1
|
Cimini M, Hansmann UHE, Gonzalez C, Chesney AD, Truongcao MM, Gao E, Wang T, Roy R, Forte E, Mallaredy V, Thej C, Magadum A, Joladarashi D, Benedict C, Koch WJ, Tükel Ç, Kishore R. Podoplanin-positive cell-derived small extracellular vesicles contribute to cardiac amyloidosis after myocardial infarction. Cell Rep 2025; 44:115408. [PMID: 40056419 PMCID: PMC12019684 DOI: 10.1016/j.celrep.2025.115408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 12/26/2024] [Accepted: 02/17/2025] [Indexed: 03/10/2025] Open
Abstract
Cardiac amyloidosis is a secondary phenomenon of an already pre-existing chronic condition. Whether cardiac amyloidosis represents one of the complications post myocardial infarction (MI) has yet to be fully understood. Here, we show that amyloidosis occurs after MI and that amyloid fibers are composed of macrophage-derived serum amyloid A 3 (SAA3) monomers. SAA3 overproduction in macrophages is triggered by exosomal communication from cardiac stromal cells (CSCs), which, in response to MI, activate the expression of a platelet aggregation-inducing type I transmembrane glycoprotein, Podoplanin (PDPN). CSCPDPN+-derived small extracellular vesicles (sEVs) are enriched in SAA3, and exosomal SAA3 engages with macrophage by Toll-like receptor 2, triggering overproduction with consequent impaired clearance and aggregation of SAA3 monomers into rigid fibers. SAA3 amyloid deposits reduce cardiac contractility and increase scar stiffness. Inhibition of SAA3 aggregation by retro-inverso D-peptide, specifically designed to bind SAA3 monomers, prevents the deposition of SAA3 amyloid fibrils and improves heart function post MI.
Collapse
Affiliation(s)
- Maria Cimini
- Aging and Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Ulrich H E Hansmann
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK 73019-5251, USA
| | - Carolina Gonzalez
- Aging and Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Andrew D Chesney
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK 73019-5251, USA
| | - May M Truongcao
- Aging and Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Erhe Gao
- Aging and Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Tao Wang
- Aging and Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Rajika Roy
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | | | - Vandana Mallaredy
- Aging and Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Charan Thej
- Aging and Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Ajit Magadum
- Aging and Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Darukeshwara Joladarashi
- Aging and Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Cindy Benedict
- Aging and Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Water J Koch
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Çağla Tükel
- Center for Microbiology & Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Raj Kishore
- Aging and Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA.
| |
Collapse
|
2
|
Kim CH. Functional regulation of cytotoxic T cells by gut microbial metabolites. GUT MICROBES REPORTS 2025; 2:1-16. [PMID: 40115123 PMCID: PMC11922538 DOI: 10.1080/29933935.2025.2454002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/21/2024] [Accepted: 01/09/2025] [Indexed: 03/23/2025]
Abstract
Metabolites from gut microbes have a wide range of functions within the host body. One important function of these metabolites is to either positively or negatively control CD8+ cytotoxic T lymphocytes (CTLs), which can kill cancer and virus-infected cells. In healthy conditions, gut microbes produce a mixture of metabolites that promote CTL activity but also suppress excessive inflammatory responses. However, gut microbial dysbiosis occurs in patients with cancer, and this leads to changes in the production of gut microbial metabolites that can suppress CTL activity, promote inflammatory responses, and/or aid cancer growth. Decreased levels of CTL-promoting metabolites such as short-chain fatty acids, indole metabolites and polyamines but increased levels of CTL-suppressing metabolites, such as certain bile acids along with oncogenic metabolites, have been observed in patients with cancer. This review summarizes the altered production of major microbial metabolites in patients with cancer and discusses the impact of these changes on anti-cancer CTL responses.
Collapse
Affiliation(s)
- Chang H Kim
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI 48109; Mary H. Weiser Food Allergy Center, Center for Gastrointestinal Research, and Rogel Center for Cancer Research, University of Michigan School of Medicine, Ann Arbor, MI 48109
| |
Collapse
|
3
|
Ma C, Xu C, Zheng M, Zhang S, Liu Q, Lyu J, Pang X, Wang Y. Utilizing Lactic Acid Bacteria to Improve Hyperlipidemia: A Comprehensive Analysis from Gut Microbiota to Metabolic Pathways. Foods 2024; 13:4058. [PMID: 39767000 PMCID: PMC11675396 DOI: 10.3390/foods13244058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 11/29/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
Hyperlipidemia poses significant risks for cardiovascular diseases, with emerging evidence underscoring the critical role of gut microbiota in metabolic regulation. This study explores Lactobacillus casei CAAS36, a probiotic strain with promising cholesterol-lowering capabilities, assessing its impact on hyperlipidemic hamsters. Utilizing 1H NMR-based metabolomics and 16S rRNA gene sequencing, we observed that L. casei CAAS36 treatment not only altered metabolic pathways but also reshaped gut microbiota composition. Notably, the treatment restored the balance between Firmicutes and Bacteroidetes and significantly increased the abundance of propionate-producing Muribaculaceae. Metabolically, L. casei CAAS36 administration led to the normalization of key lipid markers, including reductions in total cholesterol, LDL-C, and triglycerides (29.9%, 29.4% and 32.6%), while enhancing the protective HDL-C levels. These effects were accompanied by significant increases in beneficial metabolites such as propionate and succinate, which are known for their roles in preventing metabolic disorders. These findings highlight the dual regulatory effects of L. casei CAAS36 on the metabolic profile and gut microbiota, suggesting a substantial potential for this probiotic in the management of hyperlipidemia and possibly other metabolic diseases. Future applications may include its use as a natural therapeutic agent in clinical settings, aiming to reduce reliance on conventional pharmaceuticals and their associated side effects.
Collapse
Affiliation(s)
- Changlu Ma
- Department of Food and Bio-Engineering, Beijing Vocational College of Agriculture, Beijing 102442, China;
- Institute of Food Science and Technology, Chinese Academy of Agricultural Science, Beijing 100193, China; (C.X.); (M.Z.); (S.Z.); (J.L.)
| | - Chen Xu
- Institute of Food Science and Technology, Chinese Academy of Agricultural Science, Beijing 100193, China; (C.X.); (M.Z.); (S.Z.); (J.L.)
| | - Mumin Zheng
- Institute of Food Science and Technology, Chinese Academy of Agricultural Science, Beijing 100193, China; (C.X.); (M.Z.); (S.Z.); (J.L.)
| | - Shuwen Zhang
- Institute of Food Science and Technology, Chinese Academy of Agricultural Science, Beijing 100193, China; (C.X.); (M.Z.); (S.Z.); (J.L.)
| | - Qifeng Liu
- State Key Laboratory for Bioactive Substances and Functions of Natural Medicines and Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China;
| | - Jiaping Lyu
- Institute of Food Science and Technology, Chinese Academy of Agricultural Science, Beijing 100193, China; (C.X.); (M.Z.); (S.Z.); (J.L.)
| | - Xiaoyang Pang
- Institute of Food Science and Technology, Chinese Academy of Agricultural Science, Beijing 100193, China; (C.X.); (M.Z.); (S.Z.); (J.L.)
| | - Yinghong Wang
- State Key Laboratory for Bioactive Substances and Functions of Natural Medicines and Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China;
| |
Collapse
|
4
|
Balasubramanian S, Perumal E. Integrated in silico analysis of transcriptomic alterations in nanoparticle toxicity across human and mouse models. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 949:174897. [PMID: 39053559 DOI: 10.1016/j.scitotenv.2024.174897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/17/2024] [Accepted: 07/17/2024] [Indexed: 07/27/2024]
Abstract
Nanoparticles, due to their exceptional physicochemical properties are used in our day-to-day environment. They are currently not regulated which might lead to increased levels in the biological systems causing adverse effects. However, the overall mechanism behind nanotoxicity remains elusive. Previously, we analysed the transcriptome datasets of copper oxide nanoparticles using in silico tools and identified IL-17, chemokine signaling pathway, and cytokine-cytokine receptor interaction as the key pathways altered. Based on the findings, we hypothesized a common pathway could be involved in transition metal oxide nanoparticles toxicity irrespective of the variables. Further, there could be unique transcriptome changes between metal oxide nanoparticles and other nanoparticles. To accomplish this, the overall transcriptome datasets of nanoparticles consisting of microarray and RNA-Seq were obtained. >90 studies for 17 different nanoparticles, performed in humans, rats, and mice were assessed. After initial screening, 24 mouse studies (with 196 datasets) and 34 human studies (with 200 datasets) were used for further analyses. The common genes that are dysregulated upon NPs exposure were identified for human and mouse datasets separately. Further, an overrepresentation functional enrichment analysis was performed. The common genes, their gene ontology, gene-gene, and protein-protein interactions were assessed. The overall results suggest that IL-17 and its related pathways might be commonly altered in nanoparticle exposure with lung as one of the major organs affected.
Collapse
Affiliation(s)
- Satheeswaran Balasubramanian
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, Tamil Nadu 641046, India
| | - Ekambaram Perumal
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, Tamil Nadu 641046, India.
| |
Collapse
|
5
|
Cimini M, Hansmann UHE, Gonzalez C, Chesney AD, Truongcao MM, Gao E, Wang T, Roy R, Forte E, Mallaredy V, Thej C, Magadum A, Joladarashi D, Benedict C, Koch WJ, Tükel Ç, Kishore R. Podoplanin Positive Cell-derived Extracellular Vesicles Contribute to Cardiac Amyloidosis After Myocardial Infarction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.28.601297. [PMID: 39005419 PMCID: PMC11244852 DOI: 10.1101/2024.06.28.601297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Background Amyloidosis is a major long-term complication of chronic disease; however, whether it represents one of the complications of post-myocardial infarction (MI) is yet to be fully understood. Methods Using wild-type and knocked-out MI mouse models and characterizing in vitro the exosomal communication between bone marrow-derived macrophages and activated mesenchymal stromal cells (MSC) isolated after MI, we investigated the mechanism behind Serum Amyloid A 3 (SAA3) protein overproduction in injured hearts. Results Here, we show that amyloidosis occurs after MI and that amyloid fibers are composed of macrophage-derived SAA3 monomers. SAA3 overproduction in macrophages is triggered by exosomal communication from a subset of activated MSC, which, in response to MI, acquire the expression of a platelet aggregation-inducing type I transmembrane glycoprotein named Podoplanin (PDPN). Cardiac MSC PDPN+ communicate with and activate macrophages through their extracellular vesicles or exosomes. Specifically, MSC PDPN+ derived exosomes (MSC PDPN+ Exosomes) are enriched in SAA3 and exosomal SAA3 protein engages with Toll-like receptor 2 (TRL2) on macrophages, triggering an overproduction and impaired clearance of SAA3 proteins, resulting in aggregation of SAA3 monomers as rigid amyloid deposits in the extracellular space. The onset of amyloid fibers deposition alongside extra-cellular-matrix (ECM) proteins in the ischemic heart exacerbates the rigidity and stiffness of the scar, hindering the contractility of viable myocardium and overall impairing organ function. Using SAA3 and TLR2 deficient mouse models, we show that SAA3 delivered by MSC PDPN+ exosomes promotes post-MI amyloidosis. Inhibition of SAA3 aggregation via administration of a retro-inverso D-peptide, specifically designed to bind SAA3 monomers, prevents the deposition of SAA3 amyloid fibrils, positively modulates the scar formation, and improves heart function post-MI. Conclusion Overall, our findings provide mechanistic insights into post-MI amyloidosis and suggest that SAA3 may be an attractive target for effective scar reversal after ischemic injury and a potential target in multiple diseases characterized by a similar pattern of inflammation and amyloid deposition. NOVELTY AND SIGNIFICANCE What is known? Accumulation of rigid amyloid structures in the left ventricular wall impairs ventricle contractility.After myocardial infarction cardiac Mesenchymal Stromal Cells (MSC) acquire Podoplanin (PDPN) to better interact with immune cells.Amyloid structures can accumulate in the heart after chronic inflammatory conditions. What information does this article contribute? Whether accumulation of cumbersome amyloid structures in the ischemic scar impairs left ventricle contractility, and scar reversal after myocardial infarction (MI) has never been investigated.The pathophysiological relevance of PDPN acquirement by MSC and the functional role of their secreted exosomes in the context of post-MI cardiac remodeling has not been investigated.Amyloid structures are present in the scar after ischemia and are composed of macrophage-derived Serum Amyloid A (SAA) 3 monomers, although mechanisms of SAA3 overproduction is not established. SUMMARY OF NOVELTY AND SIGNIFICANCE Here, we report that amyloidosis, a secondary phenomenon of an already preexisting and prolonged chronic inflammatory condition, occurs after MI and that amyloid structures are composed of macrophage-derived SAA3 monomers. Frequently studied cardiac amyloidosis are caused by aggregation of immunoglobulin light chains, transthyretin, fibrinogen, and apolipoprotein in a healthy heart as a consequence of systemic chronic inflammation leading to congestive heart failure with various types of arrhythmias and tissue stiffness. Although chronic MI is considered a systemic inflammatory condition, studies regarding the possible accumulation of amyloidogenic proteins after MI and the mechanisms involved in that process are yet to be reported. Here, we show that SAA3 overproduction in macrophages is triggered in a Toll-like Receptor 2 (TLR2)-p38MAP Kinase-dependent manner by exosomal communication from a subset of activated MSC, which, in response to MI, express a platelet aggregation-inducing type I transmembrane glycoprotein named Podoplanin. We provide the full mechanism of this phenomenon in murine models and confirm SAA3 amyloidosis in failing human heart samples. Moreover, we developed a retro-inverso D-peptide therapeutic approach, "DRI-R5S," specifically designed to bind SAA3 monomers and prevent post-MI aggregation and deposition of SAA3 amyloid fibrils without interfering with the innate immune response.
Collapse
|
6
|
Erickson MA, Mahankali AP. Interactions of Serum Amyloid A Proteins with the Blood-Brain Barrier: Implications for Central Nervous System Disease. Int J Mol Sci 2024; 25:6607. [PMID: 38928312 PMCID: PMC11204325 DOI: 10.3390/ijms25126607] [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: 04/15/2024] [Revised: 05/30/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Serum amyloid A (SAA) proteins are highly conserved lipoproteins that are notoriously involved in the acute phase response and systemic amyloidosis, but their biological functions are incompletely understood. Recent work has shown that SAA proteins can enter the brain by crossing the intact blood-brain barrier (BBB), and that they can impair BBB functions. Once in the central nervous system (CNS), SAA proteins can have both protective and harmful effects, which have important implications for CNS disease. In this review of the thematic series on SAA, we discuss the existing literature that relates SAA to neuroinflammation and CNS disease, and the possible roles of the BBB in these relations.
