1
|
Gao A, Xie K, Gupta S, Ahmad G, Witting PK. Cyclic Nitroxide 4-Methoxy-Tempo May Decrease Serum Amyloid A-Mediated Renal Fibrosis and Reorganise Collagen Networks in Aortic Plaque. Int J Mol Sci 2024; 25:7863. [PMID: 39063104 PMCID: PMC11277023 DOI: 10.3390/ijms25147863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/09/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
Acute-phase serum amyloid A (SAA) can disrupt vascular homeostasis and is elevated in subjects with diabetes, cardiovascular disease, and rheumatoid arthritis. Cyclic nitroxides (e.g., Tempo) are a class of piperidines that inhibit oxidative stress and inflammation. This study examined whether 4-methoxy-Tempo (4-MetT) inhibits SAA-mediated vascular and renal dysfunction. Acetylcholine-mediated vascular relaxation and aortic guanosine-3',5'-cyclic monophosphate (cGMP) levels both diminished in the presence of SAA. 4-MetT dose-dependently restored vascular function with corresponding increases in cGMP. Next, male ApoE-deficient mice were administered a vehicle (control, 100 µL PBS) or recombinant SAA (100 µL, 120 µg/mL) ± 4-MetT (at 15 mg/kg body weight via i.p. injection) with the nitroxide administered before (prophylaxis) or after (therapeutic) SAA. Kidney and hearts were harvested at 4 or 16 weeks post SAA administration. Renal inflammation increased 4 weeks after SAA treatment, as judged by the upregulation of IFN-γ and concomitant increases in iNOS, p38MAPK, and matrix metalloproteinase (MMP) activities and increased renal fibrosis (Picrosirius red staining) in the same kidneys. Aortic root lesions assessed at 16 weeks revealed that SAA enhanced lesion size (vs. control; p < 0.05), with plaque presenting with a diffuse fibrous cap (compared to the corresponding aortic root from control and 4-MetT groups). The extent of renal dysfunction and aortic lesion size was largely unchanged in 4-MetT-supplemented mice, although renal fibrosis diminished at 16 weeks, and aortic lesions presented with redistributed collagen networks. These outcomes indicate that SAA stimulates renal dysfunction through promoting the IFN-γ-iNOS-p38MAPK axis, manifesting as renal damage and enhanced atherosclerotic lesions, while supplementation with 4-MetT only affected some of these pathological changes.
Collapse
Affiliation(s)
| | | | | | | | - Paul K. Witting
- Redox Biology Group, Discipline of Pathology, Faculty of Medicine and Health, Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; (A.G.); (K.X.); (S.G.); (G.A.)
| |
Collapse
|
2
|
Kasper R, Rodriguez-Alfonso A, Ständker L, Wiese S, Schneider EM. Major endothelial damage markers identified from hemadsorption filters derived from treated patients with septic shock - endoplasmic reticulum stress and bikunin may play a role. Front Immunol 2024; 15:1359097. [PMID: 38698864 PMCID: PMC11063272 DOI: 10.3389/fimmu.2024.1359097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 04/08/2024] [Indexed: 05/05/2024] Open
Abstract
Introduction In septic patients the damage of the endothelial barrier is decisive leading to circulatory septic shock with disseminated vascular coagulation, edema and multiorgan failure. Hemadsorption therapy leads to rapid resolution of clinical symptoms. We propose that the isolation of proteins adsorbed to hemadsorption devices contributes to the identification of mediators responsible for endothelial barrier dysfunction. Material and methods Plasma materials enriched to hemadsorption filters (CytoSorb®) after therapy of patients in septic shock were fractionated and functionally characterized for their effect on cell integrity, viability, proliferation and ROS formation by human endothelial cells. Fractions were further studied for their contents of oxidized nucleic acids as well as peptides and proteins by mass spectrometry. Results Individual fractions exhibited a strong effect on endothelial cell viability, the endothelial layer morphology, and ROS formation. Fractions with high amounts of DNA and oxidized DNA correlated with ROS formation in the target endothelium. In addition, defined proteins such as defensins (HNP-1), SAA1, CXCL7, and the peptide bikunin were linked to the strongest additive effects in endothelial damage. Conclusion Our results indicate that hemadsorption is efficient to transiently remove strong endothelial damage mediators from the blood of patients with septic shock, which explains a rapid clinical improvement of inflammation and endothelial function. The current work indicates that a combination of stressors leads to the most detrimental effects. Oxidized ssDNA, likely derived from mitochondria, SAA1, the chemokine CXCL7 and the human neutrophil peptide alpha-defensin 1 (HNP-1) were unique for their significant negative effect on endothelial cell viability. However, the strongest damage effect occurred, when, bikunin - cleaved off from alpha-1-microglobulin was present in high relative amounts (>65%) of protein contents in the most active fraction. Thus, a relevant combination of stressors appears to be removed by hemadsorption therapy which results in fulminant and rapid, though only transient, clinical restitution.
Collapse
Affiliation(s)
- Robin Kasper
- Clinic of Anesthesiology and Intensive Care Medicine, University Hospital Ulm, Ulm, Germany
| | - Armando Rodriguez-Alfonso
- Core Facility Functional Peptidomics, Ulm University Medical Center, Ulm, Germany
- Core Unit Mass Spectrometry and Proteomics (CUMP), Ulm University, Ulm, Germany
| | - Ludger Ständker
- Core Facility Functional Peptidomics, Ulm University Medical Center, Ulm, Germany
| | - Sebastian Wiese
- Core Unit Mass Spectrometry and Proteomics (CUMP), Ulm University, Ulm, Germany
| | - E. Marion Schneider
- Clinic of Anesthesiology and Intensive Care Medicine, University Hospital Ulm, Ulm, Germany
| |
Collapse
|
3
|
Xu Z, Wu Y, Fu G, Chen X, Sun J, Tian J, Jiang P, Wang Y, Jin B. SAA1 Has Potential as a Prognostic Biomarker Correlated with Cell Proliferation, Migration, and an Indicator for Immune Infiltration of Tumor Microenvironment in Clear Cell Renal Cell Carcinoma. Int J Mol Sci 2023; 24:ijms24087505. [PMID: 37108666 PMCID: PMC10138873 DOI: 10.3390/ijms24087505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/11/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
The tumor microenvironment (TME) plays an important part in the initiation and development of clear cell renal cell carcinoma (ccRCC). However, an understanding of the immune infiltration in TME is still unknown. Our study aims to explore the correlation between the TME and the clinical features, as well as the prognosis of ccRCC. In the present study, ESTIMATE and CIBERSORT computational methods were applied to calculate the proportion of tumor-infiltrating immune cells (TICs) and the amount of immune and stromal fractions in the ccRCC form The Cancer Genome Atlas (TCGA) database. Then, we sought to find out those immune cell types and genes which may play a significant role and validated them in the GEO database. Furthermore, an immunohistochemical analysis of our external validation dataset was used to detect SAA1 and PDL1 expression in the ccRCC cancer tissues and corresponding normal tissues. Statistical analysis was performed to study the relationship between SAA1 and clinical characteristics, as well as PDL1 expression. Furthermore, a ccRCC cell model with SAA1 knockdown was constructed, which was used for cell proliferation and the migration test. The intersection analysis of the univariate COX and PPI analysis were performed to imply Serum Amyloid A1 (SAA1) as a predictive factor. The expression of SAA1 was significantly negatively correlated to OS and positively correlated to the clinical TMN stage system. The genes in the high-expression SAA1 group were basically enriched in immune-related activities. The proportion of mast cells resting was negatively correlated with SAA1 expression, indicating that SAA1 may be involved in the maintenance of the immune status for the TME. Moreover, the PDL1 expression was positively related to the SAA1 expression and negatively correlated with the patients' prognosis. Further experiments revealed that the knockdown of SAA1 inhibited ccRCC development through suppressing cell proliferation and migration. SAA1 may be a novel marker for the prognosis prediction of ccRCC patients and may play a vital role in the TME by mast cell resting and PDL1 expression. SAA1 has the potential to become a therapeutic target and indicator for immune target therapy in ccRCC treatment.
Collapse
Affiliation(s)
- Zhijie Xu
- Department of Urology, School of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou 310009, China
- Zhejiang Engineering Research Center for Urinary Bladder Carcinoma Innovation Diagnosis and Treatment, Hangzhou 310024, China
| | - Yunfei Wu
- Department of Urology, School of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou 310009, China
- Zhejiang Engineering Research Center for Urinary Bladder Carcinoma Innovation Diagnosis and Treatment, Hangzhou 310024, China
| | - Guanghou Fu
- Department of Urology, School of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou 310009, China
- Zhejiang Engineering Research Center for Urinary Bladder Carcinoma Innovation Diagnosis and Treatment, Hangzhou 310024, China
| | - Xiaoyi Chen
- Department of Urology, School of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou 310009, China
- Zhejiang Engineering Research Center for Urinary Bladder Carcinoma Innovation Diagnosis and Treatment, Hangzhou 310024, China
| | - Junjie Sun
- Department of Urology, School of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou 310009, China
- Zhejiang Engineering Research Center for Urinary Bladder Carcinoma Innovation Diagnosis and Treatment, Hangzhou 310024, China
| | - Junjie Tian
- Department of Urology, School of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou 310009, China
- Zhejiang Engineering Research Center for Urinary Bladder Carcinoma Innovation Diagnosis and Treatment, Hangzhou 310024, China
| | - Peng Jiang
- Department of Urology, School of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou 310009, China
- Zhejiang Engineering Research Center for Urinary Bladder Carcinoma Innovation Diagnosis and Treatment, Hangzhou 310024, China
| | - Yimin Wang
- Department of Urology, School of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou 310009, China
- Zhejiang Engineering Research Center for Urinary Bladder Carcinoma Innovation Diagnosis and Treatment, Hangzhou 310024, China
| | - Baiye Jin
- Department of Urology, School of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou 310009, China
- Zhejiang Engineering Research Center for Urinary Bladder Carcinoma Innovation Diagnosis and Treatment, Hangzhou 310024, China
| |
Collapse
|
4
|
Wu R, Jiang W, Sun Y, Wu L, Di Y, Wang J, Zhong S, Wang W. Indicators of Oxidative Stress in the Prediction of Coronary Artery Lesions in Patients With Kawasaki Disease. J Clin Rheumatol 2023; 29:126-131. [PMID: 36730421 DOI: 10.1097/rhu.0000000000001925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVES The aim of this study was to examine the clinical significance of oxidative stress (OS)-related indices, including inflammatory markers and lipid and platelet (PLT) parameter, in coronary artery lesions (CALs) in Kawasaki disease (KD). METHODS Clinical data of 952 KD patients diagnosed between January 2019 and March 2022 were collected and divided into CAL and NCAL groups. All the KD patients were randomly divided into training set and verification set. The univariate analysis and multivariate logistic regression analysis of training set were used to identify the OS-related independent risk factors of CALs, which were then used to construct a predictive nomogram. Calibration curve and receiver operating characteristic curve were used to evaluate the performance of the model. The predictive nomogram was further validated on verification set. RESULTS In the training set, 137 KD patients (18.0%) showed CALs. C-reactive protein, serum amyloid A, PLT count, monocyte-to-high-density lipoprotein (HDL) ratio, and PLT-to-lymphocyte ratio were significantly higher, whereas HDL was lower in the CAL group than the NCAL group. Increased C-reactive protein, serum amyloid A, PLT, and decreased HDL were identified as independent risk factors. The nomogram constructed using these factors showed satisfactory calibration degree and discriminatory power (the area under the curve, 0.887). In the verification set, the area under the curve was 0.795. CONCLUSION The predictive nomogram constructed using 4 OS-related risk factors associated with CALs in patients with KD could be a useful tool for early diagnosis of CALs in KD.
