lipoprotein retention is thought to be an underlying mechanism for atherosclerosis development. SAA has a proteoglycan-binding domain. Lipoproteins containing SAA had increased proteoglycan binding compared with SAA-free lipoproteins. Conclusions- Thus, SAA is an exchangeable apolipoprotein and increases apoB-containing lipoproteins' proteoglycan binding. We and others have previously reported the presence of SAA on low-density lipoprotein in individuals with obesity, diabetes mellitus, and metabolic syndrome. We propose that the presence of SAA on apoB-containing lipoproteins may contribute to cardiovascular disease development in these populations.
Collapse
Affiliation(s)
- Patricia G Wilson
- Department of Veterans Affairs, Lexington, KY
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky
- Barnstable Brown Diabetes Center, College of Medicine, University of Kentucky
| | - Joel C Thompson
- Department of Veterans Affairs, Lexington, KY
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky
- Barnstable Brown Diabetes Center, College of Medicine, University of Kentucky
| | - Preetha Shridas
- Department of Internal Medicine, College of Medicine, University of Kentucky
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky
- Barnstable Brown Diabetes Center, College of Medicine, University of Kentucky
| | - Patrick J McNamara
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky
| | - Maria C de Beer
- Department of Physiology, College of Medicine, University of Kentucky
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky
- Barnstable Brown Diabetes Center, College of Medicine, University of Kentucky
| | - Frederick C de Beer
- Department of Internal Medicine, College of Medicine, University of Kentucky
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky
- Barnstable Brown Diabetes Center, College of Medicine, University of Kentucky
| | - Nancy R Webb
- Department of Veterans Affairs, Lexington, KY
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky
- Barnstable Brown Diabetes Center, College of Medicine, University of Kentucky
| | - Lisa R Tannock
- Department of Veterans Affairs, Lexington, KY
- Department of Internal Medicine, College of Medicine, University of Kentucky
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky
- Barnstable Brown Diabetes Center, College of Medicine, University of Kentucky
| |
Collapse
|
26
|
Hirsch D, Evans CA, Wong M, Machaalani R, Waters KA. Biochemical markers of cardiac dysfunction in children with obstructive sleep apnoea (OSA). Sleep Breath 2018; 23:95-101. [DOI: 10.1007/s11325-018-1666-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 04/17/2018] [Accepted: 04/19/2018] [Indexed: 12/13/2022]
|
27
|
Linhares LMC, Oliveira CP, Alvares-da-Silva MR, Stefano JT, Barbeiro HV, Barbeiro DF, Terrabuio DRB, Abdala E, Soriano FG, Carrilho FJ, Farias AQ, Siddiqui MS, D'Albuquerque LAC. Evolution of Biomarkers of Atherogenic Risk in Liver Transplantation Recipients. Transplant Proc 2018; 50:3650-3655. [PMID: 30586839 DOI: 10.1016/j.transproceed.2018.04.030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 04/12/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND Cardiovascular disease is a major contributing factor to long-term mortality after liver transplantation (LT). METHODS This study evaluated the evolution of atherogenic risk in liver transplant recipients (LTRs). Thirty-six subjects were prospectively enrolled at 12 months and followed for 48 months after liver transplantation. Serum biomarkers of endothelial dysfunction (sICAM-1 and sVCAM-1), chronic inflammation (serum amyloid A), and oxidative stress (myeloperoxidase) were measured at 12 and 48 months after LT. Additionally, at 12 months all patients underwent a cardiac computed tomography (CT) scan and a coronary artery calcium score (CACS). RESULTS The prevalence of risk factors of metabolic syndrome (MS) increased over the course of the study. The patients' sVCAM-1 and sICAM-1 increased from 1.82 ± 0.44 ng/mL to 9.10 ± 5.82 ng/mL (P < .001) and 0.23 ± 0.09 ng/mL to 2.7 ± 3.3 ng/mL, respectively from month 12 to 48. Serum myeloperoxidase increased from 0.09 ± 0.07 ng/mL to 3.46 ± 3.92 ng/mL (P < .001) over the course of the study. Serum amyloid A also increased from 21.4 ± 40.7 ng/mL at entry to 91.5 ± 143.6 ng/mL at end of study (P < .001). CONCLUSION No association between these biomarkers and MS was noted. The cardiac CT revealed mild and moderate disease in 19% and 25% of the cohort, respectively. No association between serum biomarkers and CACS was noted. Serum biomarkers of atherogenic risk increase rapidly in LTRs and precede coronary plaques.
Collapse
Affiliation(s)
- L M C Linhares
- Department of Gastroenterology (LIM-07/LIM-37), University of São Paulo School of Medicine, São Paulo, Brazil
| | - C P Oliveira
- Department of Gastroenterology (LIM-07/LIM-37), University of São Paulo School of Medicine, São Paulo, Brazil.
| | - M R Alvares-da-Silva
- Division of Gastroenterology, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - J T Stefano
- Department of Gastroenterology (LIM-07/LIM-37), University of São Paulo School of Medicine, São Paulo, Brazil
| | - H V Barbeiro
- Division of Emergency Medicine (LIM-51), University of São Paulo School of Medicine, São Paulo, Brazil
| | - D F Barbeiro
- Division of Emergency Medicine (LIM-51), University of São Paulo School of Medicine, São Paulo, Brazil
| | - D R B Terrabuio
- Department of Gastroenterology (LIM-07/LIM-37), University of São Paulo School of Medicine, São Paulo, Brazil
| | - E Abdala
- Department of Gastroenterology (LIM-07/LIM-37), University of São Paulo School of Medicine, São Paulo, Brazil
| | - F G Soriano
- Division of Emergency Medicine (LIM-51), University of São Paulo School of Medicine, São Paulo, Brazil
| | - F J Carrilho
- Department of Gastroenterology (LIM-07/LIM-37), University of São Paulo School of Medicine, São Paulo, Brazil
| | - A Q Farias
- Department of Gastroenterology (LIM-07/LIM-37), University of São Paulo School of Medicine, São Paulo, Brazil
| | - M S Siddiqui
- Division of Gastroenterology and Hepatology, Virginia Commonwealth University, Richmond, VA
| | - L A C D'Albuquerque
- Department of Gastroenterology (LIM-07/LIM-37), University of São Paulo School of Medicine, São Paulo, Brazil
| |
Collapse
|
28
|
Tannock LR, De Beer MC, Ji A, Shridas P, Noffsinger VP, den Hartigh L, Chait A, De Beer FC, Webb NR. Serum amyloid A3 is a high density lipoprotein-associated acute-phase protein. J Lipid Res 2017; 59:339-347. [PMID: 29247043 DOI: 10.1194/jlr.m080887] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 11/22/2017] [Indexed: 12/20/2022] Open
Abstract
Serum amyloid A (SAA) is a family of acute-phase reactants. Plasma levels of human SAA1/SAA2 (mouse SAA1.1/2.1) can increase ≥1,000-fold during an acute-phase response. Mice, but not humans, express a third relatively understudied SAA isoform, SAA3. We investigated whether mouse SAA3 is an HDL-associated acute-phase SAA. Quantitative RT-PCR with isoform-specific primers indicated that SAA3 and SAA1.1/2.1 are induced similarly in livers (∼2,500-fold vs. ∼6,000-fold, respectively) and fat (∼400-fold vs. ∼100-fold, respectively) of lipopolysaccharide (LPS)-injected mice. In situ hybridization demonstrated that all three SAAs are produced by hepatocytes. All three SAA isoforms were detected in plasma of LPS-injected mice, although SAA3 levels were ∼20% of SAA1.1/2.1 levels. Fast protein LC analyses indicated that virtually all of SAA1.1/2.1 eluted with HDL, whereas ∼15% of SAA3 was lipid poor/free. After density gradient ultracentrifugation, isoelectric focusing demonstrated that ∼100% of plasma SAA1.1 was recovered in HDL compared with only ∼50% of SAA2.1 and ∼10% of SAA3. Thus, SAA3 appears to be more loosely associated with HDL, resulting in lipid-poor/free SAA3. We conclude that SAA3 is a major hepatic acute-phase SAA in mice that may produce systemic effects during inflammation.
