Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 133
Filter
1.
Reprod Biol Endocrinol ; 20(1): 4, 2022 Jan 03.
Article in English | MEDLINE | ID: mdl-34980155

ABSTRACT

BACKGROUND: Insulin resistance (IR) contributes to ovarian dysfunctions in polycystic ovarian syndrome (PCOS) patients. Serum amyloid A1 (SAA1) is an acute phase protein produced primarily by the liver in response to inflammation. In addition to its role in inflammation, SAA1 may participate in IR development in peripheral tissues. Yet, expressional regulation of SAA1 in the ovary and its role in the pathogenesis of ovarian IR in PCOS remain elusive. METHODS: Follicular fluid, granulosa cells and peripheral venous blood were collected from PCOS and non-PCOS patients with and without IR to measure SAA1 abundance for analysis of its correlation with IR status. The effects of SAA1 on its own expression and insulin signaling pathway were investigated in cultured primary granulosa cells. RESULTS: Ovarian granulosa cells were capable of producing SAA1, which could be induced by SAA1 per se. Moreover, the abundance of SAA1 significantly increased in granulosa cells and follicular fluid in PCOS patients with IR. SAA1 treatment significantly attenuated insulin-stimulated membrane translocation of glucose transporter 4 and glucose uptake in granulosa cells through induction of phosphatase and tensin homolog deleted on chromosome 10 (PTEN) expression with subsequent inhibition of Akt phosphorylation. These effects of SAA1 could be blocked by inhibitors for toll-like receptors 2/4 (TLR 2/4) and nuclear factor kappa light chain enhancer of activated B (NF-κB). CONCLUSIONS: Human granulosa cells are capable of feedforward production of SAA1, which significantly increased in PCOS patients with IR. Excessive SAA1 reduces insulin sensitivity in granulosa cells via induction of PTEN and subsequent inhibition of Akt phosphorylation upon activation of TLR2/4 and NF-κB pathway. These findings highlight that elevation of SAA1 in the ovary promotes the development of IR in granulosa cells of PCOS patients.


Subject(s)
Granulosa Cells/metabolism , Insulin Resistance/genetics , Polycystic Ovary Syndrome/genetics , Serum Amyloid A Protein/physiology , Adult , Case-Control Studies , Cells, Cultured , Female , Follicular Fluid/chemistry , Follicular Fluid/metabolism , Granulosa Cells/drug effects , Humans , Ovary/drug effects , Ovary/metabolism , Polycystic Ovary Syndrome/blood , Polycystic Ovary Syndrome/metabolism , Serum Amyloid A Protein/genetics , Serum Amyloid A Protein/metabolism , Serum Amyloid A Protein/pharmacology
2.
Sci Rep ; 10(1): 10397, 2020 06 25.
Article in English | MEDLINE | ID: mdl-32587356

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.


Subject(s)
Adipose Tissue/metabolism , Gene Expression Regulation , Inflammation/pathology , Lipopolysaccharides/toxicity , Serum Amyloid A Protein/physiology , Silver Nitrate/toxicity , Animals , Anti-Infective Agents, Local/pharmacology , Anti-Infective Agents, Local/toxicity , Inflammation/blood , Inflammation/chemically induced , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
3.
PLoS Pathog ; 15(3): e1007381, 2019 03.
Article in English | MEDLINE | ID: mdl-30845179

ABSTRACT

The intestinal microbiota influences the development and function of myeloid lineages such as neutrophils, but the underlying molecular mechanisms are unresolved. Using gnotobiotic zebrafish, we identified the immune effector Serum amyloid A (Saa) as one of the most highly induced transcripts in digestive tissues following microbiota colonization. Saa is a conserved secreted protein produced in the intestine and liver with described effects on neutrophils in vitro, however its in vivo functions remain poorly defined. We engineered saa mutant zebrafish to test requirements for Saa on innate immunity in vivo. Zebrafish mutant for saa displayed impaired neutrophil responses to wounding but augmented clearance of pathogenic bacteria. At baseline, saa mutants exhibited moderate neutrophilia and altered neutrophil tissue distribution. Molecular and functional analyses of isolated neutrophils revealed that Saa suppresses expression of pro-inflammatory markers and bactericidal activity. Saa's effects on neutrophils depended on microbiota colonization, suggesting this protein mediates the microbiota's effects on host innate immunity. To test tissue-specific roles of Saa on neutrophil function, we over-expressed saa in the intestine or liver and found that sufficient to partially complement neutrophil phenotypes observed in saa mutants. These results indicate Saa produced by the intestine in response to microbiota serves as a systemic signal to neutrophils to restrict aberrant activation, decreasing inflammatory tone and bacterial killing potential while simultaneously enhancing their ability to migrate to wounds.