Collapse
Affiliation(s)
- Rouyi Wu
- From the School of Medicine, Ningbo University
| | - Wei Jiang
- Department of Pediatric Rheumatoid Immunity, Ningbo Women and Children's Hospital, Ningbo, Zhejiang, China
| | - Yangkai Sun
- From the School of Medicine, Ningbo University
| | - Ling Wu
- Department of Pediatric Rheumatoid Immunity, Ningbo Women and Children's Hospital, Ningbo, Zhejiang, China
| | - Yazhen Di
- Department of Pediatric Rheumatoid Immunity, Ningbo Women and Children's Hospital, Ningbo, Zhejiang, China
| | - Jiapei Wang
- Department of Pediatric Rheumatoid Immunity, Ningbo Women and Children's Hospital, Ningbo, Zhejiang, China
| | - Shiling Zhong
- Department of Pediatric Rheumatoid Immunity, Ningbo Women and Children's Hospital, Ningbo, Zhejiang, China
| | - Wenbo Wang
- From the School of Medicine, Ningbo University
| |
Collapse
|
5
|
Avdeeva AS. Inflammatory markers in rheumatic diseases. RHEUMATOLOGY SCIENCE AND PRACTICE 2022. [DOI: 10.47360/1995-4484-2022-561-569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Immune-mediated rheumatic diseases (IMRDs) are a broad group of pathological conditions based on impaired immunological tolerance to one’s own tissues leading to inflammation and irreversible organ damage. Laboratory diagnosis of IMRDs includes a wide range of biomarkers (autoantibodies, acute phase proteins, cytokines, markers of endothelial damage, components of the complement system, immunoglobulins, cryoglobulins, lymphocyte subpopulations, indicators of bone metabolism, apoptosis markers, genetic markers, etc). One of the leading aspects of laboratory diagnosis of IMRDs is the study of the level of inflammation markers in the blood (erythrocyte sedimentation rate, C-reactive protein (CRP), serum amyloid protein (CAA), ferritin, procalcitonin, apolipoprotein AI, calprotectin, etc). The analysis of inflammation markers makes it possible to assess the disease activity, the nature of the progression and the prognosis of the outcomes of a chronic inflammatory process, as well as the effectiveness of the therapy. The review presents the latest data on the role of the most frequently studied inflammatory markers such as CRP, CAA and ferritin.
Collapse
|
6
|
Qu R, He K, Yang Y, Fan T, Sun B, Khan AU, Huang W, Ouyang J, Pan X, Dai J. The role of serum amyloid A1 in the adipogenic differentiation of human adipose-derived stem cells basing on single-cell RNA sequencing analysis. Stem Cell Res Ther 2022; 13:187. [PMID: 35525990 PMCID: PMC9080218 DOI: 10.1186/s13287-022-02873-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 03/11/2022] [Indexed: 11/12/2022] Open
Abstract
Background Adipose-derived stem cells (ASCs) are obtained from a variety of sources in vivo where they present in large quantities. These cells are suitable for use in autologous transplantation and the construction of tissue-engineered adipose tissue. Studies have shown that ASCs differentiation is in a high degree of heterogeneity, yet the molecular basis including key regulators of differentiation remains to clarify. Methods We performed single-cell RNA sequencing and bioinformatics analysis on both undifferentiated (ASC-GM group) and adipogenically differentiated human ASCs (ASC-AD group, ASCs were cultured in adipogenic inducing medium for 1 week). And then, we verified the results of serum amyloid A1 (SAA1) with western blotting, immunofluorescence staining, oil red O staining. After these experiments, we down-regulated the expression of serum amyloid A1 (SAA1) gene to verify the adipogenic differentiation ability of ASCs.
Results In single-cell RNA sequence analyzing, we obtained 4415 cells in the ASC-GM group and 4634 cells in the ASC-AD group. The integrated sample cells could be divided into 11 subgroups (0–10 cluster). The cells in cluster 0, 2, 5 were came from ASC-GM group and the cells in cluster 1, 3, 7 came from ASC-AD group. The cells of cluster 4 and 6 came from both ASC-GM and ASC-AD groups. Fatty acid binding protein 4, fatty acid binding protein 5, complement factor D, fatty acid desaturase 1, and insulin like growth factor binding protein 5 were high expressed in category 1 and 7. Regulation of inflammatory response is the rank 1 biological processes. And cellular responses to external stimuli, negative regulation of defense response and acute inflammatory response are included in top 20 biological processes. Based on the MCODE results, we found that SAA1, C-C Motif Chemokine Ligand 5 (CCL5), and Annexin A1 (ANXA1) significantly highly expressed during adipogenic differentiation. Western blot and immunofluorescent staining results showed that SAA1 increased during adipogenesis. And the area of ORO positive staining in siSAA1 cells was significantly lower than in the siControl (negative control) cells. Conclusions Our results also indicated that our adipogenic induction was successful, and there was great heterogeneity in the adipogenic differentiation of ASCs. SAA1 with the regulation of inflammatory response were involved in adipogenesis of ASCs based on single-cell RNA sequencing analysis. The data obtained will help to elucidate the intrinsic mechanism of heterogeneity in the differentiation process of stem cells, thus, guiding the regulation of self-renewal and differentiation of adult stem cells. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-02873-5.
Collapse
Affiliation(s)
- Rongmei Qu
- Guangdong Provincial Key Laboratory of Medical Biomechanics and Guangdong Engineering Research Center for Translation of Medical 3D Printing Application and National Key Discipline of Human Anatomy, School of Basic Medical Science, Southern Medical University and National Demonstration Center for Experimental Education of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Kai He
- Guangdong Provincial Key Lab of Single Cell Technology and Application, and Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Yuchao Yang
- Guangdong Provincial Key Laboratory of Medical Biomechanics and Guangdong Engineering Research Center for Translation of Medical 3D Printing Application and National Key Discipline of Human Anatomy, School of Basic Medical Science, Southern Medical University and National Demonstration Center for Experimental Education of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Tingyu Fan
- Guangdong Provincial Key Laboratory of Medical Biomechanics and Guangdong Engineering Research Center for Translation of Medical 3D Printing Application and National Key Discipline of Human Anatomy, School of Basic Medical Science, Southern Medical University and National Demonstration Center for Experimental Education of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Bing Sun
- Guangdong Provincial Key Laboratory of Medical Biomechanics and Guangdong Engineering Research Center for Translation of Medical 3D Printing Application and National Key Discipline of Human Anatomy, School of Basic Medical Science, Southern Medical University and National Demonstration Center for Experimental Education of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Asmat Ullah Khan
- Guangdong Provincial Key Laboratory of Medical Biomechanics and Guangdong Engineering Research Center for Translation of Medical 3D Printing Application and National Key Discipline of Human Anatomy, School of Basic Medical Science, Southern Medical University and National Demonstration Center for Experimental Education of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Wenhua Huang
- Guangdong Provincial Key Laboratory of Medical Biomechanics and Guangdong Engineering Research Center for Translation of Medical 3D Printing Application and National Key Discipline of Human Anatomy, School of Basic Medical Science, Southern Medical University and National Demonstration Center for Experimental Education of Basic Medical Sciences, Southern Medical University, Guangzhou, China.
| | - Jun Ouyang
- Guangdong Provincial Key Laboratory of Medical Biomechanics and Guangdong Engineering Research Center for Translation of Medical 3D Printing Application and National Key Discipline of Human Anatomy, School of Basic Medical Science, Southern Medical University and National Demonstration Center for Experimental Education of Basic Medical Sciences, Southern Medical University, Guangzhou, China.
| | - Xinghua Pan
- Guangdong Provincial Key Lab of Single Cell Technology and Application, and Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Southern Medical University, Guangzhou, China.
| | - Jingxing Dai
- Guangdong Provincial Key Laboratory of Medical Biomechanics and Guangdong Engineering Research Center for Translation of Medical 3D Printing Application and National Key Discipline of Human Anatomy, School of Basic Medical Science, Southern Medical University and National Demonstration Center for Experimental Education of Basic Medical Sciences, Southern Medical University, Guangzhou, China.
| |
Collapse
|
7
|
Martin NJ, Chami B, Vallejo A, Mojadadi AA, Witting PK, Ahmad G. Efficacy of the Piperidine Nitroxide 4-MethoxyTEMPO in Ameliorating Serum Amyloid A-Mediated Vascular Inflammation. Int J Mol Sci 2021; 22:ijms22094549. [PMID: 33925294 PMCID: PMC8123591 DOI: 10.3390/ijms22094549] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 01/24/2023] Open
Abstract
Intracellular redox imbalance in endothelial cells (EC) can lead to endothelial dysfunction, which underpins cardiovascular diseases (CVD). The acute phase serum amyloid A (SAA) elicits inflammation through stimulating production of reactive oxygen species (ROS). The cyclic nitroxide 4-MethoxyTEMPO (4-MetT) is a superoxide dismutase mimetic that suppresses oxidant formation and inflammation. The aim of this study was to investigate whether 4-MetT inhibits SAA-mediated activation of cultured primary human aortic EC (HAEC). Co-incubating cells with 4-MetT inhibited SAA-mediated increases in adhesion molecules (VCAM-1, ICAM-1, E-selectin, and JAM-C). Pre-treatment of cells with 4-MetT mitigated SAA-mediated increases in transcriptionally activated NF-κB-p65 and P120 Catenin (a stabilizer of Cadherin expression). Mitochondrial respiration and ROS generation (mtROS) were adversely affected by SAA with decreased respiratory reserve capacity, elevated maximal respiration and proton leakage all characteristic of SAA-treated HAEC. This altered respiration manifested as a loss of mitochondrial membrane potential (confirmed by a decrease in TMRM fluorescence), and increased mtROS production as assessed with MitoSox Red. These SAA-linked impacts on mitochondria were mitigated by 4-MetT resulting in restoration of HAEC nitric oxide bioavailability as confirmed by assessing cyclic guanosine monophosphate (cGMP) levels. Thus, 4-MetT ameliorates SAA-mediated endothelial dysfunction through normalising EC redox homeostasis. Subject to further validation in in vivo settings; these outcomes suggest its potential as a therapeutic in the setting of cardiovascular pathologies where elevated SAA and endothelial dysfunction is linked to enhanced CVD.
Collapse
|
8
|
Sorić Hosman I, Kos I, Lamot L. Serum Amyloid A in Inflammatory Rheumatic Diseases: A Compendious Review of a Renowned Biomarker. Front Immunol 2021; 11:631299. [PMID: 33679725 PMCID: PMC7933664 DOI: 10.3389/fimmu.2020.631299] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 12/31/2020] [Indexed: 12/13/2022] Open
Abstract
Serum amyloid A (SAA) is an acute phase protein with a significant importance for patients with inflammatory rheumatic diseases (IRD). The central role of SAA in pathogenesis of IRD has been confirmed by recent discoveries, including its involvement in the activation of the inflammasome cascade and recruitment of interleukin 17 producing T helper cells. Clinical utility of SAA in IRD was originally evaluated nearly half a century ago. From the first findings, it was clear that SAA could be used for evaluating disease severity and monitoring disease activity in patients with rheumatoid arthritis and secondary amyloidosis. However, cost-effective and more easily applicable markers, such as C-reactive protein (CRP) and erythrocyte sedimentation rate (ESR), overwhelmed its use in clinical practice. In the light of emerging evidences, SAA has been discerned as a more sensitive biomarker in a wide spectrum of IRD, especially in case of subclinical inflammation. Furthermore, a growing number of studies are confirming the advantages of SAA over many other biomarkers in predicting and monitoring response to biological immunotherapy in IRD patients. Arising scientific discoveries regarding the role of SAA, as well as delineating SAA and its isoforms as the most sensitive biomarkers in various IRD by recently developing proteomic techniques are encouraging the revival of its clinical use. Finally, the most recent findings have shown that SAA is a biomarker of severe Coronavirus disease 2019 (COVID-19). The aim of this review is to discuss the SAA-involving immune system network with emphasis on mechanisms relevant for IRD, as well as usefulness of SAA as a biomarker in various IRD. Therefore, over a hundred original papers were collected through an extensive PubMed and Scopus databases search. These recently arising insights will hopefully lead to a better management of IRD patients and might even inspire the development of new therapeutic strategies with SAA as a target.
