Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 134
Filtrar
1.
J Allergy Clin Immunol ; 153(4): 1010-1024.e14, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38092139

RESUMEN

RATIONALE: Serum amyloid A (SAA) is bound to high-density lipoproteins (HDL) in blood. Although SAA is increased in the blood of patients with asthma, it is not known whether this modifies asthma severity. OBJECTIVE: We sought to define the clinical characteristics of patients with asthma who have high SAA levels and assess whether HDL from SAA-high patients with asthma is proinflammatory. METHODS: SAA levels in serum from subjects with and without asthma were quantified by ELISA. HDLs isolated from subjects with asthma and high SAA levels were used to stimulate human monocytes and were intravenously administered to BALB/c mice. RESULTS: An SAA level greater than or equal to 108.8 µg/mL was defined as the threshold to identify 11% of an asthmatic cohort (n = 146) as being SAA-high. SAA-high patients with asthma were characterized by increased serum C-reactive protein, IL-6, and TNF-α; older age; and an increased prevalence of obesity and severe asthma. HDL isolated from SAA-high patients with asthma (SAA-high HDL) had an increased content of SAA as compared with HDL from SAA-low patients with asthma and induced the secretion of IL-6, IL-1ß, and TNF-α from human monocytes via a formyl peptide receptor 2/ATP/P2X purinoceptor 7 axis. Intravenous administration to mice of SAA-high HDL, but not normal HDL, induced systemic inflammation and amplified allergen-induced neutrophilic airway inflammation and goblet cell metaplasia. CONCLUSIONS: SAA-high patients with asthma are characterized by systemic inflammation, older age, and an increased prevalence of obesity and severe asthma. HDL from SAA-high patients with asthma is proinflammatory and, when intravenously administered to mice, induces systemic inflammation, and amplifies allergen-induced neutrophilic airway inflammation. This suggests that systemic inflammation induced by SAA-high HDL may augment disease severity in asthma.


Asunto(s)
Asma , Lipoproteínas HDL , Humanos , Animales , Ratones , Lipoproteínas HDL/metabolismo , Lipoproteínas HDL/farmacología , Proteína Amiloide A Sérica/análisis , Proteína Amiloide A Sérica/metabolismo , Proteína Amiloide A Sérica/farmacología , Factor de Necrosis Tumoral alfa/metabolismo , Interleucina-6 , Inflamación/metabolismo , Obesidad , Alérgenos
2.
Front Endocrinol (Lausanne) ; 14: 1119227, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36817589

RESUMEN

Originally, it was thought that a single serum amyloid A (SAA) protein was involved in amyloid A amyloidosis, but in fact, SAA represents a four-membered family wherein SAA1 and SAA2 are acute phase proteins (A-SAA). SAA is highly conserved throughout evolution within a wide range of animal species suggestive of an important biological function. In fact, A-SAA has been linked to a number of divergent biological activities wherein a number of these functions are mediated via the G protein-coupled receptor (GPCR), formyl peptide receptor (FPR) 2. For instance, through the activation of FPR2, A-SAA has been described to regulate leukocyte activation, atherosclerosis, pathogen recognition, bone formation and cell survival. Moreover, A-SAA is subject to post-translational modification, primarily through proteolytic processing, generating a range of A-SAA-derived peptides. Although very little is known regarding the biological effect of A-SAA-derived peptides, they have been shown to promote neutrophil and monocyte migration through FPR2 activation via synergy with other GPCR ligands namely, the chemokines CXCL8 and CCL3, respectively. Within this review, we provide a detailed analysis of the FPR2-mediated functions of A-SAA. Moreover, we discuss the potential role of A-SAA-derived peptides as allosteric modulators of FPR2.


Asunto(s)
Receptores de Formil Péptido , Proteína Amiloide A Sérica , Animales , Receptores de Formil Péptido/fisiología , Ligandos , Proteína Amiloide A Sérica/metabolismo , Proteína Amiloide A Sérica/farmacología , Transducción de Señal , Péptidos/metabolismo
3.
Mil Med ; 188(1-2): e133-e139, 2023 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-33959775