Subject(s)
Neutrophil Activation/physiology , Serum Amyloid A Protein/physiology , Zebrafish/microbiology , Animals , Immunity, Innate/physiology , Intestines , Liver , Microbiota , Neutrophils/physiology , Serum Amyloid A Protein/metabolism , Signal Transduction
4.
Sci Rep ; 9(1): 3102, 2019 02 28.
Article in English | MEDLINE | ID: mdl-30816210

ABSTRACT

Complex associations exist between inflammation and thrombosis, with the inflammatory state tending to promote coagulation. Fibrinogen, an acute phase protein, has been shown to interact with the amyloidogenic ß-amyloid protein of Alzheimer's disease. However, little is known about the association between fibrinogen and serum amyloid A (SAA), a highly fibrillogenic protein that is one of the most dramatically changing acute phase reactants in the circulation. To study the role of SAA in coagulation and thrombosis, in vitro experiments were performed where purified human SAA, in concentrations resembling a modest acute phase response, was added to platelet-poor plasma (PPP) and whole blood (WB), as well as purified and fluorescently labelled fibrinogen. Results from thromboelastography (TEG) suggest that SAA causes atypical coagulation with a fibrin(ogen)-mediated increase in coagulation, but a decreased platelet/fibrin(ogen) interaction. In WB scanning electron microscopy analysis, SAA mediated red blood cell (RBC) agglutination, platelet activation and clumping, but not platelet spreading. Following clot formation in PPP, the presence of SAA increased amyloid formation of fibrin(ogen) as determined both with auto-fluorescence and with fluorogenic amyloid markers, under confocal microcopy. SAA also binds to fibrinogen, as determined with a fluorescent-labelled SAA antibody and correlative light electron microscopy (CLEM). The data presented here indicate that SAA can affect coagulation by inducing amyloid formation in fibrin(ogen), as well as by propelling platelets to a more prothrombotic state. The discovery of these multiple and complex effects of SAA on coagulation invite further mechanistic analyses.


Subject(s)
Acute-Phase Reaction/metabolism , Amyloid/metabolism , Blood Platelets/metabolism , Fibrinogen/metabolism , Serum Amyloid A Protein/physiology , Thrombosis/metabolism , Adult , Agglutination , Alzheimer Disease/metabolism , Blood Coagulation , Blood Platelets/pathology , Female , Humans , Middle Aged , Platelet Activation , Platelet Aggregation , Protein Binding
5.
Mol Med ; 24(1): 46, 2018 08 30.
Article in English | MEDLINE | ID: mdl-30165816

ABSTRACT

Serum amyloid A (SAA) proteins were isolated and named over 50 years ago. They are small (104 amino acids) and have a striking relationship to the acute phase response with serum levels rising as much as 1000-fold in 24 hours. SAA proteins are encoded in a family of closely-related genes and have been remarkably conserved throughout vertebrate evolution. Amino-terminal fragments of SAA can form highly organized, insoluble fibrils that accumulate in "secondary" amyloid disease. Despite their evolutionary preservation and dynamic synthesis pattern SAA proteins have lacked well-defined physiologic roles. However, considering an array of many, often unrelated, reports now permits a more coordinated perspective. Protein studies have elucidated basic SAA structure and fibril formation. Appreciating SAA's lipophilicity helps relate it to lipid transport and metabolism as well as atherosclerosis. SAA's function as a cytokine-like protein has become recognized in cell-cell communication as well as feedback in inflammatory, immunologic, neoplastic and protective pathways. SAA likely has a critical role in control and possibly propagation of the primordial acute phase response. Appreciating the many cellular and molecular interactions for SAA suggests possibilities for improved understanding of pathophysiology as well as treatment and disease prevention.