Collapse
Affiliation(s)
- Iva Sorić Hosman
- Department of Pediatrics, Zadar General Hospital, Zadar, Croatia
| | - Ivanka Kos
- Division of Nephrology, Dialysis and Transplantation, Department of Pediatrics, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Lovro Lamot
- Division of Nephrology, Dialysis and Transplantation, Department of Pediatrics, University Hospital Centre Zagreb, Zagreb, Croatia.,Department of Pediatrics, University of Zagreb School of Medicine, Zagreb, Croatia
| |
Collapse
|
9
|
Abouelasrar Salama S, De Bondt M, De Buck M, Berghmans N, Proost P, Oliveira VLS, Amaral FA, Gouwy M, Van Damme J, Struyf S. Serum Amyloid A1 (SAA1) Revisited: Restricted Leukocyte-Activating Properties of Homogeneous SAA1. Front Immunol 2020; 11:843. [PMID: 32477346 PMCID: PMC7240019 DOI: 10.3389/fimmu.2020.00843] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 04/14/2020] [Indexed: 12/11/2022] Open
Abstract
Infection, sterile injury, and chronic inflammation trigger the acute phase response in order to re-establish homeostasis. This response includes production of positive acute phase proteins in the liver, such as members of the serum amyloid A (SAA) family. In humans the major acute phase SAAs comprise a group of closely related variants of SAA1 and SAA2. SAA1 was proven to be chemotactic for several leukocyte subtypes through activation of the G protein-coupled receptor FPRL1/FPR2. Several other biological activities of SAA1, such as cytokine induction, reported to be mediated via TLRs, have been debated recently. Especially commercial SAA1, recombinantly produced in Escherichia coli, was found to be contaminated with bacterial products confounding biological assays performed with this rSAA1. We purified rSAA1 by RP-HPLC to homogeneity, removing contaminants such as lipopolysaccharides, lipoproteins and formylated peptides, and re-assessed several biological activities attributed to SAA1 (chemotaxis, cytokine induction, MMP-9 release, ROS generation, and macrophage differentiation). The homogeneous rSAA1 (hrSAA1) lacked most cell-activating properties, but its leukocyte-recruiting capacity in vivo and it’s in vitro synergy with other leukocyte attractants remained preserved. Furthermore, hrSAA1 maintained the ability to promote monocyte survival. This indicates that pure hrSAA1 retains its potential to activate FPR2, whereas TLR-mediated effects seem to be related to traces of bacterial TLR ligands in the E. coli-produced human rSAA1.
Collapse
Affiliation(s)
- Sara Abouelasrar Salama
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Mirre De Bondt
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Mieke De Buck
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Nele Berghmans
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Paul Proost
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Vivian Louise Soares Oliveira
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium.,Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Flavio A Amaral
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mieke Gouwy
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Jo Van Damme
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Sofie Struyf
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| |
Collapse
|
10
|
Kuret T, Sodin-Šemrl S, Mrak-Poljšak K, Čučnik S, Lakota K, Erman A. Interleukin-1β Induces Intracellular Serum Amyloid A1 Expression in Human Coronary Artery Endothelial Cells and Promotes its Intercellular Exchange. Inflammation 2020; 42:1413-1425. [PMID: 31011929 DOI: 10.1007/s10753-019-01003-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Serum amyloid A (SAA) is an acute-phase protein with important, pathogenic role in the development of atherosclerosis. Since dysfunctional endothelium represents a key early step in atherogenesis, we aimed to determine whether induced human coronary artery endothelial cells (HCAEC) modulate SAA1/2/4 expression and influence intracellular location and intercellular transport of SAA1. HCAEC were stimulated with 1 ng/ml IL-1β, 10 ng/ml IL-6, and/or 1 μM dexamethasone for 24 h. QPCR, Western blots, ELISA, and immunofluorescent labeling were performed for detection of SAA1/2/4 mRNA and protein levels, respectively. In SAA1 transport experiments, FITC- or Cy3-labeled SAA1 were added to HCAEC separately, for 24 h, followed by a combined incubation of SAA1-FITC and SAA1-Cy3 positive cells, with IL-1β and analysis by flow cytometry. IL-1β upregulated SAA1 (119.9-fold, p < 0.01) and SAA2 (9.3-fold; p < 0.05) mRNA expression levels, while mRNA expression of SAA4 was not affected. Intracellular SAA1 was found mainly as a monomer, while SAA2 and SAA4 formed octamers as analyzed by Western blots. Within HCAEC, SAA1/2/4 located mostly to the perinuclear area and tunneling membrane nanotubes. Co-culturing of SAA1-FITC and SAA1-Cy3 positive cells for 48 h showed a significantly higher percentage of double positive cells in IL-1β-stimulated (mean ± SD; 60 ± 4%) vs. non-stimulated cells (48 ± 2%; p < 0.05). IL-1β induces SAA1 expression in HCAEC and promotes its intercellular exchange, suggesting that direct communication between cells in inflammatory conditions could ultimately lead to faster development of atherosclerosis in coronary arteries.
Collapse
Affiliation(s)
- Tadeja Kuret
- Department of Rheumatology, University Medical Centre Ljubljana, Vodnikova 62, SI-1000, Ljubljana, Slovenia.
- Faculty of Pharmacy, Chair of Clinical Biochemistry, University of Ljubljana, Aškerčeva 7, SI-1000, Ljubljana, Slovenia.
| | - Snežna Sodin-Šemrl
- Department of Rheumatology, University Medical Centre Ljubljana, Vodnikova 62, SI-1000, Ljubljana, Slovenia
- Faculty of Mathematics, Natural Sciences and Information Technologies, University of Primorska, Glagoljaška 8, SI-6000, Koper, Slovenia
| | - Katjuša Mrak-Poljšak
- Department of Rheumatology, University Medical Centre Ljubljana, Vodnikova 62, SI-1000, Ljubljana, Slovenia
| | - Saša Čučnik
- Department of Rheumatology, University Medical Centre Ljubljana, Vodnikova 62, SI-1000, Ljubljana, Slovenia
- Faculty of Pharmacy, Chair of Clinical Biochemistry, University of Ljubljana, Aškerčeva 7, SI-1000, Ljubljana, Slovenia
| | - Katja Lakota
- Department of Rheumatology, University Medical Centre Ljubljana, Vodnikova 62, SI-1000, Ljubljana, Slovenia
- Faculty of Mathematics, Natural Sciences and Information Technologies, University of Primorska, Glagoljaška 8, SI-6000, Koper, Slovenia
| | - Andreja Erman
- Faculty of Medicine, Institute of Cell Biology, University of Ljubljana, Vrazov trg 2, SI-1000, Ljubljana, Slovenia
| |
Collapse
|
11
|
Bioassay for Endothelial Damage Mediators Retrieved by Hemoadsorption. Sci Rep 2019; 9:14522. [PMID: 31601835 PMCID: PMC6787199 DOI: 10.1038/s41598-019-50517-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 09/05/2019] [Indexed: 12/13/2022] Open
Abstract
Hemoadsorption devices are used to treat septic shock by adsorbing inflammatory cytokines and as yet incompletely defined danger and pathogen associated molecular patterns. In an ideal case, hemoadsorption results in immediate recovery of microvascular endothelial cells’ (mEC) function and rapid recovery from catecholamine-dependency and septic shock. We here tested a single device, which consists of polystyrene-divinylbenzene core particles of 450 μm diameter with a high affinity for hydrophobic compounds. The current study aimed at the proof of concept that endothelial-specific damage mediators are adsorbed and can be recovered from hemoadsorption devices. Because of excellent clinical experience, we tested protein fractions released from a hemoadsorber in a novel endothelial bioassay. Video-based, long-term imaging of mEC proliferation and cell death were evaluated and combined with apoptosis and ATP measurements. Out of a total of 39 fractions recovered from column fractionation, we identified 3 fractions that caused i) inhibition of mEC proliferation, ii) increased cell death and iii) induction of apoptosis in mEC. When adding these 3 fractions to mEC, their ATP contents were reduced. These fractions contained proteins of approximately 15 kDa, and high amounts of nucleic acid, which was at least in part oxidized. The efficacy for endothelial cell damage prevention by hemoadsorption can be addressed by a novel endothelial bioassay and long-term video observation procedures. Protein fractionation of the hemoadsorption devices used is feasible to study and define endothelial damage ligands on a molecular level. The results suggest a significant effect by circulating nucleic acids – bound to an as yet undefined protein, which may constitute a major danger-associated molecular pattern (DAMP) in the exacerbation of inflammation when patients experience septic shock. Hemoadsorption devices may thus limit endothelial damage, through the binding of nucleic acid-bearing aggregates and thus contribute to improved endothelial barrier function.
Collapse
|
12
|
Chami B, Hossain F, Hambly TW, Cai X, Aran R, Fong G, Vellajo A, Martin NJJ, Wang X, Dennis JM, Sharma A, Shihata WA, Chin-Dusting JPF, de Haan JB, Sharland A, Geczy CL, Freedman B, Witting PK. Serum Amyloid A Stimulates Vascular and Renal Dysfunction in Apolipoprotein E-Deficient Mice Fed a Normal Chow Diet. Front Immunol 2019; 10:380. [PMID: 30899260 PMCID: PMC6416175 DOI: 10.3389/fimmu.2019.00380] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 02/14/2019] [Indexed: 12/19/2022] Open
Abstract
Elevated serum amyloid A (SAA) levels may promote endothelial dysfunction, which is linked to cardiovascular and renal pathologies. We investigated the effect of SAA on vascular and renal function in apolipoprotein E-deficient (ApoE−/−) mice. Male ApoE−/− mice received vehicle (control), low-level lipopolysaccharide (LPS), or recombinant human SAA by i.p. injection every third day for 2 weeks. Heart, aorta and kidney were harvested between 3 days and 18 weeks after treatment. SAA administration increased vascular cell adhesion molecule (VCAM)-1 expression and circulating monocyte chemotactic protein (MCP)-1 and decreased aortic cyclic guanosine monophosphate (cGMP), consistent with SAA inhibiting nitric oxide bioactivity. In addition, binding of labeled leukocytes to excised aorta increased as monitored using an ex vivo leukocyte adhesion assay. Renal injury was evident 4 weeks after commencement of SAA treatment, manifesting as increased plasma urea, urinary protein, oxidized lipids, urinary kidney injury molecule (KIM)-1 and multiple cytokines and chemokines in kidney tissue, relative to controls. Phosphorylation of nuclear-factor-kappa-beta (NFκB-p-P65), tissue factor (TF), and macrophage recruitment increased in kidneys from ApoE−/− mice 4 weeks after SAA treatment, confirming that SAA elicited a pro-inflammatory and pro-thrombotic phenotype. These data indicate that SAA impairs endothelial and renal function in ApoE−/− mice in the absence of a high-fat diet.