Collapse
Affiliation(s)
- Michelle A. Erickson
- Geriatric Research Education and Clinical Center, VA Puget Sound Healthcare System, Seattle, WA 98108, USA
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington School of Medicine, Seattle, WA 98104, USA;
| | - Anvitha P. Mahankali
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington School of Medicine, Seattle, WA 98104, USA;
| |
Collapse
|
7
|
Huang S, Jiang Y, Li J, Mao L, Qiu Z, Zhang S, Jiang Y, Liu Y, Liu W, Xiong Z, Zhang W, Liu X, Zhang Y, Bai X, Guo B. Osteocytes/Osteoblasts Produce SAA3 to Regulate Hepatic Metabolism of Cholesterol. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307818. [PMID: 38613835 PMCID: PMC11199997 DOI: 10.1002/advs.202307818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 03/19/2024] [Indexed: 04/15/2024]
Abstract
Hypercholesterolaemia is a systemic metabolic disease, but the role of organs other than liver in cholesterol metabolism is unappreciated. The phenotypic characterization of the Tsc1Dmp1 mice reveal that genetic depletion of tuberous sclerosis complex 1 (TSC1) in osteocytes/osteoblasts (Dmp1-Cre) triggers progressive increase in serum cholesterol level. The resulting cholesterol metabolic dysregulation is shown to be associated with upregulation and elevation of serum amyloid A3 (SAA3), a lipid metabolism related factor, in the bone and serum respectively. SAA3, elicited from the bone, bound to toll-like receptor 4 (TLR4) on hepatocytes to phosphorylate c-Jun, and caused impeded conversion of cholesterol to bile acids via suppression on cholesterol 7 α-hydroxylase (Cyp7a1) expression. Ablation of Saa3 in Tsc1Dmp1 mice prevented the CYP7A1 reduction in liver and cholesterol elevation in serum. These results expand the understanding of bone function and hepatic regulation of cholesterol metabolism and uncover a potential therapeutic use of pharmacological modulation of SAA3 in hypercholesterolaemia.
Collapse
Affiliation(s)
- Shijiang Huang
- State Key Laboratory of Organ Failure ResearchDepartment of Cell BiologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Yuanjun Jiang
- State Key Laboratory of Organ Failure ResearchDepartment of Cell BiologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Jing Li
- Department of Obstetrics and GynecologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Linlin Mao
- State Key Laboratory of Organ Failure ResearchDepartment of Cell BiologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Zeyou Qiu
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
- Equipment Material DepartmentWest China Xiamen Hospital of Sichuan UniversityXiamenFujian361000China
| | - Sheng Zhang
- State Key Laboratory of Organ Failure ResearchDepartment of Cell BiologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Yuhui Jiang
- State Key Laboratory of Organ Failure ResearchDepartment of Cell BiologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Yong Liu
- State Key Laboratory of Organ Failure ResearchDepartment of Cell BiologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Wen Liu
- State Key Laboratory of Organ Failure ResearchDepartment of Cell BiologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Zhi Xiong
- State Key Laboratory of Organ Failure ResearchDepartment of Cell BiologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Wuju Zhang
- State Key Laboratory of Organ Failure ResearchDepartment of Cell BiologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
- Central LaboratoryThe Fifth Affiliated HospitalSouthern Medical UniversityGuangzhouGuangdong510900China
| | - Xiaolin Liu
- State Key Laboratory of Organ Failure ResearchDepartment of Cell BiologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Yue Zhang
- State Key Laboratory of Organ Failure ResearchDepartment of Cell BiologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Xiaochun Bai
- State Key Laboratory of Organ Failure ResearchDepartment of Cell BiologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
- Guangdong Provincial Key Laboratory of Bone and Joint Degenerative DiseasesThe Third Affiliated Hospital of Southern Medical UniversityGuangzhouGuangdong510630China
| | - Bin Guo
- State Key Laboratory of Organ Failure ResearchDepartment of Cell BiologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
- The Tenth Affiliated HospitalSouthern Medical UniversityDongguanGuangdong523018China
| |
Collapse
|
8
|
Shridas P, Ji A, Trumbauer AC, Noffsinger VP, Meredith LW, de Beer FC, Mullick AE, Webb NR, Karounos DG, Tannock LR. Antisense oligonucleotide targeting hepatic Serum Amyloid A limits the progression of angiotensin II-induced abdominal aortic aneurysm formation. Atherosclerosis 2024; 391:117492. [PMID: 38461759 PMCID: PMC11006562 DOI: 10.1016/j.atherosclerosis.2024.117492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 02/07/2024] [Accepted: 02/23/2024] [Indexed: 03/12/2024]
Abstract
BACKGROUND AND AIMS Obesity increases the risk for abdominal aortic aneurysms (AAA) in humans and enhances angiotensin II (AngII)-induced AAA formation in C57BL/6 mice. We reported that deficiency of Serum Amyloid A (SAA) significantly reduces AngII-induced inflammation and AAA in both hyperlipidemic apoE-deficient and obese C57BL/6 mice. The aim of this study is to investigate whether SAA plays a role in the progression of early AAA in obese C57BL/6 mice. METHODS Male C57BL/6J mice were fed a high-fat diet (60% kcal as fat) throughout the study. After 4 months of diet, the mice were infused with AngII until the end of the study. Mice with at least a 25% increase in the luminal diameter of the abdominal aorta after 4 weeks of AngII infusion were stratified into 2 groups. The first group received a control antisense oligonucleotide (Ctr ASO), and the second group received ASO that suppresses SAA (SAA-ASO) until the end of the study. RESULTS Plasma SAA levels were significantly reduced by the SAA ASO treatment. While mice that received the control ASO had continued aortic dilation throughout the AngII infusion periods, the mice that received SAA-ASO had a significant reduction in the progression of aortic dilation, which was associated with significant reductions in matrix metalloprotease activities, decreased macrophage infiltration and decreased elastin breaks in the abdominal aortas. CONCLUSIONS We demonstrate for the first time that suppression of SAA protects obese C57BL/6 mice from the progression of AngII-induced AAA. Suppression of SAA may be a therapeutic approach to limit AAA progression.
Collapse
Affiliation(s)
- Preetha Shridas
- Department of Internal Medicine, University of Kentucky, Lexington, 40536, Kentucky, USA; Saha Cardiovascular Research Center, University of Kentucky, Lexington, 40536, Kentucky, USA.
| | - Ailing Ji
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, 40536, Kentucky, USA
| | - Andrea C Trumbauer
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, 40536, Kentucky, USA
| | - Victoria P Noffsinger
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, 40536, Kentucky, USA
| | - Luke W Meredith
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, 40536, Kentucky, USA
| | - Frederick C de Beer
- Department of Internal Medicine, University of Kentucky, Lexington, 40536, Kentucky, USA; Saha Cardiovascular Research Center, University of Kentucky, Lexington, 40536, Kentucky, USA
| | | | - Nancy R Webb
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, 40536, Kentucky, USA; Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, 40536, Kentucky, USA
| | - Dennis G Karounos
- Department of Internal Medicine, University of Kentucky, Lexington, 40536, Kentucky, USA; Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, 40536, Kentucky, USA; Department of Veterans Affairs, Lexington, 40536, Kentucky, USA
| | - Lisa R Tannock
- Department of Internal Medicine, University of Kentucky, Lexington, 40536, Kentucky, USA; Saha Cardiovascular Research Center, University of Kentucky, Lexington, 40536, Kentucky, USA
| |
Collapse
|
9
|
Verstraelen P, Van Remoortel S, De Loose N, Verboven R, Garcia-Diaz Barriga G, Christmann A, Gries M, Bessho S, Li J, Guerra C, Tükel Ç, Martinez SI, Schäfer KH, Timmermans JP, De Vos WH. Serum Amyloid A3 Fuels a Feed-Forward Inflammatory Response to the Bacterial Amyloid Curli in the Enteric Nervous System. Cell Mol Gastroenterol Hepatol 2024; 18:89-104. [PMID: 38556049 PMCID: PMC11127031 DOI: 10.1016/j.jcmgh.2024.03.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 03/08/2024] [Accepted: 03/18/2024] [Indexed: 04/02/2024]
Abstract
BACKGROUND & AIMS Mounting evidence suggests the gastrointestinal microbiome is a determinant of peripheral immunity and central neurodegeneration, but the local disease mechanisms remain unknown. Given its potential relevance for early diagnosis and therapeutic intervention, we set out to map the pathogenic changes induced by bacterial amyloids in the gastrointestinal tract and its enteric nervous system. METHODS To examine the early response, we challenged primary murine myenteric networks with curli, the prototypical bacterial amyloid, and performed shotgun RNA sequencing and multiplex enzyme-linked immunosorbent assay. Using enteric neurosphere-derived glial and neuronal cell cultures, as well as in vivo curli injections into the colon wall, we further scrutinized curli-induced pathogenic pathways. RESULTS Curli induced a proinflammatory response, with strong up-regulation of Saa3 and the secretion of several cytokines. This proinflammatory state was induced primarily in enteric glia, was accompanied by increased levels of DNA damage and replication, and triggered the influx of immune cells in vivo. The addition of recombinant Serum Amyloid A3 (SAA3) was sufficient to recapitulate this specific proinflammatory phenotype while Saa3 knock-out attenuated curli-induced DNA damage and replication. Similar to curli, recombinant SAA3 caused a strong up-regulation of Saa3 transcripts, illustrating its self-amplifying potential . Since colonization of curli-producing Salmonella and dextran sulfate sodium-induced colitis triggered a significant increase in Saa3 transcripts as well, we assume SAA3plays a central role in enteric dysfunction. Inhibition of dual leucine zipper kinase, an upstream regulator of the c-Jun N-terminal kinase pathway responsible for SAA3 production, attenuated curli- and recombinant SAA3-induced Saa3 up-regulation, DNA damage, and replication in enteric glia. CONCLUSIONS Our results position SAA3 as an important mediator of gastrointestinal vulnerability to bacterial-derived amyloids and demonstrate the potential of dual leucine zipper kinase inhibition to dampen enteric pathology.
Collapse
Affiliation(s)
- Peter Verstraelen
- Laboratory of Cell Biology and Histology, University of Antwerp, Wilrijk, Belgium
| | - Samuel Van Remoortel
- Laboratory of Cell Biology and Histology, University of Antwerp, Wilrijk, Belgium
| | - Nouchin De Loose
- Laboratory of Cell Biology and Histology, University of Antwerp, Wilrijk, Belgium
| | - Rosanne Verboven
- Laboratory of Cell Biology and Histology, University of Antwerp, Wilrijk, Belgium
| | | | - Anne Christmann
- Working Group Enteric Nervous System, University of Applied Sciences Kaiserslautern, Zweibrücken, Germany
| | - Manuela Gries
- Working Group Enteric Nervous System, University of Applied Sciences Kaiserslautern, Zweibrücken, Germany
| | - Shingo Bessho
- Center for Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Jing Li
- Experimental Oncology Group, Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
| | - Carmen Guerra
- Experimental Oncology Group, Centro Nacional de Investigaciones Oncológicas, Madrid, Spain; Centro de Investigación Biomédica en Red de Cáncer, Instituto de Salud Carlos III, Madrid, Spain
| | - Çagla Tükel
- Center for Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Sales Ibiza Martinez
- Laboratory of Cell Biology and Histology, University of Antwerp, Wilrijk, Belgium
| | - Karl-Herbert Schäfer
- Working Group Enteric Nervous System, University of Applied Sciences Kaiserslautern, Zweibrücken, Germany
| | - Jean-Pierre Timmermans
- Laboratory of Cell Biology and Histology, University of Antwerp, Wilrijk, Belgium; Antwerp Centre for Advanced Microscopy, University of Antwerp, Antwerp, Belgium; μNeuro Research Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Winnok H De Vos
- Laboratory of Cell Biology and Histology, University of Antwerp, Wilrijk, Belgium; Antwerp Centre for Advanced Microscopy, University of Antwerp, Antwerp, Belgium; μNeuro Research Centre of Excellence, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
10
|
Shridas P, Ji A, Trumbauer AC, Noffsinger VP, Meredith LW, de Beer FC, Mullick AE, Webb NR, Karounos DG, Tannock LR. Antisense Oligonucleotide Targeting Hepatic Serum Amyloid A Limits the Progression of Angiotensin II-Induced Abdominal Aortic Aneurysm Formation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.22.554377. [PMID: 37662383 PMCID: PMC10473661 DOI: 10.1101/2023.08.22.554377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
OBJECTIVE Obesity increases the risk for abdominal aortic aneurysms (AAA) in humans and enhances angiotensin II (AngII)-induced AAA formation in C57BL/6 mice. Obesity is also associated with increases in serum amyloid A (SAA). We previously reported that deficiency of SAA significantly reduces AngII-induced inflammation and AAA in both hyperlipidemic apoE-deficient and obese C57BL/6 mice. In this study, we investigated whether SAA plays a role in the progression of early AAA in obese C57BL/6 mice. APPROACH AND RESULTS Male C57BL/6J mice were fed a high-fat diet (60% kcal as fat) throughout the study. After 4 months of diet, the mice were infused with angiotensin II (AngII) until the end of the study. Mice with at least a 25% increase in the luminal diameter of the abdominal aorta after 4 weeks of AngII infusion were stratified into 2 groups. The first group received a control antisense oligonucleotide (Ctr ASO), and the second group received ASO that suppresses SAA (SAA-ASO) until the end of the study. Plasma SAA levels were significantly reduced by the SAA ASO treatment. While mice that received the control ASO had continued aortic dilation throughout the AngII infusion periods, the mice that received SAA-ASO had a significant reduction in the progression of aortic dilation, which was associated with significant reductions in matrix metalloprotease activities, decreased macrophage infiltration and decreased elastin breaks in the abdominal aortas. CONCLUSION We demonstrate for the first time that suppression of SAA protects obese C57BL/6 mice from the progression of AngII-induced AAA. Suppression of SAA may be a therapeutic approach to limit AAA progression.
Collapse
|
11
|
Li X, Morel JD, Benegiamo G, Poisson J, Bachmann A, Rapin A, Sulc J, Williams E, Perino A, Schoonjans K, Bou Sleiman M, Auwerx J. Genetic and dietary modulators of the inflammatory response in the gastrointestinal tract of the BXD mouse genetic reference population. eLife 2023; 12:RP87569. [PMID: 37855835 PMCID: PMC10586803 DOI: 10.7554/elife.87569] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2023] Open
Abstract
Inflammatory gut disorders, including inflammatory bowel disease (IBD), can be impacted by dietary, environmental, and genetic factors. While the incidence of IBD is increasing worldwide, we still lack a complete understanding of the gene-by-environment interactions underlying inflammation and IBD. Here, we profiled the colon transcriptome of 52 BXD mouse strains fed with a chow or high-fat diet (HFD) and identified a subset of BXD strains that exhibit an IBD-like transcriptome signature on HFD, indicating that an interplay of genetics and diet can significantly affect intestinal inflammation. Using gene co-expression analyses, we identified modules that are enriched for IBD-dysregulated genes and found that these IBD-related modules share cis-regulatory elements that are responsive to the STAT2, SMAD3, and REL transcription factors. We used module quantitative trait locus analyses to identify genetic loci associated with the expression of these modules. Through a prioritization scheme involving systems genetics in the mouse and integration with external human datasets, we identified Muc4 and Epha6 as the top candidates mediating differences in HFD-driven intestinal inflammation. This work provides insights into the contribution of genetics and diet to IBD risk and identifies two candidate genes, MUC4 and EPHA6, that may mediate IBD susceptibility in humans.