Subject(s)
Serum Amyloid A Protein/physiology , Acute-Phase Reaction , Animals , Atherosclerosis/metabolism , Collagenases/metabolism , Humans , Lipid Metabolism , Mammary Glands, Human/metabolism , Maternal Health , Matrix Metalloproteinases/metabolism , Neoplasms/metabolism , Sarcoidosis, Pulmonary/metabolism
6.
J Biol Chem ; 293(34): 13257-13269, 2018 08 24.
Article in English | MEDLINE | ID: mdl-29976759

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-1ß (IL-1ß), 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-1ß 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-1ß secretion. Moreover, inhibiting cellular potassium efflux with glyburide or increasing extracellular potassium also significantly reduced SAA-mediated IL-1ß 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-1ß production and highlight HDL's role in regulating SAA's proinflammatory effects.


Subject(s)
Inflammasomes/metabolism , Inflammation/immunology , Lipoproteins, HDL/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Reactive Oxygen Species/metabolism , Serum Amyloid A Protein/metabolism , Serum Amyloid A Protein/physiology , Animals , Caspase 1/metabolism , Cathepsin B/metabolism , HEK293 Cells , Humans , Inflammation/metabolism , Inflammation/pathology , Interleukin-1beta/metabolism , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Serum Amyloid A Protein/genetics , Signal Transduction
7.
PLoS One ; 13(2): e0192352, 2018.
Article in English | MEDLINE | ID: mdl-29390039

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-1ß, 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.


Subject(s)
Body Weight , Immune System/physiology , Serum Amyloid A Protein/physiology , Animals , Dendritic Cells/immunology , Diet, High-Fat , Homeostasis , Insulin/physiology , Intra-Abdominal Fat/metabolism , Lipopolysaccharides/administration & dosage , Mice , Mice, Knockout , Obesity/immunology , Obesity/metabolism , Serum Amyloid A Protein/genetics
8.
Proc Natl Acad Sci U S A ; 115(6): E1147-E1156, 2018 02 06.
Article in English | MEDLINE | ID: mdl-29351990

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is characterized by the presence of abundant desmoplastic stroma primarily composed of cancer-associated fibroblasts (CAFs). It is generally accepted that CAFs stimulate tumor progression and might be implicated in drug resistance and immunosuppression. Here, we have compared the transcriptional profile of PDGFRα+ CAFs isolated from genetically engineered mouse PDAC tumors with that of normal pancreatic fibroblasts to identify genes potentially implicated in their protumorigenic properties. We report that the most differentially expressed gene, Saa3, a member of the serum amyloid A (SAA) apolipoprotein family, is a key mediator of the protumorigenic activity of PDGFRα+ CAFs. Whereas Saa3-competent CAFs stimulate the growth of tumor cells in an orthotopic model, Saa3-null CAFs inhibit tumor growth. Saa3 also plays a role in the cross talk between CAFs and tumor cells. Ablation of Saa3 in pancreatic tumor cells makes them insensitive to the inhibitory effect of Saa3-null CAFs. As a consequence, germline ablation of Saa3 does not prevent PDAC development in mice. The protumorigenic activity of Saa3 in CAFs is mediated by Mpp6, a member of the palmitoylated membrane protein subfamily of the peripheral membrane-associated guanylate kinases (MAGUK). Finally, we interrogated whether these observations could be translated to a human scenario. Indeed, SAA1, the ortholog of murine Saa3, is overexpressed in human CAFs. Moreover, high levels of SAA1 in the stromal component correlate with worse survival. These findings support the concept that selective inhibition of SAA1 in CAFs may provide potential therapeutic benefit to PDAC patients.


Subject(s)
Cancer-Associated Fibroblasts/pathology , Carcinoma, Pancreatic Ductal/pathology , Pancreas/pathology , Pancreatic Neoplasms/pathology , Serum Amyloid A Protein/metabolism , Serum Amyloid A Protein/physiology , Stromal Cells/pathology , Animals , Cancer-Associated Fibroblasts/metabolism , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Cell Movement , Cell Proliferation , Female , High-Throughput Nucleotide Sequencing , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Pancreas/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Serum Amyloid A Protein/genetics , Stromal Cells/metabolism , Tumor Microenvironment
9.
Inflamm Bowel Dis ; 23(9): 1544-1554, 2017 09.
Article in English | MEDLINE | ID: mdl-28806280