RESUMEN

BACKGROUND: Nanofiber wound dressings remain the domain of in vitro studies. The purpose of our study was to verify the benefits of chitosan (CTS) and polylactide (PLA)-based nanofiber wound dressings on a porcine model of a naturally contaminated standardized wound and compare them with the conventional dressings, i.e., gauze and Inadine. MATERIAL AND METHODS: The study group included 32 pigs randomized into four homogeneous groups according to the wound dressing type. Standardized wounds were created on their backs, and wound dressings were regularly changed. We evaluated difficulty of handling individual dressing materials and macroscopic appearance of the wounds. Wound swabs were taken for bacteriological examination. Blood samples were obtained to determine blood count values and serum levels of acute phase proteins (serum amyloid A, C-reactive protein, and haptoglobin). The crucial point of the study was histological analysis. Microscopic evaluation was focused on the defect depth and tissue reactions, including formation of the fibrin exudate with neutrophil granulocytes, the layer of granulation and cellular connective tissue, and the reepithelialization. Statistical analysis was performed by using SPSS software. The analysis was based on the Kruskal-Wallis H test and Mann-Whitney U test followed by Bonferroni correction. Significance was set at P < .05. RESULTS: Macroscopic examination did not show any difference in wound healing among the groups. However, evaluation of histological findings demonstrated that PLA-based nanofiber dressing accelerated the proliferative (P = .025) and reepithelialization (P < .001) healing phases, while chitosan-based nanofiber dressing potentiated and accelerated the inflammatory phase (P = .006). No statistically significant changes were observed in the blood count or acute inflammatory phase proteins during the trial. Different dynamics were noted in serum amyloid A values in the group treated with PLA-based nanofiber dressing (P = .006). CONCLUSION: Based on the microscopic examination, we have documented a positive effect of nanofiber wound dressings on acceleration of individual phases of the healing process. Nanofiber wound dressings have a potential to become in future part of the common wound care practice.


Asunto(s)
Quitosano , Nanofibras , Animales , Porcinos , Nanofibras/uso terapéutico , Quitosano/farmacología , Quitosano/uso terapéutico , Proteína Amiloide A Sérica/farmacología , Cicatrización de Heridas , Vendajes
4.
Reprod Biol Endocrinol ; 20(1): 4, 2022 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-34980155

RESUMEN

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.


Asunto(s)
Células de la Granulosa/metabolismo , Resistencia a la Insulina/genética , Síndrome del Ovario Poliquístico/genética , Proteína Amiloide A Sérica/fisiología , Adulto , Estudios de Casos y Controles , Células Cultivadas , Femenino , Líquido Folicular/química , Líquido Folicular/metabolismo , Células de la Granulosa/efectos de los fármacos , Humanos , Ovario/efectos de los fármacos , Ovario/metabolismo , Síndrome del Ovario Poliquístico/sangre , Síndrome del Ovario Poliquístico/metabolismo , Proteína Amiloide A Sérica/genética , Proteína Amiloide A Sérica/metabolismo , Proteína Amiloide A Sérica/farmacología
5.
Front Immunol ; 12: 691155, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34276683

RESUMEN

AA amyloidosis belongs to the group of amyloid diseases which can follow chronic inflammatory conditions of various origin. The disease is characterized by the deposition of insoluble amyloid fibrils formed by serum amyloid A1 (SAA1) leading eventually to organ failure. Macrophages are intimately involved in the fibrillogenesis as well as in the clearance of amyloid fibrils. In vivo, macrophages may occur as classically (M1) or alternatively activated (M2) macrophages. We investigate here how SAA1 might affect the macrophage phenotype and function. Gene microarray analysis revealed upregulation of 64 M1-associated genes by SAA1. M1-like polarization was further confirmed by the expression of the M1-marker MARCO, activation of the NF-κB transcription factor, and secretion of the M1-cytokines TNF-α, IL-6, and MCP-1. Additionally, we demonstrate here that M1-polarized macrophages exhibit enhanced fibrillogenic activity towards SAA1. Based on our data, we propose reconsideration of the currently used cellular amyloidosis models towards an in vitro model employing M1-polarized macrophages. Furthermore, the data suggest macrophage repolarization as potential intervention strategy in AA amyloidosis.


Asunto(s)
Macrófagos/efectos de los fármacos , Proteína Amiloide A Sérica/farmacología , Animales , Células Cultivadas , Citocinas/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Activación de Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Proteínas Recombinantes/farmacología , Proteína Amiloide A Sérica/genética
6.
Biochem Biophys Res Commun ; 555: 54-60, 2021 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-33813276