Collapse
Affiliation(s)
- Xiaoxu Li
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de LausanneLausanneSwitzerland
| | - Jean-David Morel
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de LausanneLausanneSwitzerland
| | - Giorgia Benegiamo
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de LausanneLausanneSwitzerland
| | - Johanne Poisson
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de LausanneLausanneSwitzerland
| | - Alexis Bachmann
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de LausanneLausanneSwitzerland
| | - Alexis Rapin
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de LausanneLausanneSwitzerland
| | - Jonathan Sulc
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de LausanneLausanneSwitzerland
| | - Evan Williams
- Luxembourg Centre for Systems Biomedicine, University of LuxembourgEsch-sur-AlzetteLuxembourg
| | - Alessia Perino
- Laboratory of Metabolic Signaling, Institute of Bioengineering, École Polytechnique Fédérale de LausanneLausanneSwitzerland
| | - Kristina Schoonjans
- Laboratory of Metabolic Signaling, Institute of Bioengineering, École Polytechnique Fédérale de LausanneLausanneSwitzerland
| | - Maroun Bou Sleiman
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de LausanneLausanneSwitzerland
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de LausanneLausanneSwitzerland
| |
Collapse
|
12
|
Oh M, Jang SY, Lee JY, Kim JW, Jung Y, Kim J, Seo J, Han TS, Jang E, Son HY, Kim D, Kim MW, Park JS, Song KH, Oh KJ, Kim WK, Bae KH, Huh YM, Kim SH, Kim D, Han BS, Lee SC, Hwang GS, Lee EW. The lipoprotein-associated phospholipase A2 inhibitor Darapladib sensitises cancer cells to ferroptosis by remodelling lipid metabolism. Nat Commun 2023; 14:5728. [PMID: 37714840 PMCID: PMC10504358 DOI: 10.1038/s41467-023-41462-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 08/29/2023] [Indexed: 09/17/2023] Open
Abstract
Arachidonic and adrenic acids in the membrane play key roles in ferroptosis. Here, we reveal that lipoprotein-associated phospholipase A2 (Lp-PLA2) controls intracellular phospholipid metabolism and contributes to ferroptosis resistance. A metabolic drug screen reveals that darapladib, an inhibitor of Lp-PLA2, synergistically induces ferroptosis in the presence of GPX4 inhibitors. We show that darapladib is able to enhance ferroptosis under lipoprotein-deficient or serum-free conditions. Furthermore, we find that Lp-PLA2 is located in the membrane and cytoplasm and suppresses ferroptosis, suggesting a critical role for intracellular Lp-PLA2. Lipidomic analyses show that darapladib treatment or deletion of PLA2G7, which encodes Lp-PLA2, generally enriches phosphatidylethanolamine species and reduces lysophosphatidylethanolamine species. Moreover, combination treatment of darapladib with the GPX4 inhibitor PACMA31 efficiently inhibits tumour growth in a xenograft model. Our study suggests that inhibition of Lp-PLA2 is a potential therapeutic strategy to enhance ferroptosis in cancer treatment.
Collapse
Affiliation(s)
- Mihee Oh
- Biodefense Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea
| | - Seo Young Jang
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul, 03759, Korea
| | - Ji-Yoon Lee
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea
| | - Jong Woo Kim
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea
- Department of Functional Genomics, University of Science and Technology (UST), Daejeon, 34141, Korea
| | - Youngae Jung
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul, 03759, Korea
| | - Jiwoo Kim
- Therapeutics and Biotechnology Department, Drug Discovery Platform Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon, 34114, Korea
- Graduate School of New Drug Discovery and Development, Chungnam National University, Daejeon, 305-764, Korea
| | - Jinho Seo
- Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea
| | - Tae-Su Han
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea
| | - Eunji Jang
- MediBio-Informatics Research Center, Novomics Co., Ltd., Seoul, Korea
| | - Hye Young Son
- YUHS-KRIBB Medical Convergence Research Institute, Seoul, 03722, Korea
- Department of Radiology, College of Medicine, Yonsei University, Seoul, 03722, Korea
| | - Dain Kim
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul, 03759, Korea
- Department of Life Science, Ewha Womans University, Seoul, 03760, Korea
| | - Min Wook Kim
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea
| | | | - Kwon-Ho Song
- Department of Cell Biology, Daegu Catholic University School of Medicine, Daegu, 42472, Korea
| | - Kyoung-Jin Oh
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea
- Department of Functional Genomics, University of Science and Technology (UST), Daejeon, 34141, Korea
| | - Won Kon Kim
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea
- Department of Functional Genomics, University of Science and Technology (UST), Daejeon, 34141, Korea
| | - Kwang-Hee Bae
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea
- Department of Functional Genomics, University of Science and Technology (UST), Daejeon, 34141, Korea
| | - Yong-Min Huh
- MediBio-Informatics Research Center, Novomics Co., Ltd., Seoul, Korea
- YUHS-KRIBB Medical Convergence Research Institute, Seoul, 03722, Korea
- Department of Radiology, College of Medicine, Yonsei University, Seoul, 03722, Korea
| | - Soon Ha Kim
- MitoImmune Therapeutics Inc., Seoul, 06123, Korea
| | - Doyoun Kim
- Therapeutics and Biotechnology Department, Drug Discovery Platform Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon, 34114, Korea
| | - Baek-Soo Han
- Biodefense Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea.
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea.
- Department of Functional Genomics, University of Science and Technology (UST), Daejeon, 34141, Korea.
| | - Sang Chul Lee
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea.
| | - Geum-Sook Hwang
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul, 03759, Korea.
- College of Pharmacy, Chung-Ang University, Seoul, 06974, Korea.
| | - Eun-Woo Lee
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea.
- Department of Functional Genomics, University of Science and Technology (UST), Daejeon, 34141, Korea.
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Korea.
| |
Collapse
|
13
|
Wang MM, Gai MT, Wang BZ, Maituxun M, Yesitayi G, Chen BD, Ma X. The diagnostic and prognostic value of SAA1 as a novel biomarker for acute aortic dissection. J Proteomics 2023; 286:104958. [PMID: 37422110 DOI: 10.1016/j.jprot.2023.104958] [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: 11/15/2022] [Revised: 06/20/2023] [Accepted: 07/01/2023] [Indexed: 07/10/2023]
Abstract
BACKGROUND AND AIMS Acute aortic dissection (AAD) is a serious life-threatening cardiovascular condition. It is necessary to find rapid and accurate biomarkers for the diagnosis of AAD. This study aimed to determine the efficacy of serum amyloid A1 (SAA1) in the diagnosis and prediction of long-term adverse events in AAD. MATERIALS AND METHODS Four-dimensional label-free quantification (4D-LFQ) technique was used to identify the differentially expressed proteins (DEPs) in aortic tissues of AAD. After comprehensive analysis, SAA1 was identified as a potential biomarker of AAD. ELISA was used to confirm the expression of SAA1 in serum of AAD patients. Moreover, the source of SAA1 in serum was explored by constructing AAD mouse model. RESULTS A total of 247 DEPs were identified, of which 139 were upregulated while 108 were downregulated. SAA1 was nearly 6.4-fold and 4.5-fold upregulated in AAD tissue and serum. ROC curve and Kaplan-Meier survival curve confirmed the good efficacy of SAA1 for the diagnosis and prediction of long-term adverse events in AAD. In vivo experiments revealed that SAA1 was mainly derived from the liver when AAD occurred. CONCLUSION SAA1 can be used as a potential biomarker for AAD with effective diagnostic and prognostic value. SIGNIFICANCE Despite the advances in medical technology in recent years, the mortality rate of acute aortic dissection (AAD) is still high. It is still challenging for clinicians to diagnose AAD patients on time and reduce the mortality rate. In this study, 4D-LFQ technology was used to identify serum amyloid A1 (SAA1) as a potential biomarker of AAD and was verified in subsequent work. The results of this study determined the efficacy of SAA1 in the diagnosis and prediction of long-term adverse events in patients with AAD.
Collapse
Affiliation(s)
- Meng-Meng Wang
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China; Xinjiang Key Laboratory of Cardiovascular Disease, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, China
| | - Min-Tao Gai
- Xinjiang Key Laboratory of Cardiovascular Disease, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, China
| | - Bao-Zhu Wang
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Maitudi Maituxun
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Gulinazi Yesitayi
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Bang-Dang Chen
- Xinjiang Key Laboratory of Cardiovascular Disease, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, China.
| | - Xiang Ma
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China.
| |
Collapse
|
14
|
den Hartigh LJ, May KS, Zhang XS, Chait A, Blaser MJ. Serum amyloid A and metabolic disease: evidence for a critical role in chronic inflammatory conditions. Front Cardiovasc Med 2023; 10:1197432. [PMID: 37396595 PMCID: PMC10311072 DOI: 10.3389/fcvm.2023.1197432] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 05/15/2023] [Indexed: 07/04/2023] Open
Abstract
Serum amyloid A (SAA) subtypes 1-3 are well-described acute phase reactants that are elevated in acute inflammatory conditions such as infection, tissue injury, and trauma, while SAA4 is constitutively expressed. SAA subtypes also have been implicated as playing roles in chronic metabolic diseases including obesity, diabetes, and cardiovascular disease, and possibly in autoimmune diseases such as systemic lupus erythematosis, rheumatoid arthritis, and inflammatory bowel disease. Distinctions between the expression kinetics of SAA in acute inflammatory responses and chronic disease states suggest the potential for differentiating SAA functions. Although circulating SAA levels can rise up to 1,000-fold during an acute inflammatory event, elevations are more modest (∼5-fold) in chronic metabolic conditions. The majority of acute-phase SAA derives from the liver, while in chronic inflammatory conditions SAA also derives from adipose tissue, the intestine, and elsewhere. In this review, roles for SAA subtypes in chronic metabolic disease states are contrasted to current knowledge about acute phase SAA. Investigations show distinct differences between SAA expression and function in human and animal models of metabolic disease, as well as sexual dimorphism of SAA subtype responses.
Collapse
Affiliation(s)
- Laura J. den Hartigh
- Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition, University of Washington, Seattle, WA, United States
- Diabetes Institute, University of Washington, Seattle, WA, United States
| | - Karolline S. May
- Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition, University of Washington, Seattle, WA, United States
- Diabetes Institute, University of Washington, Seattle, WA, United States
| | - Xue-Song Zhang
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, United States
| | - Alan Chait
- Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition, University of Washington, Seattle, WA, United States
- Diabetes Institute, University of Washington, Seattle, WA, United States
| | - Martin J. Blaser
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, United States
| |
Collapse
|
15
|
Wang Y, Xiao N, Hu L, Deng M, Zong F, Zhang Z, Su D, Zhou D, Yang H, Dai E. Mechanism of pulmonary plague biphasic syndrome: inhibition or activation of NF-κB signaling pathway. Future Microbiol 2023; 18:267-286. [PMID: 36971082 DOI: 10.2217/fmb-2023-0009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023] Open
Abstract
Background: Pneumonic plague is a fatal respiratory disease caused by Yersinia pestis. Time-course transcriptome analysis on the mechanism of pneumonic plague biphasic syndrome is lacking in the literature. Materials & methods: This study documented the disease course through bacterial load, histopathology, cytokine levels and flow cytometry. RNA-sequencing technology was used to investigate the global transcriptome profile of lung tissue in mice infected with Y. pestis. Results: Inflammation-related genes were significantly upregulated at 48 h post-infection, while genes related to cell adhesion and cytoskeletal structure were downregulated. Conclusion: NOD-like receptor and TNF signaling pathways play a plausible role in pneumonic plague biphasic syndrome and lung injury by controlling the activation and inhibition of the NF-κB signaling pathway.
Collapse
|
16
|
Wang Y, Groeger S, Yong J, Ruf S. Orthodontic Compression Enhances Macrophage M2 Polarization via Histone H3 Hyperacetylation. Int J Mol Sci 2023; 24:ijms24043117. [PMID: 36834533 PMCID: PMC9958841 DOI: 10.3390/ijms24043117] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/27/2023] [Accepted: 01/31/2023] [Indexed: 02/08/2023] Open
Abstract
Orthodontic tooth movement is a complex periodontal remodeling process triggered by compression that involves sterile inflammation and immune responses. Macrophages are mechanically sensitive immune cells, but their role in orthodontic tooth movement is unclear. Here, we hypothesize that orthodontic force can activate macrophages, and their activation may be associated with orthodontic root resorption. After force-loading and/or adiponectin application, the migration function of macrophages was tested via scratch assay, and Nos2, Il1b, Arg1, Il10, ApoE, and Saa3 expression levels were detected using qRT-PCR. Furthermore, H3 histone acetylation was measured using an acetylation detection kit. The specific inhibitor of H3 histone, I-BET762, was deployed to observe its effect on macrophages. In addition, cementoblasts were treated with macrophage-conditioned medium or compression force, and OPG production and cellular migration were measured. We further detected Piezo1 expression in cementoblasts via qRT-PCR and Western-blot, and its effect on the force-induced impairment of cementoblastic functions was also analyzed. Compressive force significantly inhibited macrophage migration. Nos2 was up-regulated 6 h after force-loading. Il1b, Arg1, Il10, Saa3, and ApoE increased after 24 h. Meanwhile, higher H3 histone acetylation was detected in the macrophages subjected to compression, and I-BET762 dampened the expression of M2 polarization markers (Arg1 and Il10). Lastly, even though the activated macrophage-conditioned medium showed no effect on cementoblasts, compressive force directly impaired cementoblastic function by enhancing mechanoreceptor Piezo1. Compressive force activates macrophages; specifically, it causes M2 polarization via H3 histone acetylation in the late stage. Compression-induced orthodontic root resorption is macrophage-independent, but it involves the activation of mechanoreceptor Piezo1.