Collapse
Affiliation(s)
- Lisa R Tannock
- Departments of Internal Medicine, University of Kentucky, Lexington, KY.,Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY.,Barnstable Brown Diabetes Center, University of Kentucky, Lexington, KY.,Veterans Affairs Lexington, University of Kentucky, Lexington, KY
| | - Maria C De Beer
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY.,Barnstable Brown Diabetes Center, University of Kentucky, Lexington, KY.,Departments of Physiology, University of Kentucky, Lexington, KY
| | - Ailing Ji
- Departments of Internal Medicine, University of Kentucky, Lexington, KY.,Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
| | - Preetha Shridas
- Departments of Internal Medicine, University of Kentucky, Lexington, KY.,Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY.,Barnstable Brown Diabetes Center, University of Kentucky, Lexington, KY
| | - Victoria P Noffsinger
- Departments of Internal Medicine, University of Kentucky, Lexington, KY.,Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
| | - Laura den Hartigh
- Department of Medicine University of Washington, Seattle, WA.,University of Washington Diabetes Institute, University of Washington, Seattle, WA
| | - Alan Chait
- Department of Medicine University of Washington, Seattle, WA.,University of Washington Diabetes Institute, University of Washington, Seattle, WA
| | - Frederick C De Beer
- Departments of Internal Medicine, University of Kentucky, Lexington, KY.,Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY.,Barnstable Brown Diabetes Center, University of Kentucky, Lexington, KY
| | - Nancy R Webb
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY .,Barnstable Brown Diabetes Center, University of Kentucky, Lexington, KY.,Veterans Affairs Lexington, University of Kentucky, Lexington, KY.,Departments of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY
| |
Collapse
|
29
|
Komiyama M, Shimada S, Wada H, Yamakage H, Satoh-Asahara N, Shimatsu A, Akao M, Morimoto T, Takahashi Y, Hasegawa K. Time-dependent Changes of Atherosclerotic LDL Complexes after Smoking Cessation. J Atheroscler Thromb 2016; 23:1270-1275. [PMID: 27298048 PMCID: PMC5113744 DOI: 10.5551/jat.34280] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Aim: The α1-antitrypsin – low-density lipoprotein complex (AT-LDL) and serum amyloid A-LDL complex (SAA-LDL) are oxidatively modified LDL complexes that promote atherosclerosis. The serum levels of AT-LDL and SAA-LDL are suggested to be increased by obesity and smoking. We have previously demonstrated that larger weight gain after smoking cessation (SC) perturbs a decrease in the serum level of AT-LDL at 3 months after SC. However, changes of these atherosclerotic makers >3 months after SC are unknown. This study investigated post-SC time-dependent changes in two atherogenic lipoproteins, AT-LDL and SAA-LDL, and in the extent of abdominal obesity. Methods: In 50 outpatients who had continued SC for 1 year, we measured serum AT-LDL and SAA-LDL levels by the enzyme-linked immunosorbent assay before SC, and at 3 months and 1 year after SC. Results: Both body mass index and waist circumstance significantly increased from pre-SC to 3 months after SC and from 3 months after SC to 1 year after SC. Although the serum levels of AT-LDL and SAA-LDL were unchanged from pre-SC to 3 months after SC, these levels decreased significantly from 3 months after SC to 1 year after SC. Conclusions: The extent of abdominal obesity and levels of two atherogenic lipoproteins time-dependently change after SC. Although abdominal obesity progressively worsened after SC, the beneficial effect of non-smoking overcomes the potential vascular risks by cessation-associated obesity at 1 year after SC.
Collapse
Affiliation(s)
- Maki Komiyama
- Clinical Research Institute, National Hospital Organization Kyoto Medical Center
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Hwang YG, Balasubramani GK, Metes ID, Levesque MC, Bridges SL, Moreland LW. Differential response of serum amyloid A to different therapies in early rheumatoid arthritis and its potential value as a disease activity biomarker. Arthritis Res Ther 2016; 18:108. [PMID: 27188329 PMCID: PMC4869396 DOI: 10.1186/s13075-016-1009-y] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Accepted: 04/29/2016] [Indexed: 12/23/2022] Open
Abstract
Background The aim was to compare the effect of etanercept (ETN) and conventional synthetic disease-modifying anti-rheumatic drug (DMARD) therapy on serum amyloid A (SAA) levels and to determine whether SAA reflects rheumatoid arthritis (RA) disease activity better than C-reactive protein (CRP). Methods We measured SAA and CRP at baseline, 24, 48, and 102 week follow-up visits in 594 patients participating in the Treatment of early RA (TEAR) study. We used Spearman correlation coefficients (rho) to evaluate the relationship between SAA and CRP and mixed effects models to determine whether ETN and methotrexate (MTX) treatment compared to triple DMARD therapy differentially lowered SAA. Akaike information criteria (AIC) were used to determine model fits. Results SAA levels were only moderately correlated with CRP levels (rho = 0.58, p < 0.0001). There were significant differences in SAA by both visit (p = 0.0197) and treatment arm (p = 0.0130). RA patients treated with ETN plus MTX had a larger reduction in SAA than patients treated with traditional DMARD therapy. Similar results were found for serum CRP by visit (p = 0.0254) and by treatment (p < 0.0001), with a more pronounced difference than for SAA. Across all patients and time points, models of the disease activity score of 28 joints (DAS28)-erythrocyte sedimentation rate (ESR) using SAA levels were better than models using CRP; the ΔAIC between the SAA and CRP models was 305. Conclusions SAA may be a better biomarker of RA disease activity than CRP, especially during treatment with tumor necrosis factor (TNF) antagonists. This warrants additional studies in other cohorts of patients on treatment for RA. Trial registration (ClinicalTrials.gov identifier: NCT00259610, Date of registration: 28 November 2005)
Collapse
Affiliation(s)
- Yong Gil Hwang
- Department of Medicine, Division of Rheumatology and Clinical Immunology, University of Pittsburgh, 3500 Terrace Street, Pittsburgh, PA, 15261, USA.