ABSTRACT

BACKGROUND: Inflammatory bowel diseases (IBD) are believed to be driven by dysregulated interactions between the host and the gut microbiota. Our goal is to characterize and infer relationships between mucosal T cells, the host tissue environment, and microbial communities in patients with IBD who will serve as basis for mechanistic studies on human IBD. METHODS: We characterized mucosal CD4 T cells using flow cytometry, along with matching mucosal global gene expression and microbial communities data from 35 pinch biopsy samples from patients with IBD. We analyzed these data sets using an integrated framework to identify predictors of inflammatory states and then reproduced some of the putative relationships formed among these predictors by analyzing data from the pediatric RISK cohort. RESULTS: We identified 26 predictors from our combined data set that were effective in distinguishing between regions of the intestine undergoing active inflammation and regions that were normal. Network analysis on these 26 predictors revealed SAA1 as the most connected node linking the abundance of the genus Bacteroides with the production of IL17 and IL22 by CD4 T cells. These SAA1-linked microbial and transcriptome interactions were further reproduced with data from the pediatric IBD RISK cohort. CONCLUSIONS: This study identifies expression of SAA1 as an important link between mucosal T cells, microbial communities, and their tissue environment in patients with IBD. A combination of T cell effector function data, gene expression and microbial profiling can distinguish between intestinal inflammatory states in IBD regardless of disease types.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Gastrointestinal Microbiome/immunology , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/immunology , Serum Amyloid A Protein/physiology , Adult , Biopsy , Case-Control Studies , Child , Colon/immunology , Colon/microbiology , Colon/pathology , Gene Expression , Humans , Immunity, Cellular , Inflammatory Bowel Diseases/pathology , Interleukin-17/biosynthesis , Interleukins/biosynthesis , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Th17 Cells/immunology , Interleukin-22
10.
Vet Immunol Immunopathol ; 187: 10-13, 2017 May.
Article in English | MEDLINE | ID: mdl-28494923

ABSTRACT

Serum amyloid A (SAA) concentration and plasma matrix metalloproteinase-9 (MMP-9) levels are increased in cats with lymphoma. In the present study, the association between SAA and MMP-9 production was evaluated using recombinant feline SAA (rfSAA) and three feline lymphoma-derived cell lines: 3201, MS4, and MCC. MMP-9 mRNA expression was significantly increased by rfSAA stimulation only in MCC cells. Secreted MMP-9 protein in culture media was confirmed by gelatin zymography, with clear bands of MMP-9 detected in MCC cells following rfSAA stimulation. A significant increase in semi-quantified MMP-9 levels was observed with 5 and 25µg/ml of rfSAA stimulation. The infiltrative activities of feline lymphoma cells, assessed by the matrigel transwell assay, showed that rfSAA stimulated cell infiltration in MCC cells, in addition to MMP-9 expression. Although the response to rfSAA stimulation varied between cell lines, the results showed that rfSAA can stimulate MMP-9 production and infiltration of feline lymphoma-derived cells. The findings of this study have identified a novel role for SAA in the progression of some forms of feline lymphoma.


Subject(s)
Cat Diseases/metabolism , Lymphoma/veterinary , Matrix Metalloproteinase 9/metabolism , Serum Amyloid A Protein/physiology , Animals , Cats , Cell Line, Tumor , Lymphoma/metabolism
11.
Cancer Res ; 77(7): 1586-1598, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28202524