Collapse
Affiliation(s)
- Yao Wang
- Department of Orthodontics, Faculty of Medicine, Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Sabine Groeger
- Department of Orthodontics, Faculty of Medicine, Justus Liebig University Giessen, 35392 Giessen, Germany
- Department of Periodontology, Faculty of Medicine, Justus Liebig University Giessen, 35392 Giessen, Germany
- Correspondence:
| | - Jiawen Yong
- Department of Orthodontics, Faculty of Medicine, Justus Liebig University Giessen, 35392 Giessen, Germany
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310003, China
| | - Sabine Ruf
- Department of Orthodontics, Faculty of Medicine, Justus Liebig University Giessen, 35392 Giessen, Germany
| |
Collapse
|
17
|
Gutierrez CT, Loizides C, Hafez I, Brostrøm A, Wolff H, Szarek J, Berthing T, Mortensen A, Jensen KA, Roursgaard M, Saber AT, Møller P, Biskos G, Vogel U. Acute phase response following pulmonary exposure to soluble and insoluble metal oxide nanomaterials in mice. Part Fibre Toxicol 2023; 20:4. [PMID: 36650530 PMCID: PMC9843849 DOI: 10.1186/s12989-023-00514-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 01/10/2023] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Acute phase response (APR) is characterized by a change in concentration of different proteins, including C-reactive protein and serum amyloid A (SAA) that can be linked to both exposure to metal oxide nanomaterials and risk of cardiovascular diseases. In this study, we intratracheally exposed mice to ZnO, CuO, Al2O3, SnO2 and TiO2 and carbon black (Printex 90) nanomaterials with a wide range in phagolysosomal solubility. We subsequently assessed neutrophil numbers, protein and lactate dehydrogenase activity in bronchoalveolar lavage fluid, Saa3 and Saa1 mRNA levels in lung and liver tissue, respectively, and SAA3 and SAA1/2 in plasma. Endpoints were analyzed 1 and 28 days after exposure, including histopathology of lung and liver tissues. RESULTS All nanomaterials induced pulmonary inflammation after 1 day, and exposure to ZnO, CuO, SnO2, TiO2 and Printex 90 increased Saa3 mRNA levels in lungs and Saa1 mRNA levels in liver. Additionally, CuO, SnO2, TiO2 and Printex 90 increased plasma levels of SAA3 and SAA1/2. Acute phase response was predicted by deposited surface area for insoluble metal oxides, 1 and 28 days post-exposure. CONCLUSION Soluble and insoluble metal oxides induced dose-dependent APR with different time dependency. Neutrophil influx, Saa3 mRNA levels in lung tissue and plasma SAA3 levels correlated across all studied nanomaterials, suggesting that these endpoints can be used as biomarkers of acute phase response and cardiovascular disease risk following exposure to soluble and insoluble particles.
Collapse
Affiliation(s)
- Claudia Torero Gutierrez
- grid.5254.60000 0001 0674 042XSection of Environmental Health, Department of Public Health, University of Copenhagen, Copenhagen, Denmark ,grid.418079.30000 0000 9531 3915National Research Centre for the Working Environment, Copenhagen, Denmark
| | - Charis Loizides
- grid.426429.f0000 0004 0580 3152Atmosphere and Climate Research Centre, The Cyprus Institute, Nicosia, Cyprus
| | - Iosif Hafez
- grid.426429.f0000 0004 0580 3152Atmosphere and Climate Research Centre, The Cyprus Institute, Nicosia, Cyprus
| | - Anders Brostrøm
- grid.5170.30000 0001 2181 8870National Centre for Nano Fabrication and Characterization, Technical University of Denmark, Copenhagen, Denmark
| | - Henrik Wolff
- grid.6975.d0000 0004 0410 5926Finnish Institute of Occupational Health, Helsinki, Finland
| | - Józef Szarek
- grid.412607.60000 0001 2149 6795Department of Pathophysiology, Forensic Veterinary Medicine and Administration, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Trine Berthing
- grid.418079.30000 0000 9531 3915National Research Centre for the Working Environment, Copenhagen, Denmark
| | - Alicja Mortensen
- grid.418079.30000 0000 9531 3915National Research Centre for the Working Environment, Copenhagen, Denmark
| | - Keld Alstrup Jensen
- grid.418079.30000 0000 9531 3915National Research Centre for the Working Environment, Copenhagen, Denmark
| | - Martin Roursgaard
- grid.5254.60000 0001 0674 042XSection of Environmental Health, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Anne Thoustrup Saber
- grid.418079.30000 0000 9531 3915National Research Centre for the Working Environment, Copenhagen, Denmark
| | - Peter Møller
- grid.5254.60000 0001 0674 042XSection of Environmental Health, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - George Biskos
- grid.426429.f0000 0004 0580 3152Atmosphere and Climate Research Centre, The Cyprus Institute, Nicosia, Cyprus ,grid.5292.c0000 0001 2097 4740Faculty of Civil Engineering and Geosciences, Delft University of Technology, Delft, The Netherlands
| | - Ulla Vogel
- National Research Centre for the Working Environment, Copenhagen, Denmark.
| |
Collapse
|
18
|
Sivaraman K, Wrenger S, Liu B, Schaudien D, Hesse C, Gomez-Mariano G, Perez-Luz S, Sewald K, DeLuca D, Wurm MJ, Pino P, Welte T, Martinez-Delgado B, Janciauskiene S. Mice inflammatory responses to inhaled aerosolized LPS: effects of various forms of human alpha1-antitrypsin. J Leukoc Biol 2023; 113:58-70. [PMID: 36822165 DOI: 10.1093/jleuko/qiac004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Indexed: 01/12/2023] Open
Abstract
Rodent models of lipopolysaccharide (LPS)-induced pulmonary inflammation are used for anti-inflammatory drug testing. We aimed to characterize mice responses to aerosolized LPS alone or with intraperitoneal (i.p.) delivery of alpha1-antitrypsin (AAT). Balb/c mice were exposed to clean air or aerosolized LPS (0.21 mg/mL) for 10 min per day, for 3 d. One hour after each challenge, animals were treated i.p. with saline or with (4 mg/kg body weight) one of the AAT preparations: native (AAT), oxidized (oxAAT), recombinant (recAAT), or peptide of AAT (C-36). Experiments were terminated 6 h after the last dose of AATs. Transcriptome data of mice lungs exposed to clean air versus LPS revealed 656 differentially expressed genes and 155 significant gene ontology terms, including neutrophil migration and toll-like receptor signaling pathways. Concordantly, mice inhaling LPS showed higher bronchoalveolar lavage fluid neutrophil counts and levels of myeloperoxidase, inducible nitric oxide synthase, IL-1β, TNFα, KC, IL-6, and granulocyte-macrophage colony-stimulating factor (GM-CSF). Plasma inflammatory markers did not increase. After i.p. application of AATs, about 1% to 2% of proteins reached the lungs but, except for GM-CSF, none of the proteins significantly influenced inflammatory markers. All AATs and C-36 significantly inhibited LPS-induced GM-CSF release. Surprisingly, only oxAAT decreased the expression of several LPS-induced inflammatory genes, such as Cxcl3, Cd14, Il1b, Nfkb1, and Nfkb2, in lung tissues. According to lung transcriptome data, oxAAT mostly affected genes related to transcriptional regulation while native AAT or recAAT affected genes of inflammatory pathways. Hence, we present a feasible mice model of local lung inflammation induced via aerosolized LPS that can be useful for systemic drug testing.
Collapse
Affiliation(s)
- Kokilavani Sivaraman
- Department of Pulmonary and Infectious Diseases and BREATH German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
| | - Sabine Wrenger
- Department of Pulmonary and Infectious Diseases and BREATH German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
| | - Bin Liu
- Department of Pulmonary and Infectious Diseases and BREATH German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
| | - Dirk Schaudien
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Christina Hesse
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany.,Fraunhofer Cluster of Excellence Immune-Mediated Diseases CIMD, Hannover, Germany
| | - Gema Gomez-Mariano
- Molecular Genetics, Institute of Rare Diseases Research, Institute of Health Carlos III (ISCIII), Centro de Investigacion Biomedica en red de Enfermedades Raras, U758 (CIBERER), Majadahonda, Spain
| | - Sara Perez-Luz
- Molecular Genetics, Institute of Rare Diseases Research, Institute of Health Carlos III (ISCIII), Centro de Investigacion Biomedica en red de Enfermedades Raras, U758 (CIBERER), Majadahonda, Spain
| | - Katherina Sewald
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - David DeLuca
- Department of Pulmonary and Infectious Diseases and BREATH German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
| | | | | | - Tobias Welte
- Department of Pulmonary and Infectious Diseases and BREATH German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
| | - Beatriz Martinez-Delgado
- Molecular Genetics, Institute of Rare Diseases Research, Institute of Health Carlos III (ISCIII), Centro de Investigacion Biomedica en red de Enfermedades Raras, U758 (CIBERER), Majadahonda, Spain
| | - Sabina Janciauskiene
- Department of Pulmonary and Infectious Diseases and BREATH German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany.,Department of Genetics and Clinical Immunology, National Institute of Tuberculosis and Lung Diseases, Warsaw, Poland
| |
Collapse
|
19
|
Association between Serum Amyloid A Level and White Matter Hyperintensity Burden: a Cross-Sectional Analysis in Patients with Acute Ischemic Stroke. Neurol Ther 2022; 12:161-175. [PMID: 36374429 PMCID: PMC9837367 DOI: 10.1007/s40120-022-00415-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 10/17/2022] [Indexed: 11/16/2022] Open
Abstract
INTRODUCTION This work aimed to determine the potential link between white matter hyperintensity (WMH) burden and serum amyloid A (SAA) level in patients with acute ischemic stroke. METHODS Consecutive patients with acute large artery atherosclerosis (LAA) stroke between April 2021 and May 2022 were included. WMH volumes (periventricular, deep, and total) were measured using the Fazekas score and a semiautomated volumetric analysis on fluid-attenuated inversion recovery-magnetic resonance imaging. The burdens of WMH were scored to assess the dose-dependent association between SAA and WMH volume. Multivariate regression and a two-piecewise linear regression model were used to evaluate whether SAA levels are an independent predictor of WMH, and to discover the threshold effect or saturation effect of SAA levels with respect to WMH volume. RESULTS The mean age of patients was 63.2 ± 11.5 years, with 65.9% men. The median SAA level was 3.93 mg/L and the total WMH volume of 6.86 cm3. In the multivariable analysis, SAA remained an independent predictor of total WMH volume [β = 0.82, 95% confidence interval (CI) = 0.49-1.07, p < 0.001], periventricular WMH volume (adjusted β = 0.76, 95% CI = 0.46-1.07, p < 0.001), and deep WMH volume (adjusted β = 0.26, 95% CI = 0.06-0.45, p = 0.011) after controlling for confounders. Furthermore, SAA levels were associated with periventricular Fazekas score, deep Fazekas score, and Fazekas grades. Threshold effect and saturation effect analyses demonstrated a nonlinear relationship between SAA levels and periventricular white matter hyperintensity (PVWMH) volumes, with SAA levels (2.12-19.89 mg/L) having significant dose-dependent relationships with periventricular WMH volumes (adjusted β = 1.98, 95% CI = 1.12-2.84, p < 0.001). CONCLUSION SAA level ranging from 2.12 to 19.89 mg/L is dose-dependently associated with periventricular WMH development. These findings point the way forward for future research into the pathophysiology of WMH.
Collapse
|
20
|
Diaz Villamil E, De Roeck L, Vanorlé M, Communi D. UTP Regulates the Cardioprotective Action of Transplanted Stem Cells Derived From Mouse Cardiac Adipose Tissue. Front Pharmacol 2022; 13:906173. [PMID: 35784739 PMCID: PMC9240194 DOI: 10.3389/fphar.2022.906173] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/20/2022] [Indexed: 11/26/2022] Open
Abstract
Adipose tissue is a source of stem cells with a high potential of differentiation for cell-based regenerative therapies. We previously identified mouse P2Y2, an ATP and UTP nucleotide receptor, as a regulator of adipogenic and endothelial differentiation of cardiac adipose-derived stem cells (cADSC). We investigated here the potential involvement of P2Y2 receptor in the cardioprotective action of undifferentiated cADSC transplantation in mouse ischemic heart. Transplantation of cADSC was realized in the periphery of the infarcted zone of ischemic heart, 3 days after left anterior descending artery ligation. A strong reduction of collagen stained area was observed 14 days after cADSC injection, compared to PBS injection. Interestingly, loss of P2Y2 expression totally inhibits the ability of transplanted cADSC to reduce cardiac fibrosis. A detailed gene ontology enrichment analysis was realized by comparing RNA-sequencing data obtained for UTP-treated wild type cASDC and UTP-treated P2Y2-null cASDC. We identified UTP target genes linked to extracellular matrix organization such as matrix metalloproteinases and various collagen types, UTP target genes related to macrophage chemotaxis and differentiation into pro-fibrotic foam cells, and a significant number of UTP target genes linked to angiogenesis regulation. More particularly, we showed that UTP regulated the secretion of CCL5, CXCL5, and CCL12 chemokines and serum amyloid apolipoprotein 3, in the supernatants of UTP-treated cADSC. Interestingly, CCL5 is reported as a key factor in post-infarction heart failure and in the reparative and angiogenic action of transplanted ADSC on ischemic tissue. We investigated then if a UTP-pretreatment of cADSC amplifies their effect on cardiac revascularization in mouse ischemic heart. Transplantation of cADSC was able to increase peri-infarct capillary density, 14 days after their injection. This beneficial effect on cardiac revascularization was enhanced by a UTP-pretreatment of cADSC before their transplantation, and not observed using P2Y2-null cADSC. Our data support that the efficacy of transplanted cADSC can be regulated by the release of inflammatory mediators such as extracellular nucleotides in the ischemic site. The present study highlights the P2Y2 receptor as a regulator of cADSC cardioprotective action, and as a potential target for the therapeutic use of undifferentiated cADSC in post-ischemic cardiac ischemia.
Collapse
Affiliation(s)
| | | | | | - Didier Communi
- *Correspondence: Didier Communi, , orcid.org/0000-0003-1050-1493
| |
Collapse
|
21
|
Shridas P, Ji A, Trumbauer AC, Noffsinger VP, Leung SW, Dugan AJ, Thatcher SE, Cassis LA, de Beer FC, Webb NR, Tannock LR. Adipocyte-Derived Serum Amyloid A Promotes Angiotensin II-Induced Abdominal Aortic Aneurysms in Obese C57BL/6J Mice. Arterioscler Thromb Vasc Biol 2022; 42:632-643. [PMID: 35344382 PMCID: PMC9050948 DOI: 10.1161/atvbaha.121.317225] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 03/09/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND Obesity increases the risk for human abdominal aortic aneurysms (AAAs) and enhances Ang II (angiotensin II)-induced AAA formation in C57BL/6J mice. Obesity is also associated with increases in perivascular fat that expresses proinflammatory markers including SAA (serum amyloid A). We previously reported that deficiency of SAA significantly reduces Ang II-induced inflammation and AAA in hyperlipidemic apoE-deficient mice. In this study. we investigated whether adipose tissue-derived SAA plays a role in Ang II-induced AAA in obese C57BL/6J mice. METHODS The development of AAA was compared between male C57BL/6J mice (wild type), C57BL/6J mice lacking SAA1.1, SAA2.1, and SAA3 (TKO); and TKO mice harboring a doxycycline-inducible, adipocyte-specific SAA1.1 transgene (TKO-Tgfat; SAA expressed only in fat). All mice were fed an obesogenic diet and doxycycline to induce SAA transgene expression and infused with Ang II to induce AAA. RESULTS In response to Ang II infusion, SAA expression was significantly increased in perivascular fat of obese C57BL/6J mice. Maximal luminal diameters of the abdominal aorta were determined by ultrasound before and after Ang II infusion, which indicated a significant increase in aortic luminal diameters in wild type and TKO-TGfat mice but not in TKO mice. Adipocyte-specific SAA expression was associated with MMP (matrix metalloproteinase) activity and macrophage infiltration in abdominal aortas of Ang II-infused obese mice. CONCLUSIONS We demonstrate for the first time that SAA deficiency protects obese C57BL/6J mice from Ang II-induced AAA. SAA expression only in adipocytes is sufficient to cause AAA in obese mice infused with Ang II.