Collapse
Affiliation(s)
- Belal Chami
- Discipline of Pathology, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Farjaneh Hossain
- Discipline of Pathology, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Thomas W Hambly
- Discipline of Pathology, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Xiaoping Cai
- Discipline of Pathology, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Roshanak Aran
- Discipline of Pathology, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Genevieve Fong
- Discipline of Pathology, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Abigail Vellajo
- Discipline of Pathology, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Nathan J J Martin
- Discipline of Pathology, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - XiaoSuo Wang
- Discipline of Pathology, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Joanne M Dennis
- Discipline of Pathology, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Arpeeta Sharma
- Department of Medicine, Monash University, Melbourne, VIC, Australia
| | - Waled A Shihata
- Cardiovascular Disease Program, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia.,Department of Pharmacology, Monash University, Melbourne, VIC, Australia.,Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Jaye P F Chin-Dusting
- Cardiovascular Disease Program, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia.,Department of Pharmacology, Monash University, Melbourne, VIC, Australia.,Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Judy B de Haan
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Alexandra Sharland
- Transplantation Immunobiology Group, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Carolyn L Geczy
- School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Ben Freedman
- ANZAC Research and Heart Research Institutes, Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Paul K Witting
- Discipline of Pathology, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
13
|
Silva Figueiredo P, Inada AC, Ribeiro Fernandes M, Granja Arakaki D, Freitas KDC, Avellaneda Guimarães RDC, Aragão do Nascimento V, Aiko Hiane P. An Overview of Novel Dietary Supplements and Food Ingredients in Patients with Metabolic Syndrome and Non-Alcoholic Fatty Liver Disease. Molecules 2018; 23:molecules23040877. [PMID: 29641459 PMCID: PMC6017470 DOI: 10.3390/molecules23040877] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 03/29/2018] [Accepted: 04/05/2018] [Indexed: 12/13/2022] Open
Abstract
Metabolic syndrome (MetS) is characterized by interconnected factors related to metabolic disturbances, and is directly related to the occurrence of some diseases such as cardiovascular diseases and type 2 diabetes. MetS is described as one or both of insulin resistance and visceral adiposity, considered the initial causes of abnormalities that include hyperglycemia, elevated blood pressure, dyslipidemia, elevated inflammatory markers, and prothrombotic state, as well as polycystic ovarian syndrome in women. Other than in MetS, visceral adiposity and the pro-inflammatory state are also key in the development of non-alcoholic fatty liver disease (NAFLD), which is the most prevalent chronic liver disease in modern society. Both MetS and NAFLD are related to diet and lifestyle, and their treatment may be influenced by dietary pattern changes and the use of certain dietary supplements. This study aimed to review the role of food ingredients and supplements in the management of MetS and NAFLD specifically in human clinical trials. Moreover, bioactive compounds and polyunsaturated fatty acids (PUFAs) may be used as strategies for preventing the onset of and treatment of metabolic disorders, such as MetS and NAFLD, improving the inflammatory state and other comorbidities, such as obesity, dyslipidemias, and cardiovascular diseases (CVD).
Collapse
Affiliation(s)
- Priscila Silva Figueiredo
- Post Graduate Program in Health and Development in the Central-West Region, Federal University of Mato Grosso do Sul, Campo Grande 79070900, Brazil.
| | - Aline Carla Inada
- Post Graduate Program in Health and Development in the Central-West Region, Federal University of Mato Grosso do Sul, Campo Grande 79070900, Brazil.
| | - Melina Ribeiro Fernandes
- Post Graduate Program in Health and Development in the Central-West Region, Federal University of Mato Grosso do Sul, Campo Grande 79070900, Brazil.
| | - Daniela Granja Arakaki
- Post Graduate Program in Health and Development in the Central-West Region, Federal University of Mato Grosso do Sul, Campo Grande 79070900, Brazil.
| | - Karine de Cássia Freitas
- Post Graduate Program in Health and Development in the Central-West Region, Federal University of Mato Grosso do Sul, Campo Grande 79070900, Brazil.
| | - Rita de Cássia Avellaneda Guimarães
- Post Graduate Program in Health and Development in the Central-West Region, Federal University of Mato Grosso do Sul, Campo Grande 79070900, Brazil.
- Optics and Photonics Group, Institute of Physics, Federal University of Mato Grosso do Sul, 549, Campo Grande 79070900, Brazil.
| | - Valter Aragão do Nascimento
- Post Graduate Program in Health and Development in the Central-West Region, Federal University of Mato Grosso do Sul, Campo Grande 79070900, Brazil.
- Optics and Photonics Group, Institute of Physics, Federal University of Mato Grosso do Sul, 549, Campo Grande 79070900, Brazil.
| | - Priscila Aiko Hiane
- Post Graduate Program in Health and Development in the Central-West Region, Federal University of Mato Grosso do Sul, Campo Grande 79070900, Brazil.
| |
Collapse
|
14
|
Huang M, Wei R, Wang Y, Su T, Li P, Chen X. The uremic toxin hippurate promotes endothelial dysfunction via the activation of Drp1-mediated mitochondrial fission. Redox Biol 2018; 16:303-313. [PMID: 29573704 PMCID: PMC5953222 DOI: 10.1016/j.redox.2018.03.010] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 03/14/2018] [Accepted: 03/15/2018] [Indexed: 12/31/2022] Open
Abstract
The accumulation of uremic toxins in chronic kidney disease (CKD) induces inflammation, oxidative stress and endothelial dysfunction, which is a key step in atherosclerosis. Accumulating evidence indicates increased mitochondrial fission is a contributing mechanism for impaired endothelial function. Hippurate, a uremic toxin, has been reported to be involved in cardiovascular diseases. Here, we assessed the endothelial toxicity of hippurate and the contribution of altered mitochondrial dynamics to hippurate-induced endothelial dysfunction. Treatment of human aortic endothelial cells with hippurate reduced the expression of endothelial nitric oxide synthase (eNOS) and increased the expression of intercellular cell adhesion molecule-1 (ICAM-1) and von Willebrand factor (vWF). The mechanisms of hippurate-induced endothelial dysfunction in vitro depended on the activation of Dynamin-related protein 1 (Drp1)-mediated mitochondrial fission and overproduction of mitochondrial reactive oxygen species (mitoROS). In a rat model in which CKD was induced by 5/6 nephrectomy (CKD rat), we observed increased oxidative stress, impaired endothelium-dependent vasodilation, and elevated soluble biomarkers of endothelial dysfunction (ICAM-1 and vWF). Similarly, endothelial dysfunction was identified in healthy rats treated with disease-relevant concentrations of hippurate. In aortas of CKD rats and hippurate-treated rats, we observed an increase in Drp1 protein levels and mitochondrial fission. Inhibition of Drp1 improved endothelial function in both rat models. These results indicate that hippurate, by itself, can cause endothelial dysfunction. Increased mitochondrial fission plays an active role in hippurate-induced endothelial dysfunction via an increase in mitoROS.
Collapse
Affiliation(s)
- Mengjie Huang
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease, Beijing 100853,People's Republic of China
| | - Ribao Wei
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease, Beijing 100853,People's Republic of China.
| | - Yang Wang
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease, Beijing 100853,People's Republic of China
| | - Tingyu Su
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease, Beijing 100853,People's Republic of China
| | - Ping Li
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease, Beijing 100853,People's Republic of China
| | - Xiangmei Chen
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease, Beijing 100853,People's Republic of China
| |
Collapse
|
15
|
Li XK, Yang ZD, Du J, Xing B, Cui N, Zhang PH, Li H, Zhang XA, Lu QB, Liu W. Endothelial activation and dysfunction in severe fever with thrombocytopenia syndrome. PLoS Negl Trop Dis 2017; 11:e0005746. [PMID: 28806760 PMCID: PMC5581191 DOI: 10.1371/journal.pntd.0005746] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 09/01/2017] [Accepted: 06/25/2017] [Indexed: 12/07/2022] Open
Abstract
Background Pathogenesis of severe fever with thrombocytopenia syndrome (SFTS) has not been well described yet. Recent studies indicate that SFTSV could replicate in endothelial cells. Here we performed a case-control study to determine whether endothelial activation/dysfunction occurred in SFTSV infection and to identify the biomarkers reflecting endothelial dysfunction. Methodology/Principal findings In a case-control study of 134 SFTS patients and 68 healthy controls, serum levels of plasminogen activator inhibitor 1, tissue plasminogen activator, P-selectin, platelet endothelial cell adhesion molecular, CD40 ligand, E-selectin, vascular endothelial growth factor A, serum amyloid antigen 1 (SAA-1) and vascular cell adhesion molecular 1 were significantly enhanced in the patients than the controls (all P<0.05), indicating the occurrence of endothelial activation/dysfunction in SFTS. The intercellular adhesion molecular 1 (ICAM-1) and SAA-1 at the convalescent phase were also significantly associated with severe patients, after adjusting for the potential confounders. The odds ratio was estimated to be 3.364 (95% CI 1.074–10.534) for ICAM-1, and 1.881 (95% CI 1.166–3.034) for SAA-1, respectively. Cutoff value of 1.1×107 pg/mL SAA-1 or 1.2×106 pg/mL ICAM-1 were found to have moderate power of predicting fatal cases. Conclusions The endothelial dysfunction may be one of the pathogenic mechanism of SFTS. The serum levels of ICAM-1 and SAA-1 might be used to predict adverse outcome. Severe fever with thrombocytopenia syndrome (SFTS) is a tick-borne viral disease and first reported in the rural areas of China. Pathogenesis of the disease has not been well described yet. Recent studies indicated that SFTSV replicated in endothelial cells. So, we performed a case-control study to explore whether endothelial activation/dysfunction occurred in SFTSV infection and to identify biomarkers reflecting endothelial dysfunction. We found that the occurrence of endothelial activation/dysfunction in severe fever with thrombocytopenia syndrome and the serum levels of ICAM-1 and SAA-1 might be used to predict adverse outcome.
Collapse
Affiliation(s)
- Xiao-Kun Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, P. R. China
| | - Zhen-Dong Yang
- The 154 Hospital, People’s Liberation Army, Xinyang, P. R. China
| | - Juan Du
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, P. R. China
| | - Bo Xing
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, P. R. China
| | - Ning Cui
- The 154 Hospital, People’s Liberation Army, Xinyang, P. R. China
| | - Pan-He Zhang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, P. R. China
| | - Hao Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, P. R. China
| | - Xiao-Ai Zhang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, P. R. China
| | - Qing-Bin Lu
- Department of Laboratorial Science and Technology, School of Public Health, Peking University, Beijing, P. R. China
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, P. R. China
- * E-mail: (WL); (QBL)
| | - Wei Liu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, P. R. China
- * E-mail: (WL); (QBL)
| |
Collapse
|
16
|
Serum Amyloid A Induces a Vascular Smooth Muscle Cell Phenotype Switch through the p38 MAPK Signaling Pathway. BIOMED RESEARCH INTERNATIONAL 2017. [PMID: 28642873 PMCID: PMC5469989 DOI: 10.1155/2017/4941379] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Atherosclerosis is an important pathological condition which is accompanied by a vascular smooth muscle cell (VSMC) phenotype switch toward a synthetic phenotype. As an acute-phase protein, Serum Amyloid A (SAA) is thought to have a close relationship to atherosclerosis development. However, no study has investigated the direct effect of SAA on the VSMC phenotype switch, as well as the underlying mechanisms. The purpose of our study was to explore the effect of SAA on the VSMC phenotype switch and the potential mechanisms involved. In our study, we found that SAA induced the VSMC phenotype switch which reduced expression of the smooth muscle cell (SMC) marker and enhanced expression of the matrix synthesis related marker. The proliferative ability of VSMCs was also increased by SAA treatment. Furthermore, our research found that SAA activated the ERK1/2 and p38 MAPK signaling pathways. Finally, by applying the ERK1/2 and p38 inhibitors, U0126 and SB203580, we demonstrated that the SAA-induced VSMC phenotype switch was p38-dependent. Taken together, these results indicated that SAA may play an important role in promoting the VSMC phenotype switch through the p38 MAPK signaling pathway.
Collapse
|
17
|
Artenjak A, Omersel J, Ahlin Grabnar P, Mlinarič-Raščan I, Shoenfeld Y, Sodin-Semrl S, Božič B, Čučnik S. Oxidatively altered IgG with increased immunoreactivity to β2-glycoprotein I and its peptide clusters influence human coronary artery endothelial cells. Lupus 2015; 24:448-62. [PMID: 25801888 DOI: 10.1177/0961203314561073] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Oxidative stress has been shown to play a role in modifying antibodies in favor of higher auto-immunoreactivity. We studied the immunoreactivity of oxidized IgG (oxIgG) to β2-glycoprotein I (β2GPI), six peptide sequences corresponding to amino acid clusters on its different domains, to determine their effects on human coronary artery endothelial cells (HCAEC). Human IgG was purified from seven donors, electro-oxidized and checked for immunoreactivity and avidity to β2GPI and to peptides by ELISA. Conformational stability and antibody-antigen complex formation of oxIgG was analyzed by fluorescence spectroscopy and dynamic light scattering. Resting and activated sub-confluent HCAEC were stimulated with oxIgG or IgG. Secreted cytokines were measured by ELISA. Immunoreactivity of seven oxIgG samples increased to 7.5-fold against β2GPI and to 3.8-fold against six peptides as compared to IgG. oxIgG showed low avidity "properties." Conformational changes and exposure of protein hydrophobic regions were confirmed by an elevation in fluorescence (2.4- to 5.0-fold) on bis-ANS dye binding to oxIgG. oxIgG significantly elevated the release of GROα and IL-8 in resting and activated states of HCAEC. Oxidation alters IgG in favor of autoreactivity toward whole β2GPI and corresponding peptides on different domains of β2GPI and could lead to dysfunction of arterial endothelium by upregulation of chemokines.