| | - Goundappa K Balasubramani
- Department of Epidemiology, School of Public Health, University of Pittsburgh, 130 DeSoto Street, 127 Parran Hall, Pittsburgh, PA, 15261, USA
| | - Ilinca D Metes
- Department of Medicine, Division of Rheumatology and Clinical Immunology, University of Pittsburgh, 3500 Terrace Street, Pittsburgh, PA, 15261, USA
| | - Marc C Levesque
- Department of Medicine, Division of Rheumatology and Clinical Immunology, University of Pittsburgh, 3500 Terrace Street, Pittsburgh, PA, 15261, USA.,AbbVie Inc, 100 Research Dr, Worcester, MA, 01605, USA
| | - S Louis Bridges
- Department of Medicine, Division of Clinical Immunology and Rheumatology Birmingham, University of Alabama at Birmingham, Shelby Building, Room 178B, 1825 University Blvd., Birmingham, AL, 35294-2182, USA
| | - Larry W Moreland
- Department of Medicine, Division of Rheumatology and Clinical Immunology, University of Pittsburgh, 3500 Terrace Street, Pittsburgh, PA, 15261, USA
| |
Collapse
|
31
|
Kotani K, Shuumarjav U, Taniguchi N, Yamada T. Possible relationship between the heart rates and serum amyloid A in a hyperglycemic population. Biosci Trends 2015; 9:79-81. [PMID: 25787913 DOI: 10.5582/bst.2014.01114] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Hyperglycemia predicts cardiovascular disease (CVD)-related outcomes. The resting heart rates (HRs) and serum amyloid A (SAA), an inflammatory marker, are respectively factors associated with CVD-related outcomes; however, little is known regarding the associations between these two factors. This study aimed to investigate the correlation between the HRs and SAA levels under hyperglycemic conditions. This study included 298 subjects (males, 44%; mean age, 61.1 years) without a history of CVD and/or hypertensive levels. Clinical data, including general laboratory measurements, HRs and SAA, were measured. The analyses were performed after dividing all of the subjects into two groups based on the blood glucose level (< or ≥ 6.1 mmol/L). There was a higher SAA level in the hyperglycemic group (n = 143; median [interquartile range] 6.1 [4.1-10.6] μg/mL) than in the counterpart group (n = 155; 6.0 [3.5-8.5] μg/mL; p < 0.01). There was a trend toward increased HRs in the hyperglycemic group (mean [standard deviation] 65.3 [11.2] bpm) compared to the counterpart group (63.2 [9.4] bpm; p = 0.08). In the hyperglycemic group, there was a significant positive correlation between the HRs and SAA levels (multiple variables-adjusted analysis: β = 0.21, p = 0.02), while no correlation was found in the counterpart group (β = 0.06, p = 0.50). In summary, a positive correlation between the HRs and SAA levels can present under hyperglycemic conditions. These findings may provide relevant insights into the CVD-related pathologies associated with hyperglycemia. Further studies are warranted.
Collapse
Affiliation(s)
- Kazuhiko Kotani
- Department of Clinical Laboratory Medicine, Jichi Medical University
| | | | | | | |
Collapse
|
32
|
Dysfunctional High-Density Lipoprotein: An Innovative Target for Proteomics and Lipidomics. CHOLESTEROL 2015; 2015:296417. [PMID: 26634153 PMCID: PMC4655037 DOI: 10.1155/2015/296417] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Revised: 10/12/2015] [Accepted: 10/12/2015] [Indexed: 02/02/2023]
Abstract
High-Density Lipoprotein-Cholesterol (HDL-C) is regarded as an important protective factor against cardiovascular disease, with abundant evidence of an inverse relationship between its serum levels and risk of cardiovascular disease, as well as various antiatherogenic, antioxidant, and anti-inflammatory properties. Nevertheless, observations of hereditary syndromes featuring scant HDL-C concentration in absence of premature atherosclerotic disease suggest HDL-C levels may not be the best predictor of cardiovascular disease. Indeed, the beneficial effects of HDL may not depend solely on their concentration, but also on their quality. Distinct subfractions of this lipoprotein appear to be constituted by specific protein-lipid conglomerates necessary for different physiologic and pathophysiologic functions. However, in a chronic inflammatory microenvironment, diverse components of the HDL proteome and lipid core suffer alterations, which propel a shift towards a dysfunctional state, where HDL-C becomes proatherogenic, prooxidant, and proinflammatory. This heterogeneity highlights the need for further specialized molecular studies in this aspect, in order to achieve a better understanding of this dysfunctional state; with an emphasis on the potential role for proteomics and lipidomics as valuable methods in the search of novel therapeutic approaches for cardiovascular disease.
Collapse
|
33
|
Beck HC, Overgaard M, Melholt Rasmussen L. Plasma proteomics to identify biomarkers – application to cardiovascular diseases. TRANSLATIONAL PROTEOMICS 2015. [DOI: 10.1016/j.trprot.2015.01.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
|
34
|
Webb NR, De Beer MC, Wroblewski JM, Ji A, Bailey W, Shridas P, Charnigo RJ, Noffsinger VP, Witta J, Howatt DA, Balakrishnan A, Rateri DL, Daugherty A, De Beer FC. Deficiency of Endogenous Acute-Phase Serum Amyloid A Protects apoE-/- Mice From Angiotensin II-Induced Abdominal Aortic Aneurysm Formation. Arterioscler Thromb Vasc Biol 2015; 35:1156-65. [PMID: 25745063 DOI: 10.1161/atvbaha.114.304776] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 02/13/2015] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Rupture of abdominal aortic aneurysm (AAA), a major cause of death in the aged population, is characterized by vascular inflammation and matrix degradation. Serum amyloid A (SAA), an acute-phase reactant linked to inflammation and matrix metalloproteinase induction, correlates with aortic dimensions before aneurysm formation in humans. We investigated whether SAA deficiency in mice affects AAA formation during angiotensin II (Ang II) infusion. APPROACH AND RESULTS Plasma SAA increased ≈60-fold in apoE(-/-) mice 24 hours after intraperitoneal Ang II injection (100 μg/kg; n=4) and ≈15-fold after chronic 28-day Ang II infusion (1000 ng/kg per minute; n=9). AAA incidence and severity after 28-day Ang II infusion was significantly reduced in apoE(-/-) mice lacking both acute-phase SAA isoforms (SAAKO; n=20) compared with apoE(-/-) mice (SAAWT; n=20) as assessed by in vivo ultrasound and ex vivo morphometric analyses, despite a significant increase in systolic blood pressure in SAAKO mice compared with SAAWT mice after Ang II infusion. Atherosclerotic lesion area of the aortic arch was similar in SAAKO and SAAWT mice after 28-day Ang II infusion. Immunostaining detected SAA in AAA tissues of Ang II-infused SAAWT mice that colocalized with macrophages, elastin breaks, and enhanced matrix metalloproteinase activity. Matrix metalloproteinase-2 activity was significantly lower in aortas of SAAKO mice compared with SAAWT mice after 10-day Ang II infusion. CONCLUSIONS Lack of endogenous acute-phase SAA protects against experimental AAA through a mechanism that may involve reduced matrix metalloproteinase-2 activity.
Collapse
Affiliation(s)
- Nancy R Webb
- From the Departments of Pharmacology Division of Nutritional Sciences (N.R.W.), Physiology (M.C.D.B.) and Internal Medicine (J.M.W., A.J., W.B., P.S., V.P.N., D.A.H., A.B., D.L.R., A.D., F.C.D.B.), and Saha Cardiovascular Research Center (N.R.W., M.C.D.B., J.M.W., A.J., P.S., V.P.N., D.A.H., A.B., D.L.R., A.D., F.C.D.B.), and Departments of Statistics and Biostatistics (R.J.C.), University of Kentucky, Lexington; and Foundation Gastroenterology, Nashua, NH (J.W.).
| | - Maria C De Beer
- From the Departments of Pharmacology Division of Nutritional Sciences (N.R.W.), Physiology (M.C.D.B.) and Internal Medicine (J.M.W., A.J., W.B., P.S., V.P.N., D.A.H., A.B., D.L.R., A.D., F.C.D.B.), and Saha Cardiovascular Research Center (N.R.W., M.C.D.B., J.M.W., A.J., P.S., V.P.N., D.A.H., A.B., D.L.R., A.D., F.C.D.B.), and Departments of Statistics and Biostatistics (R.J.C.), University of Kentucky, Lexington; and Foundation Gastroenterology, Nashua, NH (J.W.)