Collapse
Affiliation(s)
- Preetha Shridas
- Departments of Internal Medicine (P.S., A.J., V.P.N., S.W.L., F.C.d.B., L.R.T.), University of Kentucky, Lexington
- Saha Cardiovascular Research Center (P.S., A.C.T., S.W.L., F.C.d.B., N.R.W., L.R.T.), University of Kentucky, Lexington
- Barnstable Brown Diabetes Center (P.S., F.C.d.B., N.R.W., L.R.T.), University of Kentucky, Lexington
| | - Ailing Ji
- Departments of Internal Medicine (P.S., A.J., V.P.N., S.W.L., F.C.d.B., L.R.T.), University of Kentucky, Lexington
| | - Andrea C Trumbauer
- Saha Cardiovascular Research Center (P.S., A.C.T., S.W.L., F.C.d.B., N.R.W., L.R.T.), University of Kentucky, Lexington
| | - Victoria P Noffsinger
- Departments of Internal Medicine (P.S., A.J., V.P.N., S.W.L., F.C.d.B., L.R.T.), University of Kentucky, Lexington
| | - Steve W Leung
- Departments of Internal Medicine (P.S., A.J., V.P.N., S.W.L., F.C.d.B., L.R.T.), University of Kentucky, Lexington
- Saha Cardiovascular Research Center (P.S., A.C.T., S.W.L., F.C.d.B., N.R.W., L.R.T.), University of Kentucky, Lexington
| | - Adam J Dugan
- Biostatistics (A.J.D.), University of Kentucky, Lexington
| | - Sean E Thatcher
- Department of Pharmacology, Temple University, Philadelphia, PA (S.E.T.)
| | - Lisa A Cassis
- Pharmacology and Nutritional Sciences (L.A.C., N.R.W.), University of Kentucky, Lexington
| | - Frederick C de Beer
- Departments of Internal Medicine (P.S., A.J., V.P.N., S.W.L., F.C.d.B., L.R.T.), University of Kentucky, Lexington
- Saha Cardiovascular Research Center (P.S., A.C.T., S.W.L., F.C.d.B., N.R.W., L.R.T.), University of Kentucky, Lexington
- Barnstable Brown Diabetes Center (P.S., F.C.d.B., N.R.W., L.R.T.), University of Kentucky, Lexington
| | - Nancy R Webb
- Pharmacology and Nutritional Sciences (L.A.C., N.R.W.), University of Kentucky, Lexington
- Saha Cardiovascular Research Center (P.S., A.C.T., S.W.L., F.C.d.B., N.R.W., L.R.T.), University of Kentucky, Lexington
- Barnstable Brown Diabetes Center (P.S., F.C.d.B., N.R.W., L.R.T.), University of Kentucky, Lexington
| | - Lisa R Tannock
- Departments of Internal Medicine (P.S., A.J., V.P.N., S.W.L., F.C.d.B., L.R.T.), University of Kentucky, Lexington
- Saha Cardiovascular Research Center (P.S., A.C.T., S.W.L., F.C.d.B., N.R.W., L.R.T.), University of Kentucky, Lexington
- Barnstable Brown Diabetes Center (P.S., F.C.d.B., N.R.W., L.R.T.), University of Kentucky, Lexington
- Department of Veterans Affairs, Lexington, KY (L.R.T.)
| |
Collapse
|
22
|
Serum Amyloid A is not obligatory for high-fat, high-sucrose, cholesterol-fed diet-induced obesity and its metabolic and inflammatory complications. PLoS One 2022; 17:e0266688. [PMID: 35436297 PMCID: PMC9015120 DOI: 10.1371/journal.pone.0266688] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 03/24/2022] [Indexed: 12/26/2022] Open
Abstract
Several studies in the past have reported positive correlations between circulating Serum amyloid A (SAA) levels and obesity. However, based on limited number of studies involving appropriate mouse models, the role of SAA in the development of obesity and obesity-related metabolic consequences has not been established. Accordingly, herein, we have examined the role of SAA in the development of obesity and its associated metabolic complications in vivo using mice deficient for all three inducible forms of SAA: SAA1.1, SAA2.1 and SAA3 (TKO). Male and female mice were rendered obese by feeding a high fat, high sucrose diet with added cholesterol (HFHSC) and control mice were fed rodent chow diet. Here, we show that the deletion of SAA does not affect diet-induced obesity, hepatic lipid metabolism or adipose tissue inflammation. However, there was a modest effect on glucose metabolism. The results of this study confirm previous findings that SAA levels are elevated in adipose tissues as well as in the circulation in diet-induced obese mice. However, the three acute phase SAAs do not play a causative role in the development of obesity or obesity-associated adipose tissue inflammation and dyslipidemia.
Collapse
|
23
|
Vascular Permeability in Diseases. Int J Mol Sci 2022; 23:ijms23073645. [PMID: 35409010 PMCID: PMC8998843 DOI: 10.3390/ijms23073645] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/20/2022] [Accepted: 03/24/2022] [Indexed: 02/06/2023] Open
Abstract
Vascular permeability is a selective mechanism that maintains the exchange between vessels, tissues, and organs. The regulation was mostly studied during the nineteenth century by physiologists who defined physical laws and equations, taking blood, tissue interstitial, and oncotic pressure into account. During the last decades, a better knowledge of vascular cell functions and blood-vessel interactions opens a new area of vascular biology. Endothelial cell receptors vascular cell adhesion molecule (VCAM), intercellular cell adhesion molecule (ICAM), vascular endothelial growth factor receptor (VEGFR-2), receptor for advanced glycation end products (RAGE), and mediators were identified and their role in homeostasis and pathological situations was described. The molecular differences of endothelial cell junctions (tight, gap, and adherens junctions) and their role in vascular permeability were characterized in different organs. The main mediators of vasomotricity and permeability, such as prostaglandins, nitric oxide (NO), prostacyclin, vascular growth factor (VEGF), and cytokines, have been demonstrated to possess major functions in steady state and pathological situations. Leukocytes were shown to adhere to endothelium and migrate during inflammatory situations and infectious diseases. Increased vascular permeability is linked to endothelium integrity. Glycocalyx, when intact, may limit cancer cell metastasis. Biological modifications of blood and tissue constituents occurring in diabetes mellitus were responsible for increased permeability and, consequently, ocular and renal complications. Vascular pressure and fluidity are major determinants of pulmonary and cerebral edema. Beside the treatment of the infectious disease, of the blood circulation dysfunction and inflammatory condition, drugs (cyclooxygenase inhibitors) and specific antibodies anti-cytokine (anti-VEGF) have been demonstrated to reduce the severity and the mortality in diseases that exhibited enhanced vascular permeability.
Collapse
|
24
|
Ren Y, Zhao H, Yin C, Lan X, Wu L, Du X, Griffiths HR, Gao D. Adipokines, Hepatokines and Myokines: Focus on Their Role and Molecular Mechanisms in Adipose Tissue Inflammation. Front Endocrinol (Lausanne) 2022; 13:873699. [PMID: 35909571 PMCID: PMC9329830 DOI: 10.3389/fendo.2022.873699] [Citation(s) in RCA: 101] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 06/17/2022] [Indexed: 11/18/2022] Open
Abstract
Chronic low-grade inflammation in adipose tissue (AT) is a hallmark of obesity and contributes to various metabolic disorders, such as type 2 diabetes and cardiovascular diseases. Inflammation in ATs is characterized by macrophage infiltration and the activation of inflammatory pathways mediated by NF-κB, JNK, and NLRP3 inflammasomes. Adipokines, hepatokines and myokines - proteins secreted from AT, the liver and skeletal muscle play regulatory roles in AT inflammation via endocrine, paracrine, and autocrine pathways. For example, obesity is associated with elevated levels of pro-inflammatory adipokines (e.g., leptin, resistin, chemerin, progranulin, RBP4, WISP1, FABP4, PAI-1, Follistatin-like1, MCP-1, SPARC, SPARCL1, and SAA) and reduced levels of anti-inflammatory adipokines such as adiponectin, omentin, ZAG, SFRP5, CTRP3, vaspin, and IL-10. Moreover, some hepatokines (Fetuin A, DPP4, FGF21, GDF15, and MANF) and myokines (irisin, IL-6, and DEL-1) also play pro- or anti-inflammatory roles in AT inflammation. This review aims to provide an updated understanding of these organokines and their role in AT inflammation and related metabolic abnormalities. It serves to highlight the molecular mechanisms underlying the effects of these organokines and their clinical significance. Insights into the roles and mechanisms of these organokines could provide novel and potential therapeutic targets for obesity-induced inflammation.
Collapse
Affiliation(s)
- Yakun Ren
- Institute of Molecular and Translational Medicine, Xian Jiaotong University Health Science Center, Xi’an, China
| | - Hao Zhao
- School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Chunyan Yin
- Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Xi Lan
- Institute of Molecular and Translational Medicine, Xian Jiaotong University Health Science Center, Xi’an, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Litao Wu
- Institute of Molecular and Translational Medicine, Xian Jiaotong University Health Science Center, Xi’an, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Xiaojuan Du
- Institute of Molecular and Translational Medicine, Xian Jiaotong University Health Science Center, Xi’an, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Helen R. Griffiths
- Swansea University Medical School, Swansea University, Swansea, United Kingdom
| | - Dan Gao
- Institute of Molecular and Translational Medicine, Xian Jiaotong University Health Science Center, Xi’an, China
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Center, Xi’an, China
- *Correspondence: Dan Gao,
| |
Collapse
|
25
|
Shridas P, Patrick AC, Tannock LR. Role of Serum Amyloid A in Abdominal Aortic Aneurysm and Related Cardiovascular Diseases. Biomolecules 2021; 11:biom11121883. [PMID: 34944527 PMCID: PMC8699432 DOI: 10.3390/biom11121883] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/07/2021] [Accepted: 12/13/2021] [Indexed: 01/02/2023] Open
Abstract
Epidemiological data positively correlate plasma serum amyloid A (SAA) levels with cardiovascular disease severity and mortality. Studies by several investigators have indicated a causal role for SAA in the development of atherosclerosis in animal models. Suppression of SAA attenuates the development of angiotensin II (AngII)-induced abdominal aortic aneurysm (AAA) formation in mice. Thus, SAA is not just a marker for cardiovascular disease (CVD) development, but it is a key player. However, to consider SAA as a therapeutic target for these diseases, the pathway leading to its involvement needs to be understood. This review provides a brief description of the pathobiological significance of this enigmatic molecule. The purpose of this review is to summarize the data relevant to its role in the development of CVD, the pitfalls in SAA research, and unanswered questions in the field.
Collapse
Affiliation(s)
- Preetha Shridas
- Department of Internal Medicine, University of Kentucky, Lexington, KY 40536, USA
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY 40536, USA
- Barnstable Brown Diabetes Center, University of Kentucky, Lexington, KY 40536, USA
| | - Avery C Patrick
- Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Lisa R Tannock
- Department of Internal Medicine, University of Kentucky, Lexington, KY 40536, USA
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY 40536, USA
- Barnstable Brown Diabetes Center, University of Kentucky, Lexington, KY 40536, USA
- Veterans Affairs Lexington, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
26
|
Han B, Li C, Li H, Li Y, Luo X, Liu Y, Zhang J, Zhang Z, Yu X, Zhai Z, Xu X, Xiao F. Discovery of plasma biomarkers with data-independent acquisition mass spectrometry and antibody microarray for diagnosis and risk stratification of pulmonary embolism. J Thromb Haemost 2021; 19:1738-1751. [PMID: 33825327 DOI: 10.1111/jth.15324] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 02/22/2021] [Accepted: 03/25/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Pulmonary embolism (PE) is a leading cause of cardiovascular mortality worldwide. Rapid and accurate diagnosis and risk stratification are crucial for timely treatment options, especially in high-risk PE. OBJECTIVES The study aims to profile the comprehensive changes of plasma proteomes in PE patients and identify the potential biomarkers for both diagnosis and risk stratification. PATIENTS/METHODS Based on the data-independent acquisition mass spectrometry and antibody array proteomic technology, we screened the plasma samples (13 and 32 proteomes, respectively) in two independent studies consisting of high-risk PE patients, non-high-risk PE patients, and healthy controls. Some significantly differentially expressed proteins were quantified by ELISA in a new study group with 50 PE patients and 26 healthy controls. RESULTS We identified 207 and 70 differentially expressed proteins in PE and high-risk PE. These proteins were involved in multiple thrombosis-associated biological processes including blood coagulation, inflammation, injury, repair, and chemokine-mediated cellular response. It was verified that five proteins including SAA1, S100A8, TNC, GSN, and HRG had significant change in PE and/or in high-risk PE. The receiver operating characteristic curve analysis based on binary logistic regression showed that the area under the curve (AUC) of SAA1, S100A8, and TNC in PE diagnosis were 0.882, 0.788, and 0.795, and AUC of S100A8 and TNC in high-risk PE diagnosis were 0.773 and 0.720. CONCLUSION As predictors of inflammation or injury repair, SAA1, S100A8, and TNC are potential plasma biomarkers for the diagnosis and risk stratification of PE.