Collapse
Affiliation(s)
- A Artenjak
- Department of Rheumatology, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - J Omersel
- The chair of Clinical Biochemistry, Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - P Ahlin Grabnar
- The chair of Pharmaceutical Technology, Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - I Mlinarič-Raščan
- The chair of Clinical Biochemistry, Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Y Shoenfeld
- The Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel
| | - S Sodin-Semrl
- Department of Rheumatology, University Medical Centre Ljubljana, Ljubljana, Slovenia Faculty of Mathematics, Natural Sciences and Information Technologies, University of Primorska, Koper, Slovenia
| | - B Božič
- Department of Rheumatology, University Medical Centre Ljubljana, Ljubljana, Slovenia The chair of Clinical Biochemistry, Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - S Čučnik
- Department of Rheumatology, University Medical Centre Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
18
|
Navarro E, Funtikova AN, Fíto M, Schröder H. Can metabolically healthy obesity be explained by diet, genetics, and inflammation? Mol Nutr Food Res 2015; 59:75-93. [PMID: 25418549 DOI: 10.1002/mnfr.201400521] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 11/18/2014] [Accepted: 11/18/2014] [Indexed: 12/20/2022]
Abstract
A substantial proportion of obese individuals do not present cardiometabolic complications such as diabetes, hypertension, or dyslipidemia. Some, but not all, prospective studies observe similar risk of cardiovascular events and all-cause mortality among individuals with this so-called "metabolically healthy obese" (MHO) phenotype, compared to the metabolically healthy normal weight or metabolically healthy non-obese phenotypes. Compared to the metabolically unhealthy obese (MUO) phenotype, MHO is often characterized by a more favorable inflammatory profile, less visceral fat, less infiltration of macrophages into adipose tissue, and smaller adipocyte cell size. Tipping the inflammation balance in adipose tissue might be particularly important for metabolic health in the obese. While the potential role of genetic predisposition or lifestyle factors such as diet in the MHO phenotype is yet to be clarified, it is well known that diet affects inflammation profile and contributes to the functionality of adipose tissue. This review will discuss genetic predisposition and the molecular mechanisms underlying the potential effect of food on the development of the metabolic phenotype characteristic of obesity.
Collapse
|
19
|
McElroy AK, Erickson BR, Flietstra TD, Rollin PE, Nichol ST, Towner JS, Spiropoulou CF. Biomarker correlates of survival in pediatric patients with Ebola virus disease. Emerg Infect Dis 2015; 20:1683-90. [PMID: 25279581 PMCID: PMC4193175 DOI: 10.3201/eid2010.140430] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Children who had certain endothelial and immune function markers were more likely to survive infection. Outbreaks of Ebola virus disease (EVD) occur sporadically in Africa and are associated with high case-fatality rates. Historically, children have been less affected than adults. The 2000–2001 Sudan virus–associated EVD outbreak in the Gulu district of Uganda resulted in 55 pediatric and 161 adult laboratory-confirmed cases. We used a series of multiplex assays to measure the concentrations of 55 serum analytes in specimens from patients from that outbreak to identify biomarkers specific to pediatric disease. Pediatric patients who survived had higher levels of the chemokine regulated on activation, normal T-cell expressed and secreted marker and lower levels of plasminogen activator inhibitor 1, soluble intracellular adhesion molecule, and soluble vascular cell adhesion molecule than did pediatric patients who died. Adult patients had similar levels of these analytes regardless of outcome. Our findings suggest that children with EVD may benefit from different treatment regimens than those for adults.
Collapse
|
20
|
Serum amyloid A receptor blockade and incorporation into high-density lipoprotein modulates its pro-inflammatory and pro-thrombotic activities on vascular endothelial cells. Int J Mol Sci 2015; 16:11101-24. [PMID: 25988387 PMCID: PMC4463692 DOI: 10.3390/ijms160511101] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Revised: 03/25/2015] [Accepted: 04/27/2015] [Indexed: 02/07/2023] Open
Abstract
The acute phase protein serum amyloid A (SAA), a marker of inflammation, induces expression of pro-inflammatory and pro-thrombotic mediators including ICAM-1, VCAM-1, IL-6, IL-8, MCP-1 and tissue factor (TF) in both monocytes/macrophages and endothelial cells, and induces endothelial dysfunction—a precursor to atherosclerosis. In this study, we determined the effect of pharmacological inhibition of known SAA receptors on pro-inflammatory and pro-thrombotic activities of SAA in human carotid artery endothelial cells (HCtAEC). HCtAEC were pre-treated with inhibitors of formyl peptide receptor-like-1 (FPRL-1), WRW4; receptor for advanced glycation-endproducts (RAGE), (endogenous secretory RAGE; esRAGE) and toll-like receptors-2/4 (TLR2/4) (OxPapC), before stimulation by added SAA. Inhibitor activity was also compared to high-density lipoprotein (HDL), a known inhibitor of SAA-induced effects on endothelial cells. SAA significantly increased gene expression of TF, NFκB and TNF and protein levels of TF and VEGF in HCtAEC. These effects were inhibited to variable extents by WRW4, esRAGE and OxPapC either alone or in combination, suggesting involvement of endothelial cell SAA receptors in pro-atherogenic gene expression. In contrast, HDL consistently showed the greatest inhibitory action, and often abrogated SAA-mediated responses. Increasing HDL levels relative to circulating free SAA may prevent SAA-mediated endothelial dysfunction and ameliorate atherogenesis.
Collapse
|
21
|
Association of selenoprotein S gene polymorphism with ischemic stroke in a Chinese case–control study. Blood Coagul Fibrinolysis 2015; 26:131-5. [DOI: 10.1097/mbc.0000000000000202] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
22
|
Defagó MD, Elorriaga N, Irazola VE, Rubinstein AL. Influence of food patterns on endothelial biomarkers: a systematic review. J Clin Hypertens (Greenwich) 2014; 16:907-13. [PMID: 25376124 PMCID: PMC4270900 DOI: 10.1111/jch.12431] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Revised: 09/04/2014] [Accepted: 09/04/2014] [Indexed: 01/19/2023]
Abstract
The purpose of this study was to conduct a systematic review on the association of food patterns (FPs) and endothelial biomarkers. An electronic literature search from 1990 to 2012 was conducted and reference lists and experts were consulted. Studies without dietary intervention and without language restrictions were considered. Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines were employed. Methodological quality was assessed by Strengthening the Reporting of Observational Studies in Epidemiology guidelines. A total of 546 references were identified, of which 8 were finally included. Several FPs were identified. Healthy FPs (abundant in fruits and vegetables) had a beneficial impact on endothelial function as estimated by circulating levels of biomarkers such as C-reactive protein, soluble intercellular adhesion molecule 1, soluble vascular adhesion molecule 1, and E-selectin molecules. Westernized patterns (higher intakes of processed meats, sweets, fried foods, and refined grains) were positively associated with inflammation molecules and atherogenic promoters. The study of FPs in relation to endothelial function contributes to the development of dietary recommendations for improved cardiovascular health and therefore a better lifestyle.
Collapse
Affiliation(s)
- María Daniela Defagó
- Centro de Excelencia en Salud Cardiovascular para el Cono Sur (CESCAS/SACECH)Instituto de Efectividad Clínica y SanitariaBuenos AiresArgentina
- Facultad de Ciencias MédicasEscuela de NutriciónUniversidad Nacional de CórdobaCórdobaArgentina
| | - Natalia Elorriaga
- Centro de Excelencia en Salud Cardiovascular para el Cono Sur (CESCAS/SACECH)Instituto de Efectividad Clínica y SanitariaBuenos AiresArgentina
| | - Vilma Edith Irazola
- Centro de Excelencia en Salud Cardiovascular para el Cono Sur (CESCAS/SACECH)Instituto de Efectividad Clínica y SanitariaBuenos AiresArgentina
| | - Adolfo Luis Rubinstein
- Centro de Excelencia en Salud Cardiovascular para el Cono Sur (CESCAS/SACECH)Instituto de Efectividad Clínica y SanitariaBuenos AiresArgentina
| |
Collapse
|
23
|
Saber AT, Jacobsen NR, Jackson P, Poulsen SS, Kyjovska ZO, Halappanavar S, Yauk CL, Wallin H, Vogel U. Particle-induced pulmonary acute phase response may be the causal link between particle inhalation and cardiovascular disease. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2014; 6:517-31. [PMID: 24920450 PMCID: PMC4285160 DOI: 10.1002/wnan.1279] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Revised: 05/14/2014] [Accepted: 05/20/2014] [Indexed: 01/08/2023]
Abstract
Inhalation of ambient and workplace particulate air pollution is associated with increased risk of cardiovascular disease. One proposed mechanism for this association is that pulmonary inflammation induces a hepatic acute phase response, which increases risk of cardiovascular disease. Induction of the acute phase response is intimately linked to risk of cardiovascular disease as shown in both epidemiological and animal studies. Indeed, blood levels of acute phase proteins, such as C-reactive protein and serum amyloid A, are independent predictors of risk of cardiovascular disease in prospective epidemiological studies. In this review, we present and review emerging evidence that inhalation of particles (e.g., air diesel exhaust particles and nanoparticles) induces a pulmonary acute phase response, and propose that this induction constitutes the causal link between particle inhalation and risk of cardiovascular disease. Increased levels of acute phase mRNA and proteins in lung tissues, bronchoalveolar lavage fluid and plasma clearly indicate pulmonary acute phase response following pulmonary deposition of different kinds of particles including diesel exhaust particles, nanoparticles, and carbon nanotubes. The pulmonary acute phase response is dose-dependent and long lasting. Conversely, the hepatic acute phase response is reduced relative to lung or entirely absent. We also provide evidence that pulmonary inflammation, as measured by neutrophil influx, is a predictor of the acute phase response and that the total surface area of deposited particles correlates with the pulmonary acute phase response. We discuss the implications of these findings in relation to occupational exposure to nanoparticles. How to cite this article: WIREs Nanomed Nanobiotechnol 2014, 6:517–531. doi: 10.1002/wnan.1279
Collapse
Affiliation(s)
- Anne T Saber
- National Research Centre for the Working Environment, Copenhagen, Denmark
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Eren E, Ellidag HY, Yılmaz A, Aydın O, Yılmaz N. Acute Phase Response: Implication in ST-segment Elevation Myocardial Infarction. Open Biochem J 2014; 8:44-51. [PMID: 24894970 PMCID: PMC4040932 DOI: 10.2174/1874091x01408010044] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 03/07/2014] [Accepted: 03/09/2014] [Indexed: 12/18/2022] Open
Abstract
We aimed to investigate the relation between serum inflammatory markers, 25OHvit-D3 and oxidative stress markers, namely paraoxonase1-arylesterase (PON1-ARE), total antioxidant status (TAS) and total oxidant status (TOS) in 30 male patients with ST-elevation myocardial infarction(STEMI) . There was negative correlation between tumor necrosis factor alpha and ARE; positive correlations between serum amyloid A(SAA) and oxidative stress index, SAA and TOS, 25OHvit-D3 and ARE. There was no statistically significant correlation between inflammation makers, oxidative stress markers and Gensini score. The main finding of our study was the tendency of inflammation markers, and oxidative stress markers, to change in relatively clear opposite directions in STEMI.