| | - Joanne M Wroblewski
- From the Departments of Pharmacology Division of Nutritional Sciences (N.R.W.), Physiology (M.C.D.B.) and Internal Medicine (J.M.W., A.J., W.B., P.S., V.P.N., D.A.H., A.B., D.L.R., A.D., F.C.D.B.), and Saha Cardiovascular Research Center (N.R.W., M.C.D.B., J.M.W., A.J., P.S., V.P.N., D.A.H., A.B., D.L.R., A.D., F.C.D.B.), and Departments of Statistics and Biostatistics (R.J.C.), University of Kentucky, Lexington; and Foundation Gastroenterology, Nashua, NH (J.W.)
| | - Ailing Ji
- From the Departments of Pharmacology Division of Nutritional Sciences (N.R.W.), Physiology (M.C.D.B.) and Internal Medicine (J.M.W., A.J., W.B., P.S., V.P.N., D.A.H., A.B., D.L.R., A.D., F.C.D.B.), and Saha Cardiovascular Research Center (N.R.W., M.C.D.B., J.M.W., A.J., P.S., V.P.N., D.A.H., A.B., D.L.R., A.D., F.C.D.B.), and Departments of Statistics and Biostatistics (R.J.C.), University of Kentucky, Lexington; and Foundation Gastroenterology, Nashua, NH (J.W.)
| | - William Bailey
- From the Departments of Pharmacology Division of Nutritional Sciences (N.R.W.), Physiology (M.C.D.B.) and Internal Medicine (J.M.W., A.J., W.B., P.S., V.P.N., D.A.H., A.B., D.L.R., A.D., F.C.D.B.), and Saha Cardiovascular Research Center (N.R.W., M.C.D.B., J.M.W., A.J., P.S., V.P.N., D.A.H., A.B., D.L.R., A.D., F.C.D.B.), and Departments of Statistics and Biostatistics (R.J.C.), University of Kentucky, Lexington; and Foundation Gastroenterology, Nashua, NH (J.W.)
| | - Preetha Shridas
- From the Departments of Pharmacology Division of Nutritional Sciences (N.R.W.), Physiology (M.C.D.B.) and Internal Medicine (J.M.W., A.J., W.B., P.S., V.P.N., D.A.H., A.B., D.L.R., A.D., F.C.D.B.), and Saha Cardiovascular Research Center (N.R.W., M.C.D.B., J.M.W., A.J., P.S., V.P.N., D.A.H., A.B., D.L.R., A.D., F.C.D.B.), and Departments of Statistics and Biostatistics (R.J.C.), University of Kentucky, Lexington; and Foundation Gastroenterology, Nashua, NH (J.W.)
| | - Richard J Charnigo
- From the Departments of Pharmacology Division of Nutritional Sciences (N.R.W.), Physiology (M.C.D.B.) and Internal Medicine (J.M.W., A.J., W.B., P.S., V.P.N., D.A.H., A.B., D.L.R., A.D., F.C.D.B.), and Saha Cardiovascular Research Center (N.R.W., M.C.D.B., J.M.W., A.J., P.S., V.P.N., D.A.H., A.B., D.L.R., A.D., F.C.D.B.), and Departments of Statistics and Biostatistics (R.J.C.), University of Kentucky, Lexington; and Foundation Gastroenterology, Nashua, NH (J.W.)
| | - Victoria P Noffsinger
- From the Departments of Pharmacology Division of Nutritional Sciences (N.R.W.), Physiology (M.C.D.B.) and Internal Medicine (J.M.W., A.J., W.B., P.S., V.P.N., D.A.H., A.B., D.L.R., A.D., F.C.D.B.), and Saha Cardiovascular Research Center (N.R.W., M.C.D.B., J.M.W., A.J., P.S., V.P.N., D.A.H., A.B., D.L.R., A.D., F.C.D.B.), and Departments of Statistics and Biostatistics (R.J.C.), University of Kentucky, Lexington; and Foundation Gastroenterology, Nashua, NH (J.W.)
| | - Jassir Witta
- From the Departments of Pharmacology Division of Nutritional Sciences (N.R.W.), Physiology (M.C.D.B.) and Internal Medicine (J.M.W., A.J., W.B., P.S., V.P.N., D.A.H., A.B., D.L.R., A.D., F.C.D.B.), and Saha Cardiovascular Research Center (N.R.W., M.C.D.B., J.M.W., A.J., P.S., V.P.N., D.A.H., A.B., D.L.R., A.D., F.C.D.B.), and Departments of Statistics and Biostatistics (R.J.C.), University of Kentucky, Lexington; and Foundation Gastroenterology, Nashua, NH (J.W.)
| | - Deborah A Howatt
- From the Departments of Pharmacology Division of Nutritional Sciences (N.R.W.), Physiology (M.C.D.B.) and Internal Medicine (J.M.W., A.J., W.B., P.S., V.P.N., D.A.H., A.B., D.L.R., A.D., F.C.D.B.), and Saha Cardiovascular Research Center (N.R.W., M.C.D.B., J.M.W., A.J., P.S., V.P.N., D.A.H., A.B., D.L.R., A.D., F.C.D.B.), and Departments of Statistics and Biostatistics (R.J.C.), University of Kentucky, Lexington; and Foundation Gastroenterology, Nashua, NH (J.W.)
| | - Anju Balakrishnan
- From the Departments of Pharmacology Division of Nutritional Sciences (N.R.W.), Physiology (M.C.D.B.) and Internal Medicine (J.M.W., A.J., W.B., P.S., V.P.N., D.A.H., A.B., D.L.R., A.D., F.C.D.B.), and Saha Cardiovascular Research Center (N.R.W., M.C.D.B., J.M.W., A.J., P.S., V.P.N., D.A.H., A.B., D.L.R., A.D., F.C.D.B.), and Departments of Statistics and Biostatistics (R.J.C.), University of Kentucky, Lexington; and Foundation Gastroenterology, Nashua, NH (J.W.)
| | - Debra L Rateri
- From the Departments of Pharmacology Division of Nutritional Sciences (N.R.W.), Physiology (M.C.D.B.) and Internal Medicine (J.M.W., A.J., W.B., P.S., V.P.N., D.A.H., A.B., D.L.R., A.D., F.C.D.B.), and Saha Cardiovascular Research Center (N.R.W., M.C.D.B., J.M.W., A.J., P.S., V.P.N., D.A.H., A.B., D.L.R., A.D., F.C.D.B.), and Departments of Statistics and Biostatistics (R.J.C.), University of Kentucky, Lexington; and Foundation Gastroenterology, Nashua, NH (J.W.)
| | - Alan Daugherty
- From the Departments of Pharmacology Division of Nutritional Sciences (N.R.W.), Physiology (M.C.D.B.) and Internal Medicine (J.M.W., A.J., W.B., P.S., V.P.N., D.A.H., A.B., D.L.R., A.D., F.C.D.B.), and Saha Cardiovascular Research Center (N.R.W., M.C.D.B., J.M.W., A.J., P.S., V.P.N., D.A.H., A.B., D.L.R., A.D., F.C.D.B.), and Departments of Statistics and Biostatistics (R.J.C.), University of Kentucky, Lexington; and Foundation Gastroenterology, Nashua, NH (J.W.)