Collapse
Affiliation(s)
- Bingqing Han
- Peking University Fifth School of Clinical Medicine, Beijing, China
- The Key Laboratory of Geriatrics, Beijing Institution of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Chuanbao Li
- Department of Laboratory Medicine, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Hexin Li
- Clinical Biobank, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Ying Li
- The Key Laboratory of Geriatrics, Beijing Institution of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Xuanmei Luo
- The Key Laboratory of Geriatrics, Beijing Institution of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Ye Liu
- The Key Laboratory of Geriatrics, Beijing Institution of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Junhua Zhang
- The Key Laboratory of Geriatrics, Beijing Institution of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhu Zhang
- Department of Respiratory and Clinical Care Medicine, China-Japan, Friendship Hospital, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Xiaobo Yu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing, China
| | - Zhenguo Zhai
- Department of Respiratory and Clinical Care Medicine, China-Japan, Friendship Hospital, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Xiaomao Xu
- Department of Respiratory and Critical Care Medicine, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Fei Xiao
- Peking University Fifth School of Clinical Medicine, Beijing, China
- The Key Laboratory of Geriatrics, Beijing Institution of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Clinical Biobank, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
27
|
Zhang Y, Zhou J, Liu J, Li S, Zhou S, Zhang C, Wang Y, Shi J, Liu J, Wu Q. RNA-Seq analysis of the protection by Dendrobium nobile alkaloids against carbon tetrachloride hepatotoxicity in mice. Biomed Pharmacother 2021; 137:111307. [PMID: 33561648 DOI: 10.1016/j.biopha.2021.111307] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 11/30/2020] [Accepted: 12/26/2020] [Indexed: 01/04/2023] Open
Abstract
OBJECTIVE Dendrobium nobile is a genuine Chinese medicine. Dendrobium nobile Lindl. alkaloids (DNLA) protects against CCl4-induced acute liver injury. This study used RNA-Seq to explore the mechanisms. METHODS Mice were pretreated with DNLA (10 and 20 mg/kg, po) for 7 days, and subsequently intoxicated with CCl4 (20 μL/kg, ip for 24 h). Liver RNA was extracted and subjected to RNA-Seq. The bioinformatics, including PCA, GO, KEGG, two-dimensional clustering, Ingenuity Pathways Analysis (IPA), and Illumina BaseSpace Correlation Engine (BSCE) were used to analyze the data. qPCR was performed on selected genes to verify RNA-Seq results. RESULTS DNLA protection against CCl4 hepatotoxicity was confirmed by histopathology. PCA revealed the distinct gene expression patterns between the different treatment groups. GO showed that CCl4 induced the activation, adhesion and proliferation of immune cells. KEGG showed CCl4 induced oxidative stress, diseases and compromised adaptive responses. CCl4 induced differentially expressed genes (DEGs) were identified by DESeq2 with Padj < 0.05 and 2D-clustered with other groups. DNLA reverted CCl4-induced DEGs in a dose-dependent manner. qPCR analysis of S100 g, Sprr1, CCL3/7, Saa2/3, IL1rn, Cox7a2 and Rad15 confirmed RNA-Seq results. IPA showed that CCl4 treatment altered some signaling and metabolic pathways, which were ameliorated or returned to normal following DNLA treatment. The CCl4-activated mitochondrial oxidative phosphorylation was illustrated as an example. IPA Upstream Regulator Analysis further revealed the activated or inhibited molecules and chemicals that are responsible for CCl4-induced DEGs, and DNLA attenuated these changes. BSCE analysis verified that CCl4-induced DEGs were highly correlated with the GEO database of CCl4 hepatotoxicity in rodents, and DNLA dose-dependently attenuated such correlation. CONCLUSION RNA-Seq revealed CCl4-induced DEGs, disruption of canonical pathways, activation or inhibition of upstream regulators, which are highly correlated with database for CCl4 hepatotoxicity. All these changes were attenuated or returned to normal by DNLA, demonstrating the mechanisms for DNLA to protect against CCl4 hepatotoxicity.
Collapse
Affiliation(s)
- Ya Zhang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnocentric of Ministry of Education, Zunyi Medical University, Zunyi, China.
| | - Jinxin Zhou
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnocentric of Ministry of Education, Zunyi Medical University, Zunyi, China.
| | - Jiajia Liu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnocentric of Ministry of Education, Zunyi Medical University, Zunyi, China.
| | - Shujun Li
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnocentric of Ministry of Education, Zunyi Medical University, Zunyi, China.
| | - Shaoyu Zhou
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnocentric of Ministry of Education, Zunyi Medical University, Zunyi, China.
| | - Chengchen Zhang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnocentric of Ministry of Education, Zunyi Medical University, Zunyi, China.
| | - Yan Wang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnocentric of Ministry of Education, Zunyi Medical University, Zunyi, China.
| | - Jingshan Shi
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnocentric of Ministry of Education, Zunyi Medical University, Zunyi, China.
| | - Jie Liu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnocentric of Ministry of Education, Zunyi Medical University, Zunyi, China.
| | - Qin Wu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnocentric of Ministry of Education, Zunyi Medical University, Zunyi, China.
| |
Collapse
|
28
|
Shen S, He F, Cheng C, Xu B, Sheng J. Uric acid aggravates myocardial ischemia-reperfusion injury via ROS/NLRP3 pyroptosis pathway. Biomed Pharmacother 2021; 133:110990. [PMID: 33232925 DOI: 10.1016/j.biopha.2020.110990] [Citation(s) in RCA: 111] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 11/04/2020] [Accepted: 11/05/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The NOD-like receptor pyrin domain-containing protein 3 (NLRP3) inflammasome activation-mediated pyroptosis pathway has been linked to myocardial ischemia-reperfusion (MI/R) injury. This study explored whether uric acid (UA) aggravates MI/R injury through NLRP3 inflammasome-mediated pyroptosis. METHODS In vivo, a mouse MI/R model was established by ligating the left coronary artery, and a mouse hyperuricemia model was created by intraperitoneal injection of potassium oxonate (PO). Then, the myocardial infarction (MI) size; terminal deoxynucleotidyl transferase dUTP nick end-labeling (TUNEL) immunofluorescence; and serum levels of lactate dehydrogenase (LDH), creatine kinase isoenzyme (CK-MB), and UA, as well as the expression level of pyroptosis-related protein and caspase-3 in heart tissues, were measured. Separately, primary mouse cardiomyocytes were cultured in vitro to create a hypoxia/reoxygenation (H/R) model. We then compared cardiomyocytes viability, TUNEL immunofluorescence, and the levels of LDH, reactive oxygen species (ROS), and pyroptosis-related protein and caspase-3 in cardiomyocytes. RESULTS In vivo, the MI area, levels of CK-MB and LDH, rate of cell death, and pyroptosis-related protein and the expression of caspase-3 were significantly higher in the MI/R group than in the sham group, and high UA levels worsened these changes. In vitro, cardiomyocytes viability was significantly downregulated, and the levels of ROS, LDH, pyroptosis-related protein, caspase-3, and the rate of cardiomyocyte death were significantly higher in the H/R + UA group compared with the HR group. Administration of an NLRP3 inflammasome inhibitor and ROS scavenger reversed these effects. CONCLUSION UA aggravates MI/R-induced activation of the NLRP3 inflammatory cascade and pyroptosis by promoting ROS generation, while inflammasome inhibitors and ROS scavengers partly reverse the injury.
Collapse
Affiliation(s)
- ShiChun Shen
- Department of Cardiology, The Second Affiliated Hospital of Anhui Medical University, No. 678 FuRong Road, Hefei, Anhui Province, 230601, China.
| | - Fei He
- Department of Cardiology, The Second Affiliated Hospital of Anhui Medical University, No. 678 FuRong Road, Hefei, Anhui Province, 230601, China.
| | - Cheng Cheng
- Department of Cardiology, The Second Affiliated Hospital of Anhui Medical University, No. 678 FuRong Road, Hefei, Anhui Province, 230601, China.
| | - BangLong Xu
- Department of Cardiology, The Second Affiliated Hospital of Anhui Medical University, No. 678 FuRong Road, Hefei, Anhui Province, 230601, China.
| | - JianLong Sheng
- Department of Cardiology, The Second Affiliated Hospital of Anhui Medical University, No. 678 FuRong Road, Hefei, Anhui Province, 230601, China.
| |
Collapse
|
29
|
Han CY, Kang I, Omer M, Wang S, Wietecha T, Wight TN, Chait A. Serum amyloid A-containing HDL binds adipocyte-derived versican and macrophage-derived biglycan, reducing its antiinflammatory properties. JCI Insight 2020; 5:142635. [PMID: 32970631 PMCID: PMC7605543 DOI: 10.1172/jci.insight.142635] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 09/16/2020] [Indexed: 12/25/2022] Open
Abstract
The ability of HDL to inhibit inflammation in adipocytes and adipose tissue is reduced when HDL contains serum amyloid A (SAA) that is trapped by proteoglycans at the adipocyte surface. Because we recently found that the major extracellular matrix proteoglycan produced by hypertrophic adipocytes is versican, whereas activated adipose tissue macrophages produce mainly biglycan, we further investigated the role of proteoglycans in determining the antiinflammatory properties of HDL. The distributions of versican, biglycan, apolipoprotein A1 (the major apolipoprotein of HDL), and SAA were similar in adipose tissue from obese mice and obese human subjects. Colocalization of SAA-enriched HDL with versican and biglycan at the cell surface of adipocyte and peritoneal macrophages, respectively, was blocked by silencing these proteoglycans, which also restored the antiinflammatory property of SAA-enriched HDL despite the presence of SAA. Similar to adipocytes, normal HDL exerted its antiinflammatory function in macrophages by reducing lipid rafts, reactive oxygen species generation, and translocation of Toll-like receptor 4 and NADPH oxidase 2 into lipid rafts, effects that were not observed with SAA-enriched HDL. These findings imply that SAA present in HDL can be trapped by adipocyte-derived versican and macrophage-derived biglycan, thereby blunting HDL’s antiinflammatory properties. Versican in adiopcytes and biglycan in macrophages trap serum amyloid A-containing HDL, thereby blocking HDL’s anti-inflammatory properties.
Collapse
Affiliation(s)
- Chang Yeop Han
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Inkyung Kang
- Matrix Biology Program, Benaroya Research Institute, Seattle, Washington, USA
| | - Mohamed Omer
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Shari Wang
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Tomasz Wietecha
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Thomas N Wight
- Matrix Biology Program, Benaroya Research Institute, Seattle, Washington, USA
| | - Alan Chait
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
30
|
Structural Basis for Vital Function and Malfunction of Serum Amyloid A: an Acute-Phase Protein that Wears Hydrophobicity on Its Sleeve. Curr Atheroscler Rep 2020; 22:69. [PMID: 32968930 PMCID: PMC7511256 DOI: 10.1007/s11883-020-00888-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2020] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW This review addresses normal and pathologic functions of serum amyloid A (SAA), an enigmatic biomarker of inflammation and protein precursor of AA amyloidosis, a life-threatening complication of chronic inflammation. SAA is a small, highly evolutionarily conserved acute-phase protein whose plasma levels increase up to one thousand-fold in inflammation, infection, or after trauma. The advantage of this dramatic but transient increase is unclear, and the complex role of SAA in immune response is intensely investigated. This review summarizes recent advances in our understanding of the structure-function relationship of this intrinsically disordered protein, outlines its newly emerging beneficial roles in lipid transport and inflammation control, and discusses factors that critically influence its misfolding in AA amyloidosis. RECENT FINDINGS High-resolution structures of lipid-free SAA in crystals and fibrils have been determined by x-ray crystallography and electron cryo-microscopy. Low-resolution structural studies of SAA-lipid complexes, together with biochemical, cell-based, animal model, genetic, and clinical studies, have provided surprising new insights into a wide range of SAA functions. An emerging vital role of SAA is lipid encapsulation to remove cell membrane debris from sites of injury. The structural basis for this role has been proposed. The lysosomal origin of AA amyloidosis has solidified, and its molecular and cellular mechanisms have emerged. Recent studies have revealed molecular underpinnings for understanding complex functions of this Cambrian protein in lipid transport, immune response, and amyloid formation. These findings help guide the search for much-needed targeted therapies to block the protein deposition in AA amyloidosis.
Collapse
|
31
|
Mao K, Geng W, Liao Y, Luo P, Zhong H, Ma P, Xu J, Zhang S, Tan Q, Jin Y. Identification of robust genetic signatures associated with lipopolysaccharide-induced acute lung injury onset and astaxanthin therapeutic effects by integrative analysis of RNA sequencing data and GEO datasets. Aging (Albany NY) 2020; 12:18716-18740. [PMID: 32969837 PMCID: PMC7585091 DOI: 10.18632/aging.104042] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 08/19/2020] [Indexed: 01/24/2023]
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are life-threatening clinical conditions predominantly arising from uncontrolled inflammatory reactions. It has been found that the administration of astaxanthin (AST) can exert protective effects against lipopolysaccharide (LPS)-induced ALI; however, the robust genetic signatures underlying LPS induction and AST treatment remain obscure. Here we performed a statistical meta-analysis of five publicly available gene expression datasets from LPS-induced ALI mouse models, conducted RNA-sequencing (RNA-seq) to screen differentially expressed genes (DEGs) in response to LPS administration and AST treatment, and integrative analysis to determine robust genetic signatures associated with LPS-induced ALI onset and AST administration. Both the meta-analyses and our experimental data identified a total of 198 DEGs in response to LPS administration, and 11 core DEGs (Timp1, Ly6i, Cxcl13, Irf7, Cxcl5, Ccl7, Isg15, Saa3, Saa1, Tgtp1, and Gbp11) were identified to be associated with AST therapeutic effects. Further, the 11 core DEGs were verified by quantitative real-time PCR (qRT-PCR) and immunohistochemistry (IHC), and functional enrichment analysis revealed that these genes are primarily associated with neutrophils and chemokines. Collectively, these findings unearthed the robust genetic signatures underlying LPS administration and the molecular targets of AST for ameliorating ALI/ARDS which provide directions for further research.
Collapse
Affiliation(s)
- Kaimin Mao
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Wei Geng
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Yuhan Liao
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Ping Luo
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Hua Zhong
- College of Life Sciences, Wuhan University, Hubei Province, Wuhan, 430072, China
| | - Pei Ma
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Juanjuan Xu
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Shuai Zhang
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Qi Tan
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Yang Jin
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| |
Collapse
|
32
|
Chait A, den Hartigh LJ, Wang S, Goodspeed L, Babenko I, Altemeier WA, Vaisar T. Presence of serum amyloid A3 in mouse plasma is dependent on the nature and extent of the inflammatory stimulus. Sci Rep 2020; 10:10397. [PMID: 32587356 PMCID: PMC7316782 DOI: 10.1038/s41598-020-66898-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 05/26/2020] [Indexed: 11/09/2022] Open
Abstract
Serum amyloid A3 (Saa3) derives mainly from extrahepatic tissue and is not detected in plasma from moderately inflamed obese mice. In contrast, it is present in plasma from mice acutely inflamed by injection of high dose of lipopolysaccharide (LPS). To reconcile these differences, we evaluated whether different acute inflammatory stimuli could affect the presence of Saa3 in plasma. Saa3 appeared dose dependently in plasma after LPS injection. In contrast, only very low levels were detected after sterile inflammation with silver nitrate despite levels of Saa1 and Saa2 being comparable to high dose LPS. Saa3 was not detected in plasma following casein administration. Although most Saa3 was found in HDL, a small amount was not lipoprotein associated. Gene expression and proteomic analysis of liver and adipose tissue suggested that a major source of Saa3 in plasma after injection of LPS was adipose tissue rather than liver. We conclude that Saa3 only appears in plasma after induction of acute inflammation by some but not all inflammatory stimuli. These findings are consistent with the observation that Saa3 is not detectable in plasma in more moderate chronic inflammatory states such as obesity.