Collapse
Affiliation(s)
- Esin Eren
- Antalya Public Health Center of Ministry of Health, Antalya, Turkey
| | - Hamit Yasar Ellidag
- Central Laboratories of Antalya Education and Research Hospital of Ministry of Health, Antalya, Turkey
| | - Akar Yılmaz
- Cardiology of Antalya Education and Research Hospital of Ministry of Health, Antalya, Turkey
| | - Ozgür Aydın
- Maternity and Children's Hospital, Batman, Turkey
| | - Necat Yılmaz
- Central Laboratories of Antalya Education and Research Hospital of Ministry of Health, Antalya, Turkey
| |
Collapse
|
25
|
Abstract
S100A8, S100A9 and S100A12 are considered proinflammatory mediators of atherosclerosis. Known as calgranulins, they are major components of neutrophils and are upregulated in macrophages and foam cells. They influence leukocyte recruitment, and may propagate inflammation by binding TLR4 and/or receptor for advanced glycation endproducts (RAGE). However, the receptors for calgranulins remain an enigma; we have no evidence for TLR4 or RAGE activation by S100A8 or S100A12. Moreover, gene regulation studies suggest antiinflammatory functions for S100A8 and emerging reports indicate pleiotropic roles. Unlike S100A9, S100A8 effectively scavenges oxidants generated by the myeloperoxidase system in vivo, forming novel thiol modifications. S100A8 is also readily S-nitrosylated, stabilizing nitric oxide and transporting it to hemoglobin. S100A8-SNO reduces leukocyte transmigration in the vasculature. S-glutathionylation of S100A9 modifies its effects on leukocyte adhesion. Both S100A8 forms inhibit mast cell activation, at least partially by scavenging reactive oxygen species required for signaling. Conversely, S100A12 activates and sequesters mast cells. However S100A12 suppresses proinflammatory cytokine induction by SAA-activated monocytes and macrophages, and inhibits matrix metalloprotease activity. We propose that the abundance and types of cells expressing calgranulins in particular microenvironments, their relative concentrations and post-translational modifications may have distinct functional outcomes, including those that are protective, at different stages of atherogenesis.
Collapse
Affiliation(s)
- Carolyn L Geczy
- Inflammation and Infection Research Centre, School of Medical Sciences, University of New South Wales
| | | | | |
Collapse
|
26
|
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: 3.9] [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
|
27
|
Lakota K, Mrak-Poljsak K, Bozic B, Tomsic M, Sodin-Semrl S. Serum amyloid A activation of human coronary artery endothelial cells exhibits a neutrophil promoting molecular profile. Microvasc Res 2013; 90:55-63. [PMID: 23938271 DOI: 10.1016/j.mvr.2013.07.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Revised: 07/25/2013] [Accepted: 07/29/2013] [Indexed: 10/26/2022]
Abstract
BACKGROUND Serum amyloid A (SAA) has been shown to be an active participant in atherosclerosis and cardiovascular diseases. SAA-stimulated human coronary artery endothelial cells (HCAEC) were reported to release pro-inflammatory cytokines, chemokines and adhesion molecules; however it remains unclear which putative SAA receptors are present in these cells and how they act. We investigated the effects of inflammatory stimuli on the expression of SAA receptors, signaling pathways and molecular profiles in HCAEC. METHODOLOGY/PRINCIPLE FINDINGS HCAEC were cultured in vitro and stimulated with SAA (1000nM) or IL-1β (1000pg/ml). Expression of mRNA was determined by qPCR, and expression and quantification of proteins were assessed by dot array blots and ELISA, respectively. Protein phosphorylation was determined by dot blot arrays and Western blots. We report that all potential SAA receptors tested (FPR2/ALX, RAGE, TANIS, TLR2, TLR4 and CLA-1/hSR-B1) are expressed in HCAEC. Importantly, IL-1β or SAA significantly increased solely the expression of the innate immune receptor TLR2. SAA upregulated the phosphorylation of ERK1/2, NF-κB (p65, p105) and JNK, as well as expression/release of IL-6, IL-8, G-CSF, GM-CSF, ICAM-1 and VCAM-1, all potent molecules involved in neutrophil-related activities. A TLR2-dependent positive feedback mechanism of SAA expression was found. CONCLUSION/SIGNIFICANCE SAA stimulated responses in HCAEC target neutrophil rather than monocyte/macrophage activation.
Collapse
Affiliation(s)
- Katja Lakota
- University Medical Centre, Department of Rheumatology, Ljubljana, Slovenia
| | | | | | | | | |
Collapse
|
28
|
Saber AT, Lamson JS, Jacobsen NR, Ravn-Haren G, Hougaard KS, Nyendi AN, Wahlberg P, Madsen AM, Jackson P, Wallin H, Vogel U. Particle-induced pulmonary acute phase response correlates with neutrophil influx linking inhaled particles and cardiovascular risk. PLoS One 2013; 8:e69020. [PMID: 23894396 PMCID: PMC3722244 DOI: 10.1371/journal.pone.0069020] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Accepted: 06/02/2013] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Particulate air pollution is associated with cardiovascular disease. Acute phase response is causally linked to cardiovascular disease. Here, we propose that particle-induced pulmonary acute phase response provides an underlying mechanism for particle-induced cardiovascular risk. METHODS We analysed the mRNA expression of Serum Amyloid A (Saa3) in lung tissue from female C57BL/6J mice exposed to different particles including nanomaterials (carbon black and titanium dioxide nanoparticles, multi- and single walled carbon nanotubes), diesel exhaust particles and airborne dust collected at a biofuel plant. Mice were exposed to single or multiple doses of particles by inhalation or intratracheal instillation and pulmonary mRNA expression of Saa3 was determined at different time points of up to 4 weeks after exposure. Also hepatic mRNA expression of Saa3, SAA3 protein levels in broncheoalveolar lavage fluid and in plasma and high density lipoprotein levels in plasma were determined in mice exposed to multiwalled carbon nanotubes. RESULTS Pulmonary exposure to particles strongly increased Saa3 mRNA levels in lung tissue and elevated SAA3 protein levels in broncheoalveolar lavage fluid and plasma, whereas hepatic Saa3 levels were much less affected. Pulmonary Saa3 expression correlated with the number of neutrophils in BAL across different dosing regimens, doses and time points. CONCLUSIONS Pulmonary acute phase response may constitute a direct link between particle inhalation and risk of cardiovascular disease. We propose that the particle-induced pulmonary acute phase response may predict risk for cardiovascular disease.
Collapse
|
29
|
Chung YM, Goyette J, Tedla N, Hsu K, Geczy CL. S100A12 suppresses pro-inflammatory, but not pro-thrombotic functions of serum amyloid A. PLoS One 2013; 8:e62372. [PMID: 23638054 PMCID: PMC3634854 DOI: 10.1371/journal.pone.0062372] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Accepted: 03/20/2013] [Indexed: 12/15/2022] Open
Abstract
S100A12 is elevated in the circulation in patients with chronic inflammatory diseases and recent studies indicate pleiotropic functions. Serum amyloid A induces monocyte cytokines and tissue factor. S100A12 did not stimulate IL-6, IL-8, IL-1β or TNF-α production by human peripheral blood mononuclear cells but low amounts consistently reduced cytokine mRNA and protein levels induced by serum amyloid A, by ∼49% and ∼46%, respectively. However, S100A12 did not affect serum amyloid A-induced monocyte tissue factor. In marked contrast, LPS-induced cytokines or tissue factor were not suppressed by S100A12. S100A12 did not alter cytokine mRNA stability or the cytokine secretory pathway. S100A12 and serum amyloid A did not appear to form complexes and although they may have common receptors, suppression was unlikely via receptor competition. Serum amyloid A induces cytokines via activation of NF-κB and the MAPK pathways. S100A12 reduced serum amyloid A-, but not LPS-induced ERK1/2 phosphorylation to baseline. It did not affect JNK or p38 phosphorylation or the NF-κB pathway. Reduction in ERK1/2 phosphorylation by S100A12 was unlikely due to changes in intracellular reactive oxygen species, Ca2+ flux or to recruitment of phosphatases. We suggest that S100A12 may modulate sterile inflammation by blunting pro-inflammatory properties of lipid-poor serum amyloid A deposited in chronic lesions where both proteins are elevated as a consequence of macrophage activation.
Collapse
Affiliation(s)
- Yuen Ming Chung
- Inflammation and Infection Research Centre, University of New South Wales, Sydney, New South Wales, Australia
| | - Jesse Goyette
- Inflammation and Infection Research Centre, University of New South Wales, Sydney, New South Wales, Australia
| | - Nicodemus Tedla
- Inflammation and Infection Research Centre, University of New South Wales, Sydney, New South Wales, Australia
| | - Kenneth Hsu
- Inflammation and Infection Research Centre, University of New South Wales, Sydney, New South Wales, Australia
| | - Carolyn L. Geczy
- Inflammation and Infection Research Centre, University of New South Wales, Sydney, New South Wales, Australia
- * E-mail:
| |
Collapse
|
30
|
Leow KY, Goh WWB, Tan SZ, Lim J, Ng K, Oh VMS, Low AFH, Heng CK. Variant screening of the serum amyloid A1 gene and functional study of the p.Gly90Asp variant for its role in atherosclerosis. Atherosclerosis 2013; 227:112-7. [PMID: 23357645 DOI: 10.1016/j.atherosclerosis.2013.01.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2012] [Revised: 12/29/2012] [Accepted: 01/01/2013] [Indexed: 12/17/2022]
Abstract
OBJECTIVES Serum amyloid A1 (SAA1) is a major acute-phase protein that is increasingly used as a reliable predictor of coronary artery disease (CAD). In this study we aim to screen the SAAI promoter and exons for genetic variants and to determine their association with CAD. In addition, we also carried out functional study on a variant of p.Gly90Asp encoded by the SAA1 gene. METHODS Variant screening of SAA1 was performed using high resolution melting (HRM) analysis. Genetic association of p.Gly90Asp with CAD was determined in 800 CAD patients and 773 Chinese control subjects. Functional study of p.Gly90Asp was carried out using THP-1-derived macrophages and HL-60 promyelocytic leukemia cells. RESULTS A total of 6 SNPs were identified, of which 2 were found to be novel (c.-913G > A and c.92-5T > G). The rare allele of p.Gly90Asp has a lower frequency of 0.013 in the CAD patients although this is not statistically significant. Functional studies of p.Gly90Asp revealed that the variant has decreased upregulation of key cytokines such as IL-8, MCP-1 and TNF-α as well as SERPINB2. CONCLUSIONS We found the variant p.Gly90Asp SAA1 protein eliciting significantly reduced inflammatory responses in macrophages through a reduction in the secretion of inflammatory cytokines. Despite strong functional effects, the minor allele frequency is too low in the population to attain statistical significance difference between cases and controls.
Collapse
Affiliation(s)
- Koon-Yeow Leow
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, 1E, Kent Ridge Rd, Singapore 119228, Singapore.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Liu LR, Lin SP, Chen CC, Chen YJ, Tai CC, Chang SC, Juang RH, Tseng YW, Liu BH, Mersmann HJ, Shen TL, Ding ST. Serum amyloid A induces lipolysis by downregulating perilipin through ERK1/2 and PKA signaling pathways. Obesity (Silver Spring) 2011; 19:2301-9. [PMID: 21701568 DOI: 10.1038/oby.2011.176] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Serum amyloid A (SAA) is not only an apolipoprotein, but also a member of the adipokine family with potential to enhance lipolysis. The purpose of this study was to explore how SAA facilitates lipolysis in porcine adipocytes. We found that SAA increased the phosphorylation of perilipin and hormone-sensitive lipase (HSL) after 12-h treatment and decreased perilipin expression after 24-h treatment, and these effects were prevented by extracellular signal-regulated kinase (ERK) or protein kinase A (PKA) inhibitors in primary adipocyte cell culture. SAA treatment decreased HSL and adipose triglyceride lipase (ATGL) expression. SAA treatment also activated ERK and PKA by increasing the phosphorylation of these kinases. Moreover, SAA significantly increased porcine adipocyte glycerol release and lipase activity, which was inhibited by either ERK (PD98059) or PKA (H89) inhibitors, suggesting that ERK and PKA were involved in mediating SAA enhanced lipolysis. SAA downregulated the expression of peroxisome proliferator-activated receptor γ (PPARγ) mRNA, which was reversed by the ERK inhibitor. We performed a porcine perilipin promoter assay in differentiated 3T3-L1 adipocytes and found that SAA reduced the porcine perilipin promoter specifically through the function of its PPAR response element (PPRE), and this effect was reversed by the ERK inhibitor. These findings demonstrate that SAA-induced lipolysis is a result of downregulation of perilipin and activation of HSL via ERK/PPARγ and PKA signaling pathways. The finding could lead to developing new strategies for reducing human obesity.