ABSTRACT

Mechanisms of cross-talk between tumor cells and tumor-associated macrophages (TAM), which drive metastasis, are not fully understood. Scavenger receptor A1 (SR-A1) expressed primarily in macrophages has been associated with lung tumorigenesis. In this study, we used population genetics, transcriptomics, and functional analyses to uncover how SR-A1 is involved in lung cancer and its prognosis. SR-A1 genetic variants were investigated for possible association with survival of advanced stage NSCLC patients in the Harvard Lung Cancer Study cohort. Two SNPs (rs17484273, rs1484751) in SR-A1 were associated significantly with poor overall survival in this cohort. Data from The Cancer Genome Atlas showed considerable downregulation of SR-A1 in lung tumor tissues. The association of SR-A1 with prognosis was validated in animal models in the context of lung cancer metastasis. Macrophages derived from mice genetically deficient for SR-A1 exhibited accelerated metastasis in a model of lung cancer. On the other hand, tumor cell seeding, migration, and invasion, as well as macrophage accumulation in lung cancer tissue, were enhanced in SR-A1-deficient mice. SR-A1 deletion upregulated serum amyloid A1 (SAA1) in macrophages via MAPK/IκB/NFκB signaling. SAA1 promoted tumor cell invasion and macrophage migration in vitro and in vivo, but these effects were blocked by administration of an anti-SAA1 antibody. Overall, our findings show how SR-A1 suppresses lung cancer metastasis by downregulating SAA1 production in TAMs. Cancer Res; 77(7); 1586-98. ©2017 AACR.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , Macrophages/physiology , Neoplasm Metastasis/prevention & control , Scavenger Receptors, Class A/physiology , Serum Amyloid A Protein/physiology , Animals , Carcinoma, Non-Small-Cell Lung/mortality , Cell Movement , Female , Humans , Lung Neoplasms/mortality , Mice , Neoplasm Invasiveness , Polymorphism, Single Nucleotide , Prognosis , Scavenger Receptors, Class A/genetics , Tumor Microenvironment
12.
J Lipid Res ; 57(12): 2138-2149, 2016 12.
Article in English | MEDLINE | ID: mdl-27744369

ABSTRACT

Oxidative stress and inflammation, which involve a dramatic increase in serum amyloid A (SAA) levels, are critical in the development of atherosclerosis. Most SAA circulates on plasma HDL particles, altering their cardioprotective properties. SAA-enriched HDL has diminished anti-oxidant effects on LDL, which may contribute to atherogenesis. We determined combined effects of SAA enrichment and oxidation on biochemical changes in HDL. Normal human HDLs were incubated with SAA, oxidized by various factors (Cu2+, myeloperoxidase, H2O2, OCl-), and analyzed for lipid and protein modifications and biophysical remodeling. Three novel findings are reported: addition of SAA reduces oxidation of HDL and LDL lipids; oxidation of SAA-containing HDL in the presence of OCl- generates a covalent heterodimer of SAA and apoA-I that resists the release from HDL; and mild oxidation promotes spontaneous release of proteins (SAA and apoA-I) from SAA-enriched HDL. We show that the anti-oxidant effects of SAA extend to various oxidants and are mediated mainly by the unbound protein. We propose that free SAA sequesters lipid hydroperoxides and delays lipoprotein oxidation, though much less efficiently than other anti-oxidant proteins, such as apoA-I, that SAA displaces from HDL. These findings prompt us to reconsider the role of SAA in lipid oxidation in vivo.


Subject(s)
Antioxidants/chemistry , Lipoproteins, HDL/chemistry , Lipoproteins, LDL/chemistry , Serum Amyloid A Protein/chemistry , Animals , Antioxidants/physiology , Apolipoprotein A-I/chemistry , Copper/chemistry , Humans , Lipid Peroxidation , Mice , Peroxidase/chemistry , Serum Amyloid A Protein/physiology
13.
Infect Immun ; 84(10): 2824-32, 2016 10.
Article in English | MEDLINE | ID: mdl-27456830

ABSTRACT

Intestinal segmented filamentous bacteria (SFB) protect from ameba infection, and protection is transferable with bone marrow dendritic cells (BMDCs). SFB cause an increase in serum amyloid A (SAA), suggesting that SAA might mediate SFB's effects on BMDCs. Here we further explored the role of bone marrow in SFB-mediated protection. Transient gut colonization with SFB or SAA administration alone transiently increased the H3K27 histone demethylase Jmjd3, persistently increased bone marrow Csf2ra expression and granulocyte monocyte precursors (GMPs), and protected from ameba infection. Pharmacologic inhibition of Jmjd3 H3K27 demethylase activity during SAA treatment or blockade of granulocyte-macrophage colony-stimulating factor (GM-CSF) signaling in SFB-colonized mice prevented GMP expansion, decreased gut neutrophils, and blocked protection from ameba infection. These results indicate that alteration of the microbiota and systemic exposure to SAA can influence myelopoiesis and susceptibility to amebiasis via epigenetic mechanisms. Gut microbiota-marrow communication is a previously unrecognized mechanism of innate protection from infection.