Collapse
Affiliation(s)
- Alan Chait
- Divisions of Metabolism, Endocrinology and Nutrition, University of Washington, Seattle, WA, 98109, USA.
| | - Laura J den Hartigh
- Divisions of Metabolism, Endocrinology and Nutrition, University of Washington, Seattle, WA, 98109, USA
| | - Shari Wang
- Divisions of Metabolism, Endocrinology and Nutrition, University of Washington, Seattle, WA, 98109, USA
| | - Leela Goodspeed
- Divisions of Metabolism, Endocrinology and Nutrition, University of Washington, Seattle, WA, 98109, USA
| | - Ilona Babenko
- Divisions of Metabolism, Endocrinology and Nutrition, University of Washington, Seattle, WA, 98109, USA
| | - William A Altemeier
- Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle, WA, 98109, USA
| | - Tomas Vaisar
- Divisions of Metabolism, Endocrinology and Nutrition, University of Washington, Seattle, WA, 98109, USA
| |
Collapse
|
33
|
Zheng H, Li H, Zhang J, Fan H, Jia L, Ma W, Ma S, Wang S, You H, Yin Z, Li X. Serum amyloid A exhibits pH dependent antibacterial action and contributes to host defense against Staphylococcus aureus cutaneous infection. J Biol Chem 2019; 295:2570-2581. [PMID: 31819008 DOI: 10.1074/jbc.ra119.010626] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 12/04/2019] [Indexed: 12/21/2022] Open
Abstract
Serum amyloid A (SAA), one of the major highly conserved acute-phase proteins in most mammals, is predominantly produced by hepatocytes and also by a variety of cells in extrahepatic tissues. It is well-known that the expression of SAA is sharply increased in bacterial infections. However, the exact physiological function of SAA during bacterial infection remains unclear. Herein, we showed that SAA expression significantly increased in abscesses of Staphylococcus aureus cutaneous infected mice, which exert direct antibacterial effects by binding to the bacterial cell surface and disrupting the cell membrane in acidic conditions. Mechanically, SAA disrupts anionic liposomes by spontaneously forming small vesicles or micelles under acidic conditions. Especially, the N-terminal region of SAA is necessary for membrane disruption and bactericidal activity. Furthermore, we found that mice deficient in SAA1/2 were more susceptible to infection by S. aureus In addition, the expression of SAA in infected skin was regulated by interleukin-6. Taken together, these findings support a key role of the SAA in host defense and may provide a novel therapeutic strategy for cutaneous bacterial infection.
Collapse
Affiliation(s)
- Han Zheng
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Haifeng Li
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Jingyuan Zhang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Hanlu Fan
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Lina Jia
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Wenqiang Ma
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Shuoqian Ma
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Shenghong Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Hua You
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou 511436, China
| | - Zhinan Yin
- First Affiliated Hospital, Biomedical Translational Research Institute, Guangdong Province Key Laboratory of Molecular Immunology and Antibody Engineering, Jinan University, Guangzhou 510310, China
| | - Xiangdong Li
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China; Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou 511436, China.
| |
Collapse
|
34
|
Funcke JB, Scherer PE. Beyond adiponectin and leptin: adipose tissue-derived mediators of inter-organ communication. J Lipid Res 2019; 60:1648-1684. [PMID: 31209153 PMCID: PMC6795086 DOI: 10.1194/jlr.r094060] [Citation(s) in RCA: 203] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 06/17/2019] [Indexed: 01/10/2023] Open
Abstract
The breakthrough discoveries of leptin and adiponectin more than two decades ago led to a widespread recognition of adipose tissue as an endocrine organ. Many more adipose tissue-secreted signaling mediators (adipokines) have been identified since then, and much has been learned about how adipose tissue communicates with other organs of the body to maintain systemic homeostasis. Beyond proteins, additional factors, such as lipids, metabolites, noncoding RNAs, and extracellular vesicles (EVs), released by adipose tissue participate in this process. Here, we review the diverse signaling mediators and mechanisms adipose tissue utilizes to relay information to other organs. We discuss recently identified adipokines (proteins, lipids, and metabolites) and briefly outline the contributions of noncoding RNAs and EVs to the ever-increasing complexities of adipose tissue inter-organ communication. We conclude by reflecting on central aspects of adipokine biology, namely, the contribution of distinct adipose tissue depots and cell types to adipokine secretion, the phenomenon of adipokine resistance, and the capacity of adipose tissue to act both as a source and sink of signaling mediators.
Collapse
Affiliation(s)
- Jan-Bernd Funcke
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX
| | - Philipp E Scherer
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX
| |
Collapse
|
35
|
Abstract
Vitamin A is an essential nutrient for immune system development and function. After absorption from the diet, vitamin A is converted to retinol, which is delivered to cells and tissues by retinol-binding proteins. Serum amyloid A (SAA) proteins are retinol-binding proteins that transport retinol specifically during an infection. In this study, we illuminate the molecular details of how SAA proteins bind to retinol. We present the cocrystal structure of mouse SAA3 bound to retinol, which reveals that 3 molecules of SAA3 assemble to form a deep binding pocket that protects retinol, a fat-soluble molecule, from the aqueous environment. Our findings thus provide structural insight into how retinol is transported throughout the body during infection. Serum amyloid A (SAA) proteins are strongly induced in the liver by systemic infection and in the intestine by bacterial colonization. In infected mice, SAA proteins circulate in association with the vitamin A derivative retinol, suggesting that SAAs transport retinol during infection. Here we illuminate a structural basis for the retinol–SAA interaction. In the bloodstream of infected mice, most SAA is complexed with high-density lipoprotein (HDL). However, we found that the majority of the circulating retinol was associated with the small fraction of SAA proteins that circulate without binding to HDL, thus identifying free SAA as the predominant retinol-binding form in vivo. We then determined the crystal structure of retinol-bound mouse SAA3 at a resolution of 2.2 Å. Retinol-bound SAA3 formed a novel asymmetric trimeric assembly that was generated by the hydrophobic packing of the conserved amphipathic helices α1 and α3. This hydrophobic packing created a retinol-binding pocket in the center of the trimer, which was confirmed by mutagenesis studies. Together, these findings illuminate the molecular basis for retinol transport by SAA proteins during infection.
Collapse
|
36
|
Abstract
PURPOSE OF REVIEW Acute phase serum amyloid A (SAA) is persistently elevated in chronic inflammatory conditions, and elevated levels predict cardiovascular risk in humans. More recently, murine studies have demonstrated that over-expression of SAA increases and deficiency/suppression of SAA attenuates atherosclerosis. Thus, beyond being a biomarker, SAA appears to play a causal role in atherogenesis. The purpose of this review is to summarize the data supporting SAA as a key player in atherosclerosis development. RECENT FINDINGS A number of pro-inflammatory and pro-atherogenic activities have been ascribed to SAA. However, the literature is conflicted, as recombinant SAA, and/or lipid-free SAA, used in many of the earlier studies, do not reflect the activity of native human or murine SAA, which exists largely lipid-associated. Recent literatures demonstrate that SAA activates the NLRP3 inflammasome, alters vascular function, affects HDL function, and increases thrombosis. Importantly, SAA activity appears to be regulated by its lipid association, and HDL may serve to sequester and limit SAA activity. SUMMARY SAA has many pro-inflammatory and pro-atherogenic activities, is clearly demonstrated to affect atherosclerosis development, and may be a candidate target for clinical trials in cardiovascular diseases.
Collapse
Affiliation(s)
- Preetha Shridas
- Department of Internal Medicine
- Saha Cardiovascular Research Center
- Barnstable Brown Diabetes Center and University of Kentucky
| | - Lisa R Tannock
- Department of Internal Medicine
- Saha Cardiovascular Research Center
- Barnstable Brown Diabetes Center and University of Kentucky
- Veterans Affairs Lexington, Lexington, Kentucky, USA
| |
Collapse
|
37
|
McKay PF, Cizmeci D, Aldon Y, Maertzdorf J, Weiner J, Kaufmann SH, Lewis DJ, van den Berg RA, Del Giudice G, Shattock RJ. Identification of potential biomarkers of vaccine inflammation in mice. eLife 2019; 8:46149. [PMID: 31084714 PMCID: PMC6555592 DOI: 10.7554/elife.46149] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Accepted: 05/13/2019] [Indexed: 12/21/2022] Open
Abstract
Systems vaccinology approaches have been used successfully to define early signatures of the vaccine-induced immune response. However, the possibility that transcriptomics can also identify a correlate or surrogate for vaccine inflammation has not been fully explored. We have compared four licensed vaccines with known safety profiles, as well as three agonists of Toll-like receptors (TLRs) with known inflammatory potential, to elucidate the transcriptomic profile of an acceptable response to vaccination versus that of an inflammatory reaction. In mice, we looked at the transcriptomic changes in muscle at the injection site, the lymph node that drained the muscle, and the peripheral blood mononuclear cells (PBMCs)isolated from the circulating blood from 4 hr after injection and over the next week. A detailed examination and comparative analysis of these transcriptomes revealed a set of novel biomarkers that are reflective of inflammation after vaccination. These biomarkers are readily measurable in the peripheral blood, providing useful surrogates of inflammation, and provide a way to select candidates with acceptable safety profiles. Measles, whooping cough and other diseases can cause serious illness and death in humans, especially in young children and other vulnerable individuals. Giving people vaccines ‘trains’ their immune system to recognize and fight the microbes that cause the conditions. During an infection, the immune system triggers a set of responses that limit the spread of the infectious agent and eliminate it from the body. This can include swelling of tissues (known as inflammation), which in rare cases, can be life threatening. Inoculations work by sparking a mild immune response in the body. Before a new vaccine is licensed for use, it is thoroughly tested in mice and rodents, and then in human volunteers, to ensure it will cause little or no inflammation. Finding a way to predict early on whether a vaccine candidate will trigger dangerous levels of inflammation would improve this process. To explore this, McKay, Cizmeci et al. injected the muscle tissue of different groups of mice with one of four licensed vaccines which, by definition, cause little or no inflammation. Other groups of animals were given one of three drugs known to trigger inflammation. Over the following seven days the team repeatedly collected blood as well as cells from the muscle tissue and the lymph nodes. These samples were then analysed to find out which genes were switched on or off at any given time. The experiments show that the responses of genes in the blood and lymph cells of the mice are connected to those in the muscle cells. Therefore, blood samples may provide a quick and convenient way to assess how an animal is responding to a potential new vaccine. By comparing the genes switched on or off in response to the different vaccines and drugs, McKay, Cizemeci et al. were able to identify a set of genes (known as “biomarkers”) that are associated with inflammation in animals. These biomarkers can be used to spot early on whether a new treatment is triggering inflammation. The next step would then be to identify a similar or identical set of biomarkers in other animals used in vaccine research, and in humans. Ultimately, this approach could make the assessment of the safety of a new vaccine candidate easier.
Collapse
Affiliation(s)
- Paul F McKay
- Department of Medicine, Division of Infectious Diseases, Section of Virology, Imperial College London, London, United Kingdom
| | - Deniz Cizmeci
- Department of Medicine, Division of Infectious Diseases, Section of Virology, Imperial College London, London, United Kingdom
| | - Yoann Aldon
- Department of Medicine, Division of Infectious Diseases, Section of Virology, Imperial College London, London, United Kingdom
| | - Jeroen Maertzdorf
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - January Weiner
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Stefan He Kaufmann
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - David Jm Lewis
- The NIHR Imperial Clinical Research Facility, Imperial Centre for Translational and Experimental Medicine, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, United Kingdom
| | | | | | - Robin J Shattock
- Department of Medicine, Division of Infectious Diseases, Section of Virology, Imperial College London, London, United Kingdom
| |
Collapse
|
38
|
Wang Y, Cao F, Wang Y, Yu G, Jia BL. Silencing of SAA1 inhibits palmitate- or high-fat diet induced insulin resistance through suppression of the NF-κB pathway. Mol Med 2019; 25:17. [PMID: 31060494 PMCID: PMC6503374 DOI: 10.1186/s10020-019-0075-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Accepted: 03/13/2019] [Indexed: 12/14/2022] Open
Abstract
Background Obesity is one of the leading causes of insulin resistance. Accumulating reports have highlighted that serum amyloid A-1 (SAA1) is a potential candidate that is capable of attenuating insulin resistance. Hence, we conducted the current study with aims of investigating our proposed hypothesis that silencing SAA1 could inhibit the progression of obesity-induced insulin resistance through the NF-κB pathway. Methods Gene expression microarray analysis was initially performed to screen differentially expressed genes (DEGs) associated with obesity. Palmitate (PA)-induced insulin resistance Huh7 cell models and high-fat diet (HFD)-induced mouse models were established to elucidate the effect of SAA1/Saa1 on insulin resistance. The NF-κB pathway-related expression was subsequently determined through the application of reverse transcription quantitative polymerase chain reaction (RT-qPCR) and Western blot analysis. Results Saa1 was identified as an obesity-related gene based on the microarray data of GSE39549. Saa1 was determined to be highly expressed in HFD-induced insulin resistance mouse models. PA-induced Huh7 cells, treated with silenced SAA1 or NF-κB pathway inhibition using BAY 11–7082, displayed a marked decrease in both Saa1 and SOCS3 as well as an elevation in 2DG, IRS1 and the extent of IRS1 phosphorylation. HFD mice treated with silenced Saa1 or inhibited NF-κB pathway exhibited improved fasting blood glucose (FBG) levels as well as fasting plasma insulin (FPI) levels, glucose tolerance and systemic insulin sensitivity. Saa1/SAA1 was determined to show a stimulatory effect on the transport of the NF-κBp65 protein from the cytoplasm to the nucleus both in vivo and in vitro, suggesting that Saa1/SAA1 could activate the NF-κB pathway. Conclusion Taken together, our key findings highlight a novel mechanism by which silencing of SAA1 hinders PA or HFD-induced insulin resistance through inhibition of the NF-κB pathway. Electronic supplementary material The online version of this article (10.1186/s10020-019-0075-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yong Wang
- Department of Gastrointestinal Surgery, the Second Hospital of Anhui Medical University, No. 678, Furong Road, Economic and Technological Development Zone, Hefei, 230601, Anhui Province, People's Republic of China.