Collapse
Affiliation(s)
- Lih R Liu
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Witting PK, Song C, Hsu K, Hua S, Parry SN, Aran R, Geczy C, Freedman SB. The acute-phase protein serum amyloid A induces endothelial dysfunction that is inhibited by high-density lipoprotein. Free Radic Biol Med 2011; 51:1390-8. [PMID: 21784147 DOI: 10.1016/j.freeradbiomed.2011.06.031] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2010] [Revised: 06/24/2011] [Accepted: 06/25/2011] [Indexed: 11/17/2022]
Abstract
The acute-phase protein serum amyloid A (SAA) is elevated during inflammation and may be deposited in atheroma where it promotes atherosclerosis. We investigated the proatherogenic effects of SAA on the vascular endothelium and their regulation by high-density lipoprotein (HDL). Exposure of human aortic endothelial cells (HAEC) to SAA (0.25-25μg/ml) decreased nitric oxide ((•)NO) synthesis/bioavailability, although the endothelial NO synthase monomer-to-dimer ratio was unaffected. SAA (10μg/ml) stimulated a Ca(2+) influx linked to apocynin-sensitive superoxide radical anion (O(2)(•-)) production. Gene expression for arginase-1, nuclear factor κB (NF-κB), interleukin-8, and tissue factor (TF) increased within 4h of SAA stimulation. Enzymatically active Arg-1/2 was detected in HAEC cultured with SAA for 24h. Therefore, in addition to modulating (•)NO bioavailability by stimulating O(2)(•-) production in the endothelium, SAA modulated vascular l-Arg bioavailability. SAA also diminished relaxation of preconstricted aortic rings induced by acetylcholine, and added superoxide dismutase restored the vascular response. Preincubation of HAEC with HDL (100 or 200, but not 50, μg/ml) before (not after) SAA treatment ameliorated the Ca(2+) influx and O(2)(•-) production; decreased TF, NF-κB, and Arg-1 gene expression; and preserved overall vascular function. Thus, SAA may promote endothelial dysfunction by modulating (•)NO and l-Arg bioavailability, and HDL pretreatment may be protective. The relative HDL to SAA concentrations may regulate the proatherogenic properties of SAA on the vascular endothelium.
Collapse
Affiliation(s)
- Paul K Witting
- Discipline of Pathology, Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Dong Z, Wu T, Qin W, An C, Wang Z, Zhang M, Zhang Y, Zhang C, An F. Serum amyloid A directly accelerates the progression of atherosclerosis in apolipoprotein E-deficient mice. Mol Med 2011; 17:1357-64. [PMID: 21953420 DOI: 10.2119/molmed.2011.00186] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Accepted: 09/20/2011] [Indexed: 11/06/2022] Open
Abstract
Although serum amyloid A (SAA) is an excellent marker for coronary artery disease, its direct effect on atherogenesis in vivo is obscure. In this study we investigated the direct effect of SAA on promoting the formation of atherosclerosis in apolipoprotein E-deficient (ApoE⁻/⁻) mice. Murine SAA lentivirus was constructed and injected into ApoE⁻/⁻ mice intravenously. Then, experimental mice were fed a chow diet (5% fat and no added cholesterol) for 14 wks. The aortic atherosclerotic lesion area was larger with than without SAA treatment. With increased SAA levels, the plasma levels of interleukin-6 and tumor necrosis factor-α were significantly increased. Macrophage infiltration in atherosclerotic regions was enhanced with SAA treatment. A migration assay revealed prominent dose-dependent chemotaxis of SAA to macrophages. Furthermore, the expression of monocyte chemotactic protein-1 and vascular cell adhesion molecule-1 (VCAM-1) was upregulated significantly with SAA treatment. SAA-induced VCAM-1 production was detected in human aortic endothelial cells in vitro. Thus, an increase in plasma SAA directly accelerates the progression of atherosclerosis in ApoE⁻/⁻ mice. SAA is not only a risk marker for atherosclerosis but also an active participant in atherogenesis.
Collapse
Affiliation(s)
- Zhe Dong
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Shandong University Qilu Hospital, Jinan, Shandong, China
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Yang Q, Xue HM, Wong WT, Tian XY, Huang Y, Tsui SKW, Ng PKS, Wohlfart P, Li H, Xia N, Tobias S, Underwood MJ, He GW. AVE3085, an enhancer of endothelial nitric oxide synthase, restores endothelial function and reduces blood pressure in spontaneously hypertensive rats. Br J Pharmacol 2011; 163:1078-1085. [PMID: 21385179 PMCID: PMC3130953 DOI: 10.1111/j.1476-5381.2011.01308.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2010] [Revised: 01/03/2011] [Accepted: 01/31/2011] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND AND PURPOSE Nitric oxide (NO) plays an important role in endothelial function, and impaired NO production is involved in hypertension. Therefore, compounds that regulate endothelial NO synthase (eNOS) may be of therapeutic benefit. A novel, low molecular weight compound AVE3085 is a recently developed compound with the ability to enhance eNOS transcription. The present study investigated the effects of AVE3085 in endothelial dysfunction associated with hypertension. EXPERIMENTAL APPROACH Spontaneously hypertensive rats (SHRs) were treated with AVE 3085 (10 mg·kg·day(-1) , orally) for 4 weeks. Isometric force measurement was performed on rings of isolated aortae in organ baths. Protein expression of eNOS, phosphorylated-eNOS and nitrotyrosine in the aortae were examined by Western blotting. mRNA for eNOS in rat aortae were examined by reverse-transcriptase polymerase chain reaction (RT-PCR). KEY RESULTS AVE3085 greatly improved endothelium-dependent relaxations in the aortae of SHRs. This functional change was accompanied by up-regulated expression of eNOS protein and mRNA, enhanced eNOS phosphorylation and decreased formation of nitrotyrosine. Furthermore, AVE3085 treatment reduced the blood pressure in SHR without affecting that of hypertensive eNOS(-/-) mice. CONCLUSIONS AND IMPLICATIONS The eNOS-transcription enhancer AVE3085 restored impaired endothelial function in a hypertensive model. The present study provides a solid basis for the potential development of eNOS-targeting drugs to restore down-regulated eNOS, as a new strategy in hypertension.
Collapse
MESH Headings
- Administration, Oral
- Animals
- Antihypertensive Agents/administration & dosage
- Antihypertensive Agents/pharmacology
- Antihypertensive Agents/therapeutic use
- Aorta, Thoracic/cytology
- Aorta, Thoracic/drug effects
- Aorta, Thoracic/enzymology
- Benzodioxoles/administration & dosage
- Benzodioxoles/pharmacology
- Benzodioxoles/therapeutic use
- Blood Pressure/drug effects
- Blotting, Western
- Dose-Response Relationship, Drug
- Endothelial Cells/drug effects
- Endothelial Cells/enzymology
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/physiology
- Hypertension/drug therapy
- Hypertension/enzymology
- Hypertension/physiopathology
- Indans/administration & dosage
- Indans/pharmacology
- Indans/therapeutic use
- Male
- Nitric Oxide Synthase Type III/genetics
- Nitric Oxide Synthase Type III/metabolism
- Rats
- Rats, Inbred SHR
- Rats, Inbred WKY
- Reverse Transcriptase Polymerase Chain Reaction
Collapse
Affiliation(s)
- Qin Yang
- Department of Surgery, The Chinese University of Hong KongHong Kong, China & TEDA International Cardiovascular Hospital, Medical College, Nankai UniversityTianjin, China
| | - Hong-Mei Xue
- Department of Surgery, The Chinese University of Hong KongHong Kong, China & TEDA International Cardiovascular Hospital, Medical College, Nankai UniversityTianjin, China
| | - Wing-Tak Wong
- Institute of Vascular Medicine, Li Ka Shing Institute of Health Sciences and School of Biomedical Sciences, The Chinese University of Hong KongHong Kong, China
| | - Xiao-Yu Tian
- Institute of Vascular Medicine, Li Ka Shing Institute of Health Sciences and School of Biomedical Sciences, The Chinese University of Hong KongHong Kong, China
| | - Yu Huang
- Institute of Vascular Medicine, Li Ka Shing Institute of Health Sciences and School of Biomedical Sciences, The Chinese University of Hong KongHong Kong, China
| | - Stephen KW Tsui
- Institute of Vascular Medicine, Li Ka Shing Institute of Health Sciences and School of Biomedical Sciences, The Chinese University of Hong KongHong Kong, China
| | - Patrick KS Ng
- Institute of Vascular Medicine, Li Ka Shing Institute of Health Sciences and School of Biomedical Sciences, The Chinese University of Hong KongHong Kong, China
| | - Paulus Wohlfart
- Department of Molecular and Cell Biology, Sanofi-Aventis, Therapeutic Domain CardiovascularGermany
| | - Huige Li
- Department of Pharmacology, Johannes Gutenberg UniversityMainz, Germany
| | - Ning Xia
- Department of Pharmacology, Johannes Gutenberg UniversityMainz, Germany
| | - Silke Tobias
- Department of Pharmacology, Johannes Gutenberg UniversityMainz, Germany
| | - Malcolm John Underwood
- Department of Surgery, The Chinese University of Hong KongHong Kong, China & TEDA International Cardiovascular Hospital, Medical College, Nankai UniversityTianjin, China
| | - Guo-Wei He
- Department of Surgery, The Chinese University of Hong KongHong Kong, China & TEDA International Cardiovascular Hospital, Medical College, Nankai UniversityTianjin, China
- Providence Heart & Vascular Institute, Albert Starr Academic Center, Department of Surgery, Oregon Health and Science UniversityPortland, OR, USA
| |
Collapse
|
35
|
|
36
|
Salini V, Saggini A, Maccauro G, Caraffa A, Shaik-Dasthagirisaheb Y, Conti P. Inflammatory Markers: Serum Amyloid A, Fibrinogen and C-Reactive Protein — A Revisited Study. EUR J INFLAMM 2011. [DOI: 10.1177/1721727x1100900202] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The acute phase response is the part of the innate defence system of an animal against trauma, inflammation or infection. During this response, there is increased production and release of certain plasma proteins known as acute phase proteins, which include C-reactive protein (CRP), serum amyloid A (SAA) and fibrinogen (Fg). CRP consists of five identical subunits of 206 amino acids with a molecular weight of approximately 23 kDa. There is strong evidence from numerous studies that CRP is a predictor of inflammation. The acute-phase protein serum amyloid A (SAA) is a clinically useful marker of inflammation. SAA plays not only an important role in the development of AA amyloidosis (an important complication of rheumatoid arthritis) but also interacts with events closely involved in the metabolic syndrome as a high- and low-grade inflammatory modulator. Fibrinogen (Fg) is a high molecular weight plasma adhesion protein and is a biomarker of inflammation. It is synthesized and assembled in hepatocytes and fibroblasts and when secreted into the circulation, its plasma half-life ranges from 3 to 4 days. Several cytokines, are involved in the induction of acute phase protein synthesis, but the mutual importance of these cytokines seems to be cell-type specific and to vary in various experimental settings. Here we revisited the classic acute phase proteins SAA, C-Reactive protein and fibrinogen in their role in inflammation and their interrelationship with some cytokines.