Subject(s)
Bone Marrow Cells/cytology , Entamoeba histolytica/physiology , Entamoebiasis/physiopathology , Gastrointestinal Tract/microbiology , Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Serum Amyloid A Protein/physiology , Animals , Bacteria , Bone Marrow/metabolism , Bone Marrow Cells/physiology , Dendritic Cells/metabolism , Disease Models, Animal , Granulocyte-Macrophage Progenitor Cells , Jumonji Domain-Containing Histone Demethylases/metabolism , Male , Mice , Mice, Inbred C57BL , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism
14.
J. physiol. biochem ; 72(1): 71-81, mar. 2016.
Article in English | IBECS | ID: ibc-168208

ABSTRACT

Serum amyloid A (SAA), a major acute-phase reactant, modulates angiogenesis in many diseases. Vascular endothelial growth factor receptor 2 (VEGFR2) is the primary angiogenic receptor for vascular endothelial growth factor (VEGF), but the possibility of an interaction between SAA and VEGFR2 has not yet been resolved. Here, we investigated if SAA stimulates the expression of VEGFR2 and promotes angiogenesis in vitro. Human umbilical vein endothelial cells (HUVECs) were stimulated with recombinant SAA (rSAA), and the messenger RNA (mRNA) and protein expression of VEGFR2 was detected by Western blot analysis and quantitative real-time PCR. Formyl peptide receptor-like 1 (FPRL1) agonist (WKYMVm) and antagonist (WRW4) and inhibitors of mitogen-activated protein kinases (MAPKs) were used to investigate the mechanism of regulation of VEGFR2.We show that SAA induces VEGFR2 expression in a time- and dose-dependent manner in HUVECs. In addition, SAA promotes tube formation in HUVECs. The effect of SAA on tube formation was shown to be the result of an increase in VEGFR2 expression, which was blocked by the multi-angiokinase receptor inhibitor BIBF1120. These activities of SAA appear to be mediated by FPRL1/MAPK signaling pathways, as they were mimicked by WKYMVm and abrogated by WRW4 and inhibitors of MAPKs. These observations indicate that SAA induces VEGFR2 expression and promotes tube formation in HUVECs via the FPRL1/MAPK signaling pathway, thus providing a potential target for the control of angiogénesis (AU)


No disponible


Subject(s)
Humans , Neovascularization, Physiologic , Vascular Endothelial Growth Factor Receptor-2/physiology , Serum Amyloid A Protein/physiology , Human Umbilical Vein Endothelial Cells
15.
J Clin Invest ; 126(1): 266-81, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26642365

ABSTRACT

HDL from healthy humans and lean mice inhibits palmitate-induced adipocyte inflammation; however, the effect of the inflammatory state on the functional properties of HDL on adipocytes is unknown. Here, we found that HDL from mice injected with AgNO3 fails to inhibit palmitate-induced inflammation and reduces cholesterol efflux from 3T3-L1 adipocytes. Moreover, HDL isolated from obese mice with moderate inflammation and humans with systemic lupus erythematosus had similar effects. Since serum amyloid A (SAA) concentrations in HDL increase with inflammation, we investigated whether elevated SAA is a causal factor in HDL dysfunction. HDL from AgNO3-injected mice lacking Saa1.1 and Saa2.1 exhibited a partial restoration of antiinflammatory and cholesterol efflux properties in adipocytes. Conversely, incorporation of SAA into HDL preparations reduced antiinflammatory properties but not to the same extent as HDL from AgNO3-injected mice. SAA-enriched HDL colocalized with cell surface-associated extracellular matrix (ECM) of adipocytes, suggesting impaired access to the plasma membrane. Enzymatic digestion of proteoglycans in the ECM restored the ability of SAA-containing HDL to inhibit palmitate-induced inflammation and cholesterol efflux. Collectively, these findings indicate that inflammation results in a loss of the antiinflammatory properties of HDL on adipocytes, which appears to partially result from the SAA component of HDL binding to cell-surface proteoglycans, thereby preventing access of HDL to the plasma membrane.