| | - Feng Cao
- Department of Gastrointestinal Surgery, the Second Hospital of Anhui Medical University, No. 678, Furong Road, Economic and Technological Development Zone, Hefei, 230601, Anhui Province, People's Republic of China
| | - Yang Wang
- Department of Gastrointestinal Surgery, the Second Hospital of Anhui Medical University, No. 678, Furong Road, Economic and Technological Development Zone, Hefei, 230601, Anhui Province, People's Republic of China
| | - Gang Yu
- Department of Gastrointestinal Surgery, the Second Hospital of Anhui Medical University, No. 678, Furong Road, Economic and Technological Development Zone, Hefei, 230601, Anhui Province, People's Republic of China
| | - Ben-Li Jia
- Department of Gastrointestinal Surgery, the Second Hospital of Anhui Medical University, No. 678, Furong Road, Economic and Technological Development Zone, Hefei, 230601, Anhui Province, People's Republic of China
| |
Collapse
|
39
|
Hirai K, Furusho H, Kawashima N, Xu S, de Beer M, Battaglino R, Van Dyke T, Stashenko P, Sasaki H. Serum Amyloid A Contributes to Chronic Apical Periodontitis via TLR2 and TLR4. J Dent Res 2019; 98:117-125. [PMID: 30189157 PMCID: PMC6304714 DOI: 10.1177/0022034518796456] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
In the current concept of bacterial infections, pathogen-associated molecular patterns (PAMPs) derived from pathogens and damage-associated molecular patterns (DAMPs) released from damaged/necrotic host cells are crucial factors in induction of innate immune responses. However, the implication of DAMPs in apical and marginal periodontitis is unknown. Serum amyloid A (SAA) is a DAMP that is involved in the development of various chronic inflammatory diseases, such as rheumatoid arthritis. In the present study, we tested whether SAA is involved in the pathogenesis of periapical lesions, using human periapical surgical specimens and mice deficient in SAA and Toll-like receptors (TLR). SAA1/2 was locally expressed in human periapical lesions at the mRNA and protein levels. The level of SAA protein appeared to be positively associated with the inflammatory status of the lesions. In the development of mouse periapical inflammation, SAA1.1/2.1 was elevated locally and systemically in wild-type (WT) mice. Although SAA1.1/2.1 double-knockout and SAA3 knockout mice had redundant attenuation of the extent of periapical lesions, these animals showed strikingly improved inflammatory cell infiltration versus WT. Recombinant human SAA1 (rhSAA1) directly induced chemotaxis of WT neutrophils in a dose-dependent manner in vitro. In addition, rhSAA1 stimulation significantly prolonged the survival of WT neutrophils as compared with nonstimulated neutrophils. Furthermore, rhSAA1 activated the NF-κB pathway and subsequent IL-1α production in macrophages in a dose-dependent manner. However, TLR2/TLR4 double deficiency substantially diminished these SAA-mediated proinflammatory responses. Taken together, the SAA-TLR axis plays an important role in the chronicity of periapical inflammation via induction of inflammatory cell infiltration and prolonged cell survival. The interactions of PAMPs and DAMPs require further investigation in dental/oral inflammation.
Collapse
Affiliation(s)
- K. Hirai
- Department of Cariology, Restorative Sciences
and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
- Department of Immunology and Infectious
Diseases, The Forsyth Institute, Cambridge, MA, USA
| | - H. Furusho
- Department of Oral and Maxillofacial
Pathobiology, Hiroshima University, Hiroshima, Japan
| | - N. Kawashima
- Department of Pulp Biology and Endodontics,
Tokyo Medical and Dental University, Tokyo, Japan
| | - S. Xu
- Department of Immunology and Infectious
Diseases, The Forsyth Institute, Cambridge, MA, USA
- Department of Medicine, Brigham and Women’s
Hospital and Harvard Medical School, Boston, MA, USA
| | - M.C. de Beer
- Department of Physiology, University of
Kentucky, Lexington, KY, USA
| | - R. Battaglino
- Department of Physical Medicine and
Rehabilitation, School of Medicine, University of Colorado, Aurora, CO, USA
| | - T. Van Dyke
- The Forsyth Institute, Cambridge, MA,
USA
- Department of Oral Medicine, Infection, and
Immunity, Harvard School of Dental Medicine, Boston, MA, USA
| | - P. Stashenko
- Department of Immunology and Infectious
Diseases, The Forsyth Institute, Cambridge, MA, USA
- Department of Oral Medicine, Infection, and
Immunity, Harvard School of Dental Medicine, Boston, MA, USA
- Department of Endodontics, Goldman School of
Dental Medicine, Boston University, Boston, MA, USA
| | - H. Sasaki
- Department of Cariology, Restorative Sciences
and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
- Department of Immunology and Infectious
Diseases, The Forsyth Institute, Cambridge, MA, USA
- Department of Oral Medicine, Infection, and
Immunity, Harvard School of Dental Medicine, Boston, MA, USA
| |
Collapse
|
40
|
Wilson PG, Thompson JC, Shridas P, McNamara PJ, de Beer MC, de Beer FC, Webb NR, Tannock LR. Serum Amyloid A Is an Exchangeable Apolipoprotein. Arterioscler Thromb Vasc Biol 2018; 38:1890-1900. [PMID: 29976766 PMCID: PMC6202200 DOI: 10.1161/atvbaha.118.310979] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Objective- SAA (serum amyloid A) is a family of acute-phase reactants that have proinflammatory and proatherogenic activities. SAA is more lipophilic than apoA-I (apolipoprotein A-I), and during an acute-phase response, <10% of plasma SAA is found lipid-free. In most reports, SAA is found exclusively associated with high-density lipoprotein; however, we and others have reported SAA on apoB (apolipoprotein B)-containing lipoproteins in both mice and humans. The goal of this study was to determine whether SAA is an exchangeable apolipoprotein. Approach and Results- Delipidated human SAA was incubated with SAA-free human lipoproteins; then, samples were reisolated by fast protein liquid chromatography, and SAA analyzed by ELISA and immunoblot. Both in vitro and in vivo, we show that SAA associates with any lipoprotein and does not remain in a lipid-free form. Although SAA is preferentially found on high-density lipoprotein, it can exchange between lipoproteins. In the presence of CETP (cholesterol ester transfer protein), there is greater exchange of SAA between lipoproteins. Subjects with diabetes mellitus, but not those with metabolic syndrome, showed altered SAA lipoprotein distribution postprandially. Proteoglycan-mediated lipoprotein retention is thought to be an underlying mechanism for atherosclerosis development. SAA has a proteoglycan-binding domain. Lipoproteins containing SAA had increased proteoglycan binding compared with SAA-free lipoproteins. Conclusions- Thus, SAA is an exchangeable apolipoprotein and increases apoB-containing lipoproteins' proteoglycan binding. We and others have previously reported the presence of SAA on low-density lipoprotein in individuals with obesity, diabetes mellitus, and metabolic syndrome. We propose that the presence of SAA on apoB-containing lipoproteins may contribute to cardiovascular disease development in these populations.
Collapse
Affiliation(s)
- Patricia G Wilson
- Department of Veterans Affairs, Lexington, KY
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky
- Barnstable Brown Diabetes Center, College of Medicine, University of Kentucky
| | - Joel C Thompson
- Department of Veterans Affairs, Lexington, KY
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky
- Barnstable Brown Diabetes Center, College of Medicine, University of Kentucky
| | - Preetha Shridas
- Department of Internal Medicine, College of Medicine, University of Kentucky
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky
- Barnstable Brown Diabetes Center, College of Medicine, University of Kentucky
| | - Patrick J McNamara
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky
| | - Maria C de Beer
- Department of Physiology, College of Medicine, University of Kentucky
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky
- Barnstable Brown Diabetes Center, College of Medicine, University of Kentucky
| | - Frederick C de Beer
- Department of Internal Medicine, College of Medicine, University of Kentucky
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky
- Barnstable Brown Diabetes Center, College of Medicine, University of Kentucky
| | - Nancy R Webb
- Department of Veterans Affairs, Lexington, KY
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky
- Barnstable Brown Diabetes Center, College of Medicine, University of Kentucky
| | - Lisa R Tannock
- Department of Veterans Affairs, Lexington, KY
- Department of Internal Medicine, College of Medicine, University of Kentucky
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky
- Barnstable Brown Diabetes Center, College of Medicine, University of Kentucky
| |
Collapse
|
41
|
Shridas P, De Beer MC, Webb NR. High-density lipoprotein inhibits serum amyloid A-mediated reactive oxygen species generation and NLRP3 inflammasome activation. J Biol Chem 2018; 293:13257-13269. [PMID: 29976759 DOI: 10.1074/jbc.ra118.002428] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 06/18/2018] [Indexed: 12/11/2022] Open
Abstract
Serum amyloid A (SAA) is a high-density apolipoprotein whose plasma levels can increase more than 1000-fold during a severe acute-phase inflammatory response and are more modestly elevated in chronic inflammation. SAA is thought to play important roles in innate immunity, but its biological activities have not been completely delineated. We previously reported that SAA deficiency protects mice from developing abdominal aortic aneurysms (AAAs) induced by chronic angiotensin II (AngII) infusion. Here, we report that SAA is required for AngII-induced increases in interleukin-1β (IL-1β), a potent proinflammatory cytokine that is tightly controlled by the Nod-like receptor protein 3 (NLRP3) inflammasome and caspase-1 and has been implicated in both human and mouse AAAs. We determined that purified SAA stimulates IL-1β secretion in murine J774 and bone marrow-derived macrophages through a mechanism that depends on NLRP3 expression and caspase-1 activity, but is independent of P2X7 nucleotide receptor (P2X7R) activation. Inhibiting reactive oxygen species (ROS) by N-acetyl-l-cysteine or mito-TEMPO and inhibiting activation of cathepsin B by CA-074 blocked SAA-mediated inflammasome activation and IL-1β secretion. Moreover, inhibiting cellular potassium efflux with glyburide or increasing extracellular potassium also significantly reduced SAA-mediated IL-1β secretion. Of note, incorporating SAA into high-density lipoprotein (HDL) prior to its use in cell treatments completely abolished its ability to stimulate ROS generation and inflammasome activation. These results provide detailed insights into SAA-mediated IL-1β production and highlight HDL's role in regulating SAA's proinflammatory effects.
Collapse
Affiliation(s)
- Preetha Shridas
- From the Departments of Internal Medicine, .,Barnstable Brown Diabetes Center, University of Kentucky, Lexington, Kentucky 40536
| | - Maria C De Beer
- Barnstable Brown Diabetes Center, University of Kentucky, Lexington, Kentucky 40536.,Physiology, and.,Pharmacology and Nutritional Sciences
| | - Nancy R Webb
- Barnstable Brown Diabetes Center, University of Kentucky, Lexington, Kentucky 40536.,Pharmacology and Nutritional Sciences.,Saha Cardiovascular Research Center, and
| |
Collapse
|
42
|
Choudhary S, Santone E, Yee SP, Lorenzo J, Adams DJ, Goetjen A, McCarthy MB, Mazzocca AD, Pilbeam C. Continuous PTH in Male Mice Causes Bone Loss Because It Induces Serum Amyloid A. Endocrinology 2018; 159:2759-2776. [PMID: 29757436 PMCID: PMC6692876 DOI: 10.1210/en.2018-00265] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 05/05/2018] [Indexed: 12/12/2022]
Abstract
Increased bone resorption is considered to explain why intermittent PTH is anabolic for bone but continuous PTH is catabolic. However, when cyclooxygenase-2 (COX2) is absent in mice, continuous PTH becomes anabolic without decreased resorption. In murine bone marrow stromal cells (BMSCs), serum amyloid A (SAA)3, induced in the hematopoietic lineage by the combination of COX2-produced prostaglandin and receptor activator of nuclear factor κB ligand (RANKL), suppresses PTH-stimulated osteoblast differentiation. To determine whether SAA3 inhibits the anabolic effects of PTH in vivo, wild-type (WT) and SAA3 knockout (KO) mice were infused with PTH. In WT mice, continuous PTH induced SAA3 and was catabolic for bone. In KO mice, PTH was anabolic, increasing trabecular bone, serum markers of bone formation, and osteogenic gene expression. In contrast, PTH increased all measurements associated with bone resorption, as well as COX2 gene expression, similarly in KO and WT mice. SAA1 and SAA2 in humans are likely to have analogous functions to SAA3 in mice. RANKL induced both SAA1 and SAA2 in human bone marrow macrophages in a COX2-dependent manner. PTH stimulated osteogenesis in human BMSCs only when COX2 or RANKL was inhibited. Addition of recombinant SAA1 or SAA2 blocked PTH-stimulated osteogenesis. In summary, SAA3 suppresses the bone formation responses but not the bone resorption responses to PTH in mice, and in the absence of SAA3, continuous PTH is anabolic. In vitro studies in human bone marrow suggest that SAA may be a target for enhancing the therapeutic effects of PTH in treating osteoporosis.
Collapse
Affiliation(s)
- Shilpa Choudhary
- Musculoskeletal Institute, UConn Health, Farmington, Connecticut
- Department of Medicine, UConn Health, Farmington, Connecticut
| | | | - Sui-Pok Yee
- Department of Cell Biology, UConn Health, Farmington, Connecticut
- Center for Mouse Genome Modification, UConn Health, Farmington, Connecticut
| | - Joseph Lorenzo
- Musculoskeletal Institute, UConn Health, Farmington, Connecticut
- Department of Medicine, UConn Health, Farmington, Connecticut
| | - Douglas J Adams
- Musculoskeletal Institute, UConn Health, Farmington, Connecticut
| | | | | | | | - Carol Pilbeam
- Musculoskeletal Institute, UConn Health, Farmington, Connecticut
- Department of Medicine, UConn Health, Farmington, Connecticut
| |
Collapse
|
43
|
Ather JL, Poynter ME. Serum amyloid A3 is required for normal weight and immunometabolic function in mice. PLoS One 2018; 13:e0192352. [PMID: 29390039 PMCID: PMC5794179 DOI: 10.1371/journal.pone.0192352] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 01/22/2018] [Indexed: 02/06/2023] Open
Abstract
Serum amyloid A (SAA) is an apolipoprotein that is robustly upregulated in numerous inflammatory diseases and has been implicated as a candidate pro-inflammatory mediator. However, studies comparing endogenous SAAs and recombinant forms of the acute phase protein have generated conflicting data on the function of SAA in immunity. We generated SAA3 knockout mice to evaluate the contribution of SAA3 to immune-mediated disease, and found that mice lacking SAA3 develop adult-onset obesity and metabolic dysfunction along with defects in innate immune development. Mice that lack SAA3 gain more weight, exhibit increased visceral adipose deposition, and develop hepatic steatosis compared to wild-type littermates. Leukocytes from the adipose tissue of SAA3-/- mice express a pro-inflammatory phenotype, and bone marrow derived dendritic cells from mice lacking SAA3 secrete increased levels of IL-1β, IL-6, IL-23, and TNFα in response to LPS compared to cells from wild-type mice. Finally, BMDC lacking SAA3 demonstrate an impaired endotoxin tolerance response and inhibited responses to retinoic acid. Our findings indicate that endogenous SAA3 modulates metabolic and immune homeostasis.
Collapse
Affiliation(s)
- Jennifer L. Ather
- Vermont Lung Center, Division of Pulmonary Disease and Critical Care, Department of Medicine, University of Vermont, Burlington, VT, United States of America
| | - Matthew E. Poynter
- Vermont Lung Center, Division of Pulmonary Disease and Critical Care, Department of Medicine, University of Vermont, Burlington, VT, United States of America
| |
Collapse
|