| | - Frederick C De Beer
- From the Departments of Pharmacology Division of Nutritional Sciences (N.R.W.), Physiology (M.C.D.B.) and Internal Medicine (J.M.W., A.J., W.B., P.S., V.P.N., D.A.H., A.B., D.L.R., A.D., F.C.D.B.), and Saha Cardiovascular Research Center (N.R.W., M.C.D.B., J.M.W., A.J., P.S., V.P.N., D.A.H., A.B., D.L.R., A.D., F.C.D.B.), and Departments of Statistics and Biostatistics (R.J.C.), University of Kentucky, Lexington; and Foundation Gastroenterology, Nashua, NH (J.W.)
| |
Collapse
|
35
|
Ardila CM, Guzmán IC. Comparison of serum amyloid A protein and C-reactive protein levels as inflammatory markers in periodontitis. J Periodontal Implant Sci 2015; 45:14-22. [PMID: 25722922 PMCID: PMC4341202 DOI: 10.5051/jpis.2015.45.1.14] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 12/24/2014] [Indexed: 12/20/2022] Open
Abstract
PURPOSE The purpose of this study was to compare serum amyloid A (SAA) protein levels with high-sensitive C-reactive protein (hs-CRP) levels as markers of systemic inflammation in patients with chronic periodontitis. The association of serum titers of antibodies to periodontal microbiota and SAA/hs-CRP levels in periodontitis patients was also studied. METHODS A total of 110 individuals were included in this study. Patients were assessed for levels of hs-CRP and SAA. Nonfasting blood samples were collected from participants at the time of clinical examination. The diagnosis of adipose tissue disorders was made according to previously defined criteria. To determine SAA levels, a sandwich enzyme-linked immunosorbent assay was utilized. Paper points were transferred to a sterile tube to obtain a pool of samples for polymerase chain reaction processing and the identification of Porphyromonas gingivalis, Aggregatibacter actinomycetemcomitans, and Tannerella forsythia. The serum level of IgG1 and IgG2 antibodies to P. gingivalis, A. actinomycetemcomitans, and T. forsythia was also determined. RESULTS SAA and hs-CRP levels were higher in periodontitis patients than in controls (P<0.05). In bivariate analysis, high levels of hs-CRP (>3 mg/L) and SAA (>10 mg/L) were significantly associated with chronic periodontitis (P=0.004). The Spearman correlation analysis between acute-phase proteins showed that SAA positively correlated with hs-CRP (r=0.218, P=0.02). In the adjusted model, chronic periodontitis was associated with high levels of SAA (odds ratio [OR], 5.5; 95% confidence interval [CI], 1.6-18.2; P=0.005) and elevated hs-CRP levels (OR, 6.1, 95% CI, 1.6-23.6; P=0.008). Increased levels of serum IgG2 antibodies to P. gingivalis were associated with high levels of SAA (OR, 3.6; 95% CI, 1.4-8.5; P=0.005) and high concentrations of hs-CRP (OR, 4.3; 95% CI, 1.9-9.8; P<0.001). CONCLUSIONS SAA and hs-CRP concentrations in patients with chronic periodontitis are comparably elevated. High serum titers of antibodies to P. gingivalis and the presence of periodontal disease are independently related to high SAA and hs-CRP levels.
Collapse
|
36
|
Jitpean S, Pettersson A, Höglund OV, Holst BS, Olsson U, Hagman R. Increased concentrations of Serum amyloid A in dogs with sepsis caused by pyometra. BMC Vet Res 2014; 10:273. [PMID: 25430894 PMCID: PMC4247870 DOI: 10.1186/s12917-014-0273-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 11/08/2014] [Indexed: 01/21/2023] Open
Abstract
Background Sepsis is a serious and potentially life-threatening condition and early diagnosis and appropriate treatment is crucial for survival. Pyometra is one of the most common diseases in intact female dogs. The disease often leads to sepsis (systemic inflammatory response syndrome, SIRS, caused by infection). Diagnostic markers for detecting sepsis are gaining increasing interest in veterinary medicine. Acute phase proteins (APPs) such as C-reactive protein (CRP) are useful for detecting systemic inflammation in dogs. Serum amyloid A (SAA) is another major APP in dogs that is not yet as widely used. Albumin is regarded as a negative APP and has earlier been evaluated for prediction of prognosis in septic dogs. The aim of the present study was to determine SAA, CRP and albumin concentrations in dogs with sepsis and pyometra and to evaluate whether these inflammatory markers are associated with length of postoperative hospitalization. Results Thirty-one surgically treated bitches with pyometra were included, whereof 23 septic (SIRS-positive) and eight non-septic (SIRS-negative). Albumin concentrations were analyzed by routine automated methods. SAA and CRP analyses were performed with previously validated commercially available assays (ELISA and immunoturbidimetric). Mean (±SE) serum concentrations of SAA were significantly higher in septic (130.8 ± 8.0 mg/L) compared to non-septic bitches (88.5 ± 12.5 mg/L). Using a cut-off value for SAA of 109.07 mg/L (n = 31 bitches), the sensitivity and specificity for detecting sepsis was 74% and 50%, respectively. Serum albumin concentrations were not significantly different in septic compared to non-septic bitches (mean ± SE, 25 ± 1 g/L and 26 ± 1 g/L, respectively). CRP concentrations were also not significantly different in septic (mean ± SE 225.6 ± 16.0 mg/L) compared to non-septic bitches (mean ± SE, 176.0 ± 27.1 mg/L). None of these inflammatory markers were associated with the outcome as measured by length of hospitalization. Conclusions SAA concentrations were increased in dogs with sepsis induced by pyometra and may be useful as an adjunctive diagnostic marker for sepsis. To evaluate the full potential of SAA as a marker for sepsis also in other diseases, further studies are warranted.
Collapse
Affiliation(s)
- Supranee Jitpean
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, SE-750 07, Sweden.
| | | | | | | | | | | |
Collapse
|
37
|
Serum amyloid A as a marker of persistent inflammation and an indicator of cardiovascular and renal involvement in patients with rheumatoid arthritis. Mediators Inflamm 2014; 2014:793628. [PMID: 25525305 PMCID: PMC4265690 DOI: 10.1155/2014/793628] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Revised: 09/24/2014] [Accepted: 11/13/2014] [Indexed: 02/03/2023] Open
Abstract
OBJECTIVES Rheumatoid arthritis (RA) is a systemic, inflammatory disease. Serum amyloid A (SAA) is an acute-phase protein, involved in pathogenesis of atherosclerosis. The aim of the study was to assess serum concentration of SAA in RA patients, with reference to other inflammatory parameters and markers of extra-articular involvement. METHODS The study population consisted of 140 RA patients, low/moderate disease activity (L/MDA) in 98 (70%) patients and high disease activity (HDA) in 42 (30%). Comprehensive clinical and laboratory assessment was performed with evaluation of electrocardiogram and carotid intima-media thickness. RESULTS The mean SAA concentration [327.0 (263.4) mg/L] was increased highly above the normal value, even in patients with L/MDA. Simultaneously, SAA was significantly higher in patients with HDA versus L/MDA. The mean SAA concentration was significantly higher in patients treated with glucocorticoids, was inversely associated with QTc duration, and was markedly higher in patients with atherosclerotic plaques, emphasizing increased CV risk. SAA was significantly higher in patients with increased cystatin-C level. CONCLUSIONS In RA patients, high serum SAA concentration was strongly associated with activity of the disease and risk of CV and renal involvement. Recurrent assessment of SAA may facilitate searching patients with persistent inflammation and risk of extra-articular complications.