Subject(s)
Lipoproteins, HDL/physiology , Serum Amyloid A Protein/physiology , 3T3-L1 Cells , Adipocytes/metabolism , Animals , C-Reactive Protein/analysis , Cholesterol/metabolism , Humans , Inflammation/prevention & control , Male , Mice , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , Silver Nitrate/pharmacology , Toll-Like Receptor 4/physiology
16.
J Physiol Biochem ; 72(1): 71-81, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26714823

ABSTRACT

Serum amyloid A (SAA), a major acute-phase reactant, modulates angiogenesis in many diseases. Vascular endothelial growth factor receptor 2 (VEGFR2) is the primary angiogenic receptor for vascular endothelial growth factor (VEGF), but the possibility of an interaction between SAA and VEGFR2 has not yet been resolved. Here, we investigated if SAA stimulates the expression of VEGFR2 and promotes angiogenesis in vitro. Human umbilical vein endothelial cells (HUVECs) were stimulated with recombinant SAA (rSAA), and the messenger RNA (mRNA) and protein expression of VEGFR2 was detected by Western blot analysis and quantitative real-time PCR. Formyl peptide receptor-like 1 (FPRL1) agonist (WKYMVm) and antagonist (WRW(4)) and inhibitors of mitogen-activated protein kinases (MAPKs) were used to investigate the mechanism of regulation of VEGFR2.We show that SAA induces VEGFR2 expression in a time- and dose-dependent manner in HUVECs. In addition, SAA promotes tube formation in HUVECs. The effect of SAA on tube formation was shown to be the result of an increase in VEGFR2 expression, which was blocked by the multi-angiokinase receptor inhibitor BIBF1120. These activities of SAA appear to be mediated by FPRL1/MAPK signaling pathways, as they were mimicked by WKYMVm and abrogated by WRW(4) and inhibitors of MAPKs. These observations indicate that SAA induces VEGFR2 expression and promotes tube formation in HUVECs via the FPRL1/MAPK signaling pathway, thus providing a potential target for the control of angiogenesis.


Subject(s)
Neovascularization, Physiologic , Serum Amyloid A Protein/physiology , Vascular Endothelial Growth Factor Receptor-2/physiology , Human Umbilical Vein Endothelial Cells , Humans
17.
Eur Heart J ; 36(43): 3007-16, 2015 Nov 14.
Article in English | MEDLINE | ID: mdl-26248570

ABSTRACT

AIMS: High-density lipoproteins (HDLs) are considered as anti-atherogenic. Recent experimental findings suggest that their biological properties can be modified in certain clinical conditions by accumulation of serum amyloid A (SAA). The effect of SAA on the association between HDL-cholesterol (HDL-C) and cardiovascular outcome remains unknown. METHODS AND RESULTS: We examined the association of SAA and HDL-C with mortality in the Ludwigshafen Risk and Cardiovascular Health (LURIC) study, which included 3310 patients undergoing coronary angiography. To validate our findings, we analysed 1255 participants of the German Diabetes and Dialysis study (4D) and 4027 participants of the Cooperative Health Research in the Region of Augsburg (KORA) S4 study. In LURIC, SAA concentrations predicted all-cause and cardiovascular mortality. In patients with low SAA, higher HDL-C was associated with lower all-cause and cardiovascular mortality. In contrast, in patients with high SAA, higher HDL-C was associated with increased all-cause and cardiovascular mortality, indicating that SAA indeed modifies the beneficial properties of HDL. We complemented these clinical observations by in vitro experiments, in which SAA impaired vascular functions of HDL. We further derived a formula for the simple calculation of the amount of biologically 'effective' HDL-C based on measured HDL-C and SAA from the LURIC study. In 4D and KORA S4 studies, we found that measured HDL-C was not associated with clinical outcomes, whereas calculated 'effective' HDL-C significantly predicted better outcome. CONCLUSION: The acute-phase protein SAA modifies the biological effects of HDL-C in several clinical conditions. The concomitant measurement of SAA is a simple, useful, and clinically applicable surrogate for the vascular functionality of HDL.