Collapse
Affiliation(s)
- V. Salini
- Orthopaedic Division University of Chieti-Pescara, Medical School, Chieti, Italy
| | - A. Saggini
- Department of Dermatology, University of Rome Tor Vergata, Rome, Italy
| | - G. Maccauro
- Department of Orthopaedics, Catholic University of Rome, Rome, Italy
| | - A. Caraffa
- Orthopaedics Division, University of Perugia, Perugia, Italy
| | | | - P. Conti
- Immunology Division, University of Chieti-Pescara, Medical School, Chieti, Italy
| |
Collapse
|
37
|
Nakarai H, Yamashita A, Nagayasu S, Iwashita M, Kumamoto S, Ohyama H, Hata M, Soga Y, Kushiyama A, Asano T, Abiko Y, Nishimura F. Adipocyte-macrophage interaction may mediate LPS-induced low-grade inflammation: Potential link with metabolic complications. Innate Immun 2011; 18:164-70. [DOI: 10.1177/1753425910393370] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Chronic low-grade infection has been suggested to be associated with metabolic disorder such as diabetes. However, the molecular mechanism underlying this important association is largely unknown. The only clue established so far is that many subjects exhibit elevated levels of C-reactive protein as measured by highly sensitive assay. Here, we hypothesized that adipocyte–macrophage interaction plays a key role in amplifying such low grade infection to the level of influencing metabolic disorders. The presence of macrophages in abdominal adipose tissues was investigated by immunohistochemistry. To see whether molecules associated with acute phase protein, LPS signaling, and persistent recruitment of monocytes, are produced at higher amounts in adipocytes co-cultured with macrophages stimulated with low concentration of LPS (1 ng/ml), we measured serum amyloid A (SAA), LPS binding protein (LBP), soluble CD14 (sCD14), and RANTES levels in culture supernatant of co-cultures. Lastly, we investigated in vivo effect of low-grade LPS infusion on the production of these molecules using obese model mice. The macrophages were certainly identified in abdominal adipose tissues. Investigated molecules, especially LBP, SAA, and RANTES were produced at higher amounts in co-cultures stimulated with LPS compared with the cells without LPS. The ob/ob, and high-fat diet-induced obesity mice produced higher amounts of LBP, SAA, and RANTES one day after LPS infusion (1 ng/ml/g body weight) compared with ob/– and normal-fat fed control mice. Thus, adipocytes and infiltrated macrophages, and their interaction with low endotoxin stimulation appear to play an important role in amplifying and maintaining LPS-induced low-grade inflammation.
Collapse
Affiliation(s)
- Hideo Nakarai
- Departments of Dental Science for Health Promotion, Hiroshima University Graduate School of Biomedical Science, Minami-ku, Hiroshima, Japan
| | - Akiko Yamashita
- Departments of Dental Science for Health Promotion, Hiroshima University Graduate School of Biomedical Science, Minami-ku, Hiroshima, Japan
| | - Shintaro Nagayasu
- Departments of Dental Science for Health Promotion, Hiroshima University Graduate School of Biomedical Science, Minami-ku, Hiroshima, Japan
| | - Misaki Iwashita
- Departments of Dental Science for Health Promotion, Hiroshima University Graduate School of Biomedical Science, Minami-ku, Hiroshima, Japan
| | - Sonoko Kumamoto
- Departments of Dental Science for Health Promotion, Hiroshima University Graduate School of Biomedical Science, Minami-ku, Hiroshima, Japan
| | - Hideki Ohyama
- Department of Pathology, Hyogo College of Medicine, Hyogo, Japan
| | - Masaki Hata
- Department of Pathology, Hyogo College of Medicine, Hyogo, Japan
| | - Yoshihiko Soga
- Department of Pathophysiology Periodontal Science, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | | | - Tomoichiro Asano
- Department of Biomedical Chemistry, Hiroshima University Graduate School of Biomedical Sciences, Hiroshima, Japan
| | - Yoshimitsu Abiko
- Department of Biochemistry and Molecular Biology, Nihon University School of Dentistry at Matsudo, Chiba, Japan
| | - Fusanori Nishimura
- Departments of Dental Science for Health Promotion, Hiroshima University Graduate School of Biomedical Science, Minami-ku, Hiroshima, Japan
| |
Collapse
|
38
|
Tanaka T, Ikeda K, Yamamoto Y, Iida H, Kikuchi H, Morita T, Yamasoba T, Nagai R, Nakajima T. Effects of Serum Amyloid A and Lysophosphatidylcholine on Intracellular Calcium Concentration in Human Coronary Artery Smooth Muscle Cells. Int Heart J 2011; 52:185-93. [DOI: 10.1536/ihj.52.185] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Tomofumi Tanaka
- Department of Cardiovascular Medicine, The University of Tokyo
| | - Kenichi Ikeda
- Department of Cardiovascular Medicine, The University of Tokyo
| | - Yumiko Yamamoto
- Department of Cardiovascular Medicine, The University of Tokyo
| | - Haruko Iida
- Department of Ischemic Circulatory Physiology, The University of Tokyo
| | | | - Toshihiro Morita
- Department of Ischemic Circulatory Physiology, The University of Tokyo
| | | | - Ryozo Nagai
- Department of Cardiovascular Medicine, The University of Tokyo
| | - Toshiaki Nakajima
- Department of Ischemic Circulatory Physiology, The University of Tokyo
| |
Collapse
|
39
|
Green tea minimally affects biomarkers of inflammation in obese subjects with metabolic syndrome. Nutrition 2010; 27:206-13. [PMID: 20605696 DOI: 10.1016/j.nut.2010.01.015] [Citation(s) in RCA: 130] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2009] [Revised: 01/12/2010] [Accepted: 01/22/2010] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Green tea (Camellia sinensis) has shown to exert cardioprotective benefits in observational studies. The objective of this clinical trial was to assess the effects of green tea on features of metabolic syndrome and inflammation in obese subjects. METHODS We conducted a randomized controlled trial in obese subjects with metabolic syndrome. Thirty-five subjects [(mean ± SE) age 42.5 ± 1.7 y, body mass index 36.1 ± 1.3 kg/m(2)] completed the 8-wk study and were randomly assigned to receive green tea (4 cups/d), green tea extract (2 capsules and 4 cups water/d), or no treatment (4 cups water/d). Both the beverage and extract groups had similar dosing of epigallocatechin-3-gallate, the active green tea polyphenol. Fasting blood samples were collected at screening, 4 and 8 wk of the study. RESULTS Green tea beverage or extract supplementation did not significantly alter features of metabolic syndrome or biomarkers of inflammation including adiponectin, C-reactive protein, interleukin-6, interleukin-1β, soluble vascular cell adhesion molecule-1, soluble intercellular adhesion molecule-1, leptin, or leptin:adiponectin ratio. However, both green tea beverage and extracts significantly reduced plasma serum amyloid alpha versus no treatment (P < 0.005). CONCLUSION This study suggests that the daily consumption of green tea beverage or extracts for 8 wk was well tolerated but did not affect the features of metabolic syndrome. However, green tea significantly reduced plasma serum amyloid alpha, an independent cardiovascular disease risk factor, in obese subjects with metabolic syndrome.
Collapse
|
40
|
Song C, Hsu K, Yamen E, Yan W, Fock J, Witting PK, Geczy CL, Freedman SB. Serum amyloid A induction of cytokines in monocytes/macrophages and lymphocytes. Atherosclerosis 2009; 207:374-83. [DOI: 10.1016/j.atherosclerosis.2009.05.007] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2008] [Revised: 05/04/2009] [Accepted: 05/06/2009] [Indexed: 11/28/2022]
|
41
|
Jenkins NT, Witkowski S, Spangenburg EE, Hagberg JM. Effects of acute and chronic endurance exercise on intracellular nitric oxide in putative endothelial progenitor cells: role of NAPDH oxidase. Am J Physiol Heart Circ Physiol 2009; 297:H1798-805. [PMID: 19717732 DOI: 10.1152/ajpheart.00347.2009] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
We sought to delineate the effects of acute and chronic exercise on the regulation of intracellular nitric oxide (NO(i)) production in putative endothelial progenitor cells (EPCs). Putative EPC colony-forming units (CFU-EC) were cultured from blood drawn before and after 30 min of treadmill exercise at 75% of maximal oxygen uptake in active (n = 8) and inactive (n = 8) men. CFU-EC were similar between groups at baseline, but increased after exercise in active men only (P = 0.04). CFU-EC expressed lower NADPH oxidase subunit gp91(phox) mRNA and elevated endothelial nitric oxide synthase mRNA in active relative to inactive men at baseline (P < 0.05). Acute exercise reduced gp91(phox) mRNA in CFU-EC of both groups (P < 0.05), whereas p47(phox) mRNA levels were reduced in the inactive group only (P = 0.02). There were no differences between groups or with acute exercise in xanthine oxidase, superoxide dismutase isoforms, or gluthathione peroxidase-1 mRNA levels. NO(i) was significantly greater in CFU-EC of active men at baseline (P = 0.004). NO(i) increased in CFU-EC of inactive men with acute exercise, and in vitro experiments with apocynin indicated the increased NO(i) production was caused by suppression of NADPH oxidase. However, the increases in NO(i) with the different treatments in the inactive group did not reach the baseline levels in the active group (P < 0.05). We conclude that acute exercise increases NO(i) in cells generated by the CFU-EC assay through an NADPH oxidase-inhibition mechanism in sedentary men. However, differences due to chronic exercise must involve additional factors. Our findings support exercise as a means to improve putative EPC function and suggest a novel mechanism that may explain this effect.
Collapse
Affiliation(s)
- Nathan T Jenkins
- Department of Kinesiology, School of Public Health, University of Maryland College Park. College Park, MD 20742, USA
| | | | | | | |
Collapse
|
42
|
Low-density lipoprotein apheresis as a treatment option for hyperlipidemia. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2009; 11:279-88. [DOI: 10.1007/s11936-009-0029-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
43
|
Duffy P, Wang X, Lin PH, Yao Q, Chen C. HIV Nef protein causes endothelial dysfunction in porcine pulmonary arteries and human pulmonary artery endothelial cells. J Surg Res 2009; 156:257-64. [PMID: 19540523 DOI: 10.1016/j.jss.2009.02.005] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2009] [Revised: 01/09/2009] [Accepted: 02/04/2009] [Indexed: 12/17/2022]
Abstract
BACKGROUND Infection of human immunodeficiency virus (HIV) has been associated with several chronic diseases, including pulmonary artery hypertension and atherosclerosis. However, the underlying mechanisms of these vascular complications are largely unknown. The objective of this study was to test a novel hypothesis that HIV Nef, an accessory HIV protein, may directly affect endothelial functions and gene expression in pulmonary arteries. METHODS Fresh porcine pulmonary artery rings and human pulmonary artery endothelial cells (HPAECs) were treated with HIV Nef for 24 h. With a myograph device, vasomotor function was determined with thromboxane A2 analog, U46619, for contraction, bradykinin, and sodium nitroprusside for relaxation. The expression of endothelial nitric oxide synthase (eNOS) was determined with real-time PCR and immunohistochemistry. Nitric oxide (NO) production was determined by Calorimetric Nitric Oxide Assay kit. Superoxide anion levels were detected with lucigenin-enhanced chemiluminescence assay and dihydroethidium (DHE) staining. RESULTS The endothelium-dependent vasorelaxation in response to bradykinin was significantly reduced in HIV Nef-treated porcine pulmonary artery rings in a concentration-dependent manner. In response to bradykinin (10(-8) mol/L), HIV Nef (10 ng/mL) significantly reduced vasorelaxation by 32% compared with untreated controls (P < 0.05). In addition, HIV Nef significantly decreased eNOS expression in the vessels and HPAECs. HIV Nef at 10 ng/mL significantly decreased NO production in HPAECs by 21% compared with controls (P < 0.05). Furthermore, HIV Nef significantly increased superoxide anion production in porcine pulmonary arteries and HPAECs compared with controls (P < 0.05). Consequently, Mn (III) tetrakis porphyrin, a superoxide dismutase mimic, effectively blocked HIV Nef-induced vasomotor dysfunction and superoxide anion production. The specificity of HIV Nef action was confirmed by anti-Nef antibody blocking and Nef heat inactivation. CONCLUSIONS HIV Nef protein significantly decreases endothelium-dependent vasorelaxation in porcine pulmonary arteries. It also reduces eNOS expression and induces oxidative stress in both porcine pulmonary arteries and HPAECs. This study demonstrates a new mechanism of HIV Nef, which causes endothelial dysfunction and may contribute to the human pulmonary artery disease in HIV-infected patients.
Collapse
Affiliation(s)
- Patrick Duffy
- Molecular Surgeon Research Center, Division of Vascular Surgery and Endovascular Therapy, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Michael E. DeBakey VA Medical Center, Houston, Texas 77030, USA
| | | | | | | | | |
Collapse
|