Collapse
|
38
|
A pro-atherogenic HDL profile in coronary heart disease patients: an iTRAQ labelling-based proteomic approach. PLoS One 2014; 9:e98368. [PMID: 24859250 PMCID: PMC4032332 DOI: 10.1371/journal.pone.0098368] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2013] [Accepted: 05/01/2014] [Indexed: 12/15/2022] Open
Abstract
Objectives This study aims to compare the protein composition of high-density lipoprotein (HDL) particles in coronary heart disease (CHD) patients and controls by proteomic methods. Background HDL has been reported to exert pro-atherogenic properties in CHD patients. Accumulating evidence indicates that HDL composition, rather than the HDL-C level, determines its functions. The changes in HDL composition involved in the conversion of anti-atherogenic to pro-atherogenic properties in CHD patients are currently unknown. Methods and Results iTRAQ combined with nanoLC-MS/MS was performed to obtain a differential expression profile of the HDL pooled samples of the male age-matched CHD patients and controls (n = 10/group). Of the 196 proteins identified in the examined HDL, 12 were differentially expressed between the CHD patients and the controls, including five up-regulated proteins and seven down-regulated proteins. Using GO analysis, we determined that the up-regulated proteins were mostly involved in inflammatory reactions, displaying a potential pro-atherogenic profile. In contrast, the down-regulated proteins were mostly involved in lipid metabolism processes, displaying anti-atherogenic properties. To confirm the proteomic results, serum amyloid A (SAA) and apoC-I were selected and quantified by ELISA, in the same population as the proteomic analysis, as well as another independent population (n = 120/group). Consistent with the proteomic results, the amount of SAA was significantly increased, and apoC-I was significantly decreased in the HDL particles of CHD patients compared with those of controls (P<0.05). Conclusions Our study shows that the HDL proteome changes to a pro-atherogenic profile in CHD patients, which might compromise the protective effects of HDL. Proteomic analysis of HDL composition may provide more relevant information regarding their functional properties than steady-state HDL-C levels.
Collapse
|
39
|
Ahlin S, Olsson M, Wilhelmson AS, Skålén K, Borén J, Carlsson LMS, Svensson PA, Sjöholm K. Adipose tissue-derived human serum amyloid a does not affect atherosclerotic lesion area in hSAA1+/-/ApoE-/- mice. PLoS One 2014; 9:e95468. [PMID: 24751653 PMCID: PMC3994058 DOI: 10.1371/journal.pone.0095468] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Accepted: 03/27/2014] [Indexed: 11/18/2022] Open
Abstract
Chronically elevated serum levels of serum amyloid A (SAA) are linked to increased risk of cardiovascular disease. However, whether SAA is directly involved in atherosclerosis development is still not known. The aim of this study was to investigate the effects of adipose tissue-derived human SAA on atherosclerosis in mice. hSAA1+/- transgenic mice (hSAA1 mice) with a specific expression of human SAA1 in adipose tissue were bred with ApoE-deficient mice. The hSAA1 mice and their wild type (wt) littermates were fed normal chow for 35 weeks. At the end of the experiment, the mice were euthanized and blood, gonadal adipose tissue and aortas were collected. Plasma levels of SAA, cholesterol and triglycerides were measured. Atherosclerotic lesion areas were analyzed in the aortic arch, the thoracic aorta and the abdominal aorta in en face preparations of aorta stained with Sudan IV. The human SAA protein was present in plasma from hSAA1 mice but undetectable in wt mice. Similar plasma levels of cholesterol and triglycerides were observed in hSAA1 mice and their wt controls. There were no differences in atherosclerotic lesion areas in any sections of the aorta in hSAA1 mice compared to wt mice. In conclusion, our data suggest that adipose tissue-derived human SAA does not influence atherosclerosis development in mice.
Collapse
Affiliation(s)
- Sofie Ahlin
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Maja Olsson
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Anna S. Wilhelmson
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Kristina Skålén
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Jan Borén
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Lena M. S. Carlsson
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Per-Arne Svensson
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Kajsa Sjöholm
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
40
|
Kristensen LP, Larsen MR, Mickley H, Saaby L, Diederichsen AC, Lambrechtsen J, Rasmussen LM, Overgaard M. Plasma proteome profiling of atherosclerotic disease manifestations reveals elevated levels of the cytoskeletal protein vinculin. J Proteomics 2014; 101:141-53. [DOI: 10.1016/j.jprot.2013.12.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Revised: 12/12/2013] [Accepted: 12/16/2013] [Indexed: 11/16/2022]
|
41
|
Kim JB, Hama S, Hough G, Navab M, Fogelman AM, MacLellan WR, Horwich TB, Fonarow GC. Heart failure is associated with impaired anti-inflammatory and antioxidant properties of high-density lipoproteins. Am J Cardiol 2013; 112:1770-7. [PMID: 24050409 DOI: 10.1016/j.amjcard.2013.07.045] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Revised: 07/26/2013] [Accepted: 07/26/2013] [Indexed: 10/26/2022]
Abstract
Oxidative stress and inflammation are hallmarks of the heart failure (HF) disease state. In the present study, we investigated the inflammatory/anti-inflammatory characteristics of high-density lipoproteins (HDL) in patients with HF. Ninety-six consecutive patients with systolic HF were followed in an advanced HF center, and 21 healthy subjects were recruited. Plasma was tested for HDL inflammatory index (HII) using a monocyte chemotactic activity assay, with HII >1.0 indicating proinflammatory HDL. We found significantly increased inflammatory properties of HDL in patients with HF (median HII 1.56 vs 0.59 in controls; p <0.0001). Serum amyloid A level was markedly elevated and the activity of paraoxonase-1, an HDL antioxidant enzyme, was significantly reduced in patients versus controls. HDL and albumin from patients with HF contained markedly elevated levels of oxidized products of arachidonic and linoleic acids. HDL function improved when plasma was treated in vitro with 4F, an apolipoprotein A-I mimetic peptide (40% reduction in HII, p <0.0001). There was no correlation found between HII level and ejection fraction or New York Heart Association functional class. In conclusion, HDL function is significantly impaired and oxidation products of arachidonic and linoleic acids are markedly elevated in patients with HF compared with non-HF controls.
Collapse
|
42
|
De Beer MC, Wroblewski JM, Noffsinger VP, Rateri DL, Howatt DA, Balakrishnan A, Ji A, Shridas P, Thompson JC, van der Westhuyzen DR, Tannock LR, Daugherty A, Webb NR, De Beer FC. Deficiency of endogenous acute phase serum amyloid A does not affect atherosclerotic lesions in apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol 2013; 34:255-61. [PMID: 24265416 DOI: 10.1161/atvbaha.113.302247] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Although elevated plasma concentrations of serum amyloid A (SAA) are associated strongly with increased risk for atherosclerotic cardiovascular disease in humans, the role of SAA in the pathogenesis of lesion formation remains obscure. Our goal was to determine the impact of SAA deficiency on atherosclerosis in hypercholesterolemic mice. APPROACH AND RESULTS Apolipoprotein E-deficient (apoE(-/-)) mice, either wild type or deficient in both major acute phase SAA isoforms, SAA1.1 and SAA2.1, were fed a normal rodent diet for 50 weeks. Female mice, but not male apoE-/- mice deficient in SAA1.1 and SAA2.1, had a modest increase (22%; P≤0.05) in plasma cholesterol concentrations and a 53% increase in adipose mass compared with apoE-/- mice expressing SAA1.1 and SAA2.1 that did not affect the plasma cytokine levels or the expression of adipose tissue inflammatory markers. SAA deficiency did not affect lipoprotein cholesterol distributions or plasma triglyceride concentrations in either male or female mice. Atherosclerotic lesion areas measured on the intimal surfaces of the arch, thoracic, and abdominal regions were not significantly different between apoE-/- mice deficient in SAA1.1 and SAA2.1 and apoE-/- mice expressing SAA1.1 and SAA2.1 in either sex. To accelerate lesion formation, mice were fed a Western diet for 12 weeks. SAA deficiency had effect neither on diet-induced alterations in plasma cholesterol, triglyceride, or cytokine concentrations nor on aortic atherosclerotic lesion areas in either male or female mice. In addition, SAA deficiency in male mice had no effect on lesion areas or macrophage accumulation in the aortic roots. CONCLUSIONS The absence of endogenous SAA1.1 and 2.1 does not affect atherosclerotic lipid deposition in apolipoprotein E-deficient mice fed either normal or Western diets.