Subject(s)
Cardiovascular Diseases/mortality , Cholesterol, HDL/metabolism , Serum Amyloid A Protein/metabolism , Acute Coronary Syndrome/mortality , Adult , Aged , Aorta/metabolism , Biomarkers/metabolism , Cardiovascular Diseases/blood , Cause of Death , Cells, Cultured , Diabetes Mellitus, Type 2/mortality , Diabetic Nephropathies/mortality , Endothelium, Vascular/metabolism , Female , Humans , Kidney Failure, Chronic/mortality , Male , Middle Aged , Nitric Oxide/biosynthesis , Prognosis , Prospective Studies , Reactive Oxygen Species/metabolism , Renal Dialysis/mortality , Risk Factors , Serum Amyloid A Protein/physiology , Vascular Cell Adhesion Molecule-1/metabolism
18.
Oncogene ; 34(4): 424-35, 2015 Jan 22.
Article in English | MEDLINE | ID: mdl-24469032

ABSTRACT

S100A4 is implicated in metastasis and chronic inflammation, but its function remains uncertain. Here we establish an S100A4-dependent link between inflammation and metastatic tumor progression. We found that the acute-phase response proteins serum amyloid A (SAA) 1 and SAA3 are transcriptional targets of S100A4 via Toll-like receptor 4 (TLR4)/nuclear factor-κB signaling. SAA proteins stimulated the transcription of RANTES (regulated upon activation normal T-cell expressed and presumably secreted), G-CSF (granulocyte-colony-stimulating factor) and MMP2 (matrix metalloproteinase 2), MMP3, MMP9 and MMP13. We have also shown for the first time that SAA stimulate their own transcription as well as that of proinflammatory S100A8 and S100A9 proteins. Moreover, they strongly enhanced tumor cell adhesion to fibronectin, and stimulated migration and invasion of human and mouse tumor cells. Intravenously injected S100A4 protein induced expression of SAA proteins and cytokines in an organ-specific manner. In a breast cancer animal model, ectopic expression of SAA1 or SAA3 in tumor cells potently promoted widespread metastasis formation accompanied by a massive infiltration of immune cells. Furthermore, coordinate expression of S100A4 and SAA in tumor samples from colorectal carcinoma patients significantly correlated with reduced overall survival. These data show that SAA proteins are effectors for the metastasis-promoting functions of S100A4, and serve as a link between inflammation and tumor progression.


Subject(s)
Inflammation/complications , Neoplasm Metastasis , S100 Proteins/physiology , Serum Amyloid A Protein/genetics , Animals , Cell Line, Tumor , Colonic Neoplasms/mortality , ErbB Receptors/physiology , Humans , Mice , Organ Specificity , S100 Calcium-Binding Protein A4 , Serum Amyloid A Protein/physiology
19.
Proc Natl Acad Sci U S A ; 111(14): 5189-94, 2014 Apr 08.
Article in English | MEDLINE | ID: mdl-24706838

ABSTRACT

Serum amyloid A (SAA) represents an evolutionarily conserved family of inflammatory acute-phase proteins. It is also a major constituent of secondary amyloidosis. To understand its function and structural transition to amyloid, we determined a structure of human SAA1.1 in two crystal forms, representing a prototypic member of the family. Native SAA1.1 exists as a hexamer, with subunits displaying a unique four-helix bundle fold stabilized by its long C-terminal tail. Structure-based mutational studies revealed two positive-charge clusters, near the center and apex of the hexamer, that are involved in SAA association with heparin. The binding of high-density lipoprotein involves only the apex region of SAA and can be inhibited by heparin. Peptide amyloid formation assays identified the N-terminal helices 1 and 3 as amyloidogenic peptides of SAA1.1. Both peptides are secluded in the hexameric structure of SAA1.1, suggesting that the native SAA is nonpathogenic. Furthermore, dissociation of the SAA hexamer appears insufficient to initiate amyloidogenic transition, and proteolytic cleavage or removal of the C-terminal tail of SAA resulted in formation of various-sized structural aggregates containing ∼5-nm regular repeating protofibril-like units. The combined structural and functional studies provide mechanistic insights into the pathogenic contribution of glycosaminoglycan in SAA1.1-mediated AA amyloid formation.


Subject(s)
Amyloidosis/physiopathology , Inflammation/physiopathology , Serum Amyloid A Protein/physiology , Amino Acid Sequence , Binding Sites , Glycosaminoglycans/metabolism , Humans , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein Folding , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Sequence Homology, Amino Acid , Serum Amyloid A Protein/chemistry , Serum Amyloid A Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...