Collapse
Affiliation(s)
- Maria C De Beer
- From the Graduate Center for Nutritional Science (M.C.D.B., J.M.W., V.P.N., A.J., P.S., J.C.T., D.R.v.d.W., L.R.T., N.R.W., F.C.D.B.), Saha Cardiovascular Research Center (M.C.D.B., J.M.W., V.P.N., D.L.R., D.A.H., A.B., A.J., P.S., J.C.T., D.R.v.d.W., L.R.T., A.D., N.R.W., F.C.D.B.), and the Departments of Physiology (M.C.D.B.) and Internal Medicine (J.M.W., V.P.N., D.L.R., D.A.H., A.B., A.J., P.S., J.C.T., D.R.v.d.W., L.R.T., A.D., N.R.W., F.C.D.B.), University of Kentucky Medical Center, Lexington, KY; and Department of Veterans Affairs Medical Center, Lexington, KY (D.R.v.d.W., L.R.T.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Tsun JGS, Shiu SWM, Wong Y, Yung S, Chan TM, Tan KCB. Impact of serum amyloid A on cellular cholesterol efflux to serum in type 2 diabetes mellitus. Atherosclerosis 2013; 231:405-10. [PMID: 24267259 DOI: 10.1016/j.atherosclerosis.2013.10.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Revised: 09/07/2013] [Accepted: 10/10/2013] [Indexed: 10/26/2022]
Abstract
OBJECTIVE Serum amyloid A (SAA) is an acute phase response protein and has apolipoprotein properties. Since type 2 diabetes is associated with chronic subclinical inflammation, the objective of this study is to investigate the changes in SAA level in type 2 diabetic patients and to evaluate the relationship between SAA and the capacity of serum to induce cellular cholesterol efflux via the two known cholesterol transporters, scavenger receptor class B type I (SR-BI) and ATP-binding cassette transporter G1 (ABCG1). METHODS 264 patients with type 2 diabetes mellitus (42% with normoalbuminuria, 30% microalbuminuria, and 28% proteinuria) and 275 non-diabetic controls were recruited. SAA was measured by ELISA. SR-BI and ABCG1-mediated cholesterol efflux to serum were determined by measuring the transfer of [(3)H]cholesterol from Fu5AH rat hepatoma cells expressing SR-BI and from human ABCG1-transfected CHO-K1 cells to the medium containing the tested serum respectively. RESULTS SAA was significantly increased in diabetic patients with incipient or overt nephropathy. Both SR-BI and ABCG1-mediated cholesterol efflux to serum were significantly impaired in all three groups of diabetic patients (p < 0.01). SAA inversely correlated with SR-BI-mediated cholesterol efflux (r = -0.36, p < 0.01) but did not correlate with ABCG1-mediated cholesterol efflux. Stepwise linear regression analysis showed that HDL, the presence or absence of diabetes, and log(SAA) were significant independent determinants of SR-BI-mediated cholesterol efflux to serum. CONCLUSION SAA was increased in type 2 diabetic patients with incipient or overt nephropathy, and SAA was associated with impairment of SR-BI-mediated cholesterol efflux to serum.
Collapse
Affiliation(s)
- J G S Tsun
- Department of Medicine, University of Hong Kong, Queen Mary Hospital, Pokfulam Road, Hong Kong
| | | | | | | | | | | |
Collapse
|
44
|
Yayan J. Emerging families of biomarkers for coronary artery disease: inflammatory mediators. Vasc Health Risk Manag 2013; 9:435-56. [PMID: 23983474 PMCID: PMC3751465 DOI: 10.2147/vhrm.s45704] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Introduction Inflammation has been implicated in the development of atherosclerosis in patients with acute coronary syndrome. C-reactive protein is an established nonspecific prognostic inflammatory biomarker for patients with acute coronary syndrome in the medical literature. This has led to a concerted effort to identify circulating inflammatory biomarkers to facilitate predicting the risk for and diagnosing coronary artery disease in at-risk subjects. The objective of this study was to search after novel inflammatory biomarkers reported as useful for diagnosing coronary artery disease. Methods The PubMed database was searched for reports published from January 1, 2000 to June 30, 2012 of novel circulating biomarkers for coronary artery disease in addition to the established biomarker, C-reactive protein. The search terms used were “infarction”, “biomarkers”, and “markers”, and only original articles describing clinical trials that were written in English were included. All published articles were separately examined carefully after novel inflammatory markers for acute coronary syndrome. All irrelevant publications without content pertaining to inflammatory biomarkers for acute coronary syndrome were excluded from this study. Our results reflect all articles concerning biomarkers in humans. Results The PubMed search yielded 4,415 research articles. After further analysis, all relevant published original articles examining 53 biomarkers were included in this review, which identified 46 inflammation biomarkers useful for detecting coronary artery disease. Conclusion The emergence of diverse novel biomarkers for coronary artery disease has provided insight into the varied pathophysiology of this disease. Inflammatory biomarkers have tremendous potential in aiding the prediction of acute coronary syndrome and recurrent ischemic episodes, and will eventually help improve patient care and management.
Collapse
Affiliation(s)
- Josef Yayan
- Department of internal Medicine, University Hospital of Saarland, Homburg/Saar, Germany.
| |
Collapse
|
45
|
Jahangiri A, Wilson PG, Hou T, Brown A, King VL, Tannock LR. Serum amyloid A is found on ApoB-containing lipoproteins in obese humans with diabetes. Obesity (Silver Spring) 2013; 21:993-6. [PMID: 23784902 PMCID: PMC3695410 DOI: 10.1002/oby.20126] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2011] [Accepted: 09/06/2012] [Indexed: 11/20/2022]
Abstract
OBJECTIVE In murine models of obesity/diabetes, there is an increase in plasma serum amyloid A (SAA) levels along with redistribution of SAA from high-density lipoprotein (HDL) to apolipoprotein B (apoB)-containing lipoprotein particles, namely, low-density lipoprotein and very low-density lipoprotein. The goal of this study was to determine if obesity is associated with similar SAA lipoprotein redistribution in humans. DESIGN AND METHODS Three groups of obese individuals were recruited from a weight loss clinic: healthy obese (n = 14), metabolic syndrome (MetS) obese (n = 8), and obese with type 2 diabetes (n = 6). Plasma was separated into lipoprotein fractions by fast protein liquid chromatography, and SAA was measured in lipid fractions using enzyme-linked immunosorbent assay and Western blotting. RESULTS Only the obese diabetic group had SAA detectable in apoB-containing lipoproteins, and SAA reverted back to HDL with active weight loss. CONCLUSIONS In human subjects, SAA is found in apoB-containing lipoprotein particles only in obese subjects with type 2 diabetes, but not in healthy obese or obese subjects with MetS.
Collapse
Affiliation(s)
- Anisa Jahangiri
- Division of Endocrinology and Molecular Medicine, University of Kentucky, Lexington, KY, USA
| | - Patricia G Wilson
- Division of Endocrinology and Molecular Medicine, University of Kentucky, Lexington, KY, USA
| | - Tianfei Hou
- Graduate Center for Nutritional Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - Aparna Brown
- Division of Endocrinology and Molecular Medicine, University of Kentucky, Lexington, KY, USA
| | - Victoria L. King
- Graduate Center for Nutritional Sciences, University of Kentucky, Lexington, Kentucky, USA
- Division of Cardiovascular Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Lisa R. Tannock
- Division of Endocrinology and Molecular Medicine, University of Kentucky, Lexington, KY, USA
- Graduate Center for Nutritional Sciences, University of Kentucky, Lexington, Kentucky, USA
- Department of Veterans Affairs, Lexington, KY, USA
| |
Collapse
|
46
|
Eren MA, Vural M, Cece H, Camuzcuoglu H, Yildiz S, Toy H, Aksoy N. Association of serum amyloid A with subclinical atherosclerosis in women with gestational diabetes. Gynecol Endocrinol 2012; 28:1010-3. [PMID: 22827403 DOI: 10.3109/09513590.2012.705371] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The aim of our study was to evaluate serum amyloid A (SAA), an acute phase reactant, and carotid intima-media thickness (CIMT) as a valid predictor of atherosclerosis in women with gestational diabetes mellitus (GDM). Serum samples from 39 pregnant women with GDM and 25 healthy pregnant women were collected for the analysis of SAA. CIMT was measured in both groups to evaluate future atherosclerotic heart disease risk. The SAA level was measured with ELISA. The mean arterial blood pressure (MABP), CIMT and SAA levels were significantly higher in women with GDM compared with healthy pregnant controls (p = 0.033, p = 0.001 and p = 0.004, respectively). There were significant correlations between SAA and age, BMI, MABP, 50-g oral glucose tolerance test (OGTT), and A1c (p = 0.048, p = 0.037, p = 0.035, p = 0.042 and p = 0.048, respectively) and between CIMT and BMI, MABP, and 50-g OGTT, (p = 0.001, p = 0.004 and p < 0.001, respectively) in correlation analysis. Furthermore, there was a correlation between SAA and CIMT (p = 0.048). Increased SAA and CIMT values in GDM compared with healthy controls might indicate an increased risk of subclinical atherosclerosis and future atherosclerotic heart disease and the importance of inflammation in this process. These changes were associated with obesity, hypertension and glucose intolerance-related factors (BMI, MABP, and 50-g OGTT), which may be relevant to GDM pathophysiology.
Collapse
Affiliation(s)
- Mehmet Ali Eren
- Department of Endocrinology, School of Medicine, Harran University, Sanliurfa, Turkey.
| | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
PURPOSE OF REVIEW HDL cholesterol concentration is inversely correlated with cardiovascular disease and has a wide range of functions involved in many systems. The purpose of this review is to summarize HDL functionality, its relevance to atherosclerosis and factors affecting HDL functions. RECENT FINDINGS The contribution of HDL to reverse cholesterol transport may not be as great as first envisaged. However, it still plays an important role in cholesterol efflux from peripheral tissues. The capacity of HDL to promote cellular cholesterol efflux in an ex-vivo model has been reported to correlate more closely with carotid intima-media thickness than HDL cholesterol concentration. Recently, a variety of other functions of HDL have been described including antimicrobial, antioxidant, antiglycation, anti-inflammatory, nitric oxide--inducing, antithrombotic and antiatherogenic activity and immune modulation as well as a potential role in glucose homeostasis, diabetes pathophysiology and complications. SUMMARY HDL has a wide range of functions some of which are independent of its cholesterol content. Its cargo of apolipoproteins, various proteins and phospholipids contributes most to its various functions. These functions are affected by a number of genetic, physiological and pathological factors.
Collapse
Affiliation(s)
- Handrean Soran
- University Department of Medicine, Central Manchester and Manchester Children University Hospital NHS Foundation Trust, School of Biomedicine, University of Manchester, Manchester, UK
| | | | | | | |
Collapse
|
48
|
Wan W, Gao JL. Leukocyte chemoattractant receptor FPR2 may accelerate atherogenesis. Med Hypotheses 2012; 79:101-3. [PMID: 22541858 PMCID: PMC3361548 DOI: 10.1016/j.mehy.2012.04.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2012] [Revised: 03/06/2012] [Accepted: 04/03/2012] [Indexed: 01/31/2023]
Abstract
Atherosclerosis is a chronic inflammatory disease and the number one cause of mortality worldwide. The fundamental causes of atherosclerosis have not been precisely delineated, although pathogenesis clearly involves endothelial dysfunction and both innate and adaptive immunity. Recent evidence suggests that formyl peptide receptor 2 (FPR2), a G protein-coupled receptor (GPCR), mediates a range of inflammatory responses including superoxide production in neutrophils, chemotaxis of monocytes and neutrophils, CCL2 production in endothelial cells (ECs) and monocytes, and increased CXCL8 expression in neutrophils, which are all related with atherogenesis. Therefore, we propose that FPR2 may play a pathogenic role in atherogenesis.
Collapse
Affiliation(s)
- Wuzhou Wan
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, United States.
| | | |
Collapse
|
49
|
Ahmed MS, Jadhav AB, Hassan A, Meng QH. Acute phase reactants as novel predictors of cardiovascular disease. ISRN INFLAMMATION 2012; 2012:953461. [PMID: 24049653 PMCID: PMC3767354 DOI: 10.5402/2012/953461] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Accepted: 04/01/2012] [Indexed: 12/21/2022]
Abstract
Acute phase reaction is a systemic response which usually follows a physiological condition that takes place in the beginning of an inflammatory process. This physiological change usually lasts 1-2 days. However, the systemic acute phase response usually lasts longer. The aim of this systemic response is to restore homeostasis. These events are accompanied by upregulation of some proteins (positive acute phase reactants) and downregulation of others (negative acute phase reactants) during inflammatory reactions. Cardiovascular diseases are accompanied by the elevation of several positive acute phase reactants such as C-reactive protein (CRP), serum amyloid A (SAA), fibrinogen, white blood cell count, secretory nonpancreatic phospholipase 2-II (sPLA2-II), ferritin, and ceruloplasmin. Cardiovascular disease is also accompanied by the reduction of negative acute phase reactants such as albumin, transferrin, transthyretin, retinol-binding protein, antithrombin, and transcortin. In this paper, we will be discussing the biological activity and diagnostic and prognostic values of acute phase reactants with cardiovascular importance. The potential therapeutic targets of these reactants will be also discussed.
Collapse
Affiliation(s)
- M S Ahmed
- Department of Medicine, Royal University Hospital, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK, Canada S7N 5E5 ; Department of Pharmacology, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK, Canada S7N 5E5
| | | | | | | |
Collapse
|
50
|
Maly MA, Majek P, Reicheltova Z, Kotlin R, Suttnar J, Oravec M, Veselka J, Dyr JE. Proteomic analysis of plasma samples from acute coronary syndrome patients — The pilot study. Int J Cardiol 2012; 157:126-8. [DOI: 10.1016/j.ijcard.2012.03.045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Accepted: 03/03/2012] [Indexed: 10/28/2022]
|