RESUMEN

Non-alcoholic fatty liver disease (NAFLD) is the pathological manifestation of metabolic syndrome in liver. Its pathological changes may evolve from the initial simple steatosis to non-alcoholic steatohepatitis, liver fibrosis and even liver cancer. Numerous studies have proved that platelets play a vital role in liver disease and homeostasis. Particularly, anti-platelet therapy can reduce intrahepatic platelet aggregation and improve the inflammation of fatty liver. Previous study has also confirmed that SAA is a gene closely related to high-fat diet (HFD) induced obesity, and SAA1 can promote liver insulin resistance induced by Palmitate or HFD. Here, we found that SAA1 treated platelets presented increased sensitivity of platelet aggregation, enhanced activation and increased adhesion ability, and such function was partly dependent on Toll-Like Receptor (TLR) 2 signaling. In addition, blocking SAA1 expression in vivo not only inhibited platelet aggregation in the liver tissues of NAFLD mice, but also alleviated the inflammation of fatty liver. In conclusion, our findings identify that HFD-induced hepatic overexpressed SAA1 aggravates fatty liver inflammation by promoting intrahepatic platelet aggregation, these results also imply that SAA1 may serve as a potential target for ameliorating NAFLD.


Asunto(s)
Hepatocitos/metabolismo , Enfermedad del Hígado Graso no Alcohólico/fisiopatología , Agregación Plaquetaria/efectos de los fármacos , Proteína Amiloide A Sérica/metabolismo , Animales , Adhesión Celular/efectos de los fármacos , Dieta Alta en Grasa/efectos adversos , Femenino , Hepatitis/etiología , Hepatocitos/patología , Humanos , Ratones Endogámicos BALB C , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Proteína Amiloide A Sérica/genética , Proteína Amiloide A Sérica/farmacología , Receptor Toll-Like 2/metabolismo
7.
Neurosci Lett ; 738: 135352, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32931862

RESUMEN

The blood-brain barrier (BBB) is the multicellular interface located between the peripheral circulation and the brain parenchyma. BBB dysfunction is reported in many CNS diseases, such cognitive impairment, depression, Alzheimer's disease (AD), and multiple sclerosis (MS). Emerging evidence indicates that liver-derived inflammatory mediators are upregulated in neurological diseases with BBB dysfunction. Serum amyloid A (SAA), an acute phase protein secreted by hepatocytes, could be a candidate inflammatory signaling molecule transmitted from the liver to the brain; however, its contribution to BBB dysfunction is poorly understood. The present study aimed to elucidate the involvement of SAA in BBB impairment in an in vitro BBB model using rat brain microvascular endothelial cells (RBECs). We demonstrated that Apo-SAA significantly decreased transendothelial electrical resistance (TEER) and increased sodium fluorescein (Na-F) permeability in RBEC monolayers. Apo-SAA also decreased claudin-5 expression levels in RBECs. Furthermore, the Apo-SAA-mediated impairment of the BBB with decreased claudin-5 expression was inhibited by the addition of a high-density lipoprotein (HDL) related to SAA in plasma. These findings suggest that HDL counteracts the effects of SAA on BBB function. Therefore, the functional imbalance between SAA and HDL may induce BBB impairment, thereby triggering development of neuroinflammation. SAA could be a significant endogenous mediator in the liver-to-brain inflammation axis.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Encéfalo/efectos de los fármacos , Claudina-5/metabolismo , Células Endoteliales/efectos de los fármacos , Lipoproteínas HDL/farmacología , Proteína Amiloide A Sérica/farmacología , Animales , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Permeabilidad Capilar/efectos de los fármacos , Células Endoteliales/metabolismo , Ratas , Ratas Wistar
8.
J Neuroinflammation ; 17(1): 254, 2020 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-32861245

RESUMEN

BACKGROUND: The accumulation of astrocytes around senile plaques is one of the pathological characteristics in Alzheimer's disease (AD). Serum amyloid A (SAA), known as a major acute-phase protein, colocalizes with senile plaques in AD patients. Here, we demonstrate the role of SAA in astrocyte migration. METHODS: The effects of SAA on astrocyte activation and accumulation around amyloid ß (Aß) deposits were detected in APP/PS1 transgenic mice mated with Saa3-/- mice. SAA expression, astrocyte activation, and colocalization with Aß deposits were evaluated in mice using immunofluorescence staining and/or Western blotting. The migration of primary cultures of mouse astrocytes and human glioma U251 cells was examined using Boyden chamber assay and scratch-would assay. The actin and microtubule networks, protrusion formation, and Golgi apparatus location in astrocytes were determined using scratch-would assay and immunofluorescence staining. RESULTS: Saa3 expression was significantly induced in aged APP/PS1 transgenic mouse brain. Saa3 deficiency exacerbated astrocyte activation and increased the number of astrocytes around Aß deposits in APP/PS1 mice. In vitro studies demonstrated that SAA inhibited the migration of primary cultures of astrocytes and U251 cells. Mechanistic studies showed that SAA inhibited astrocyte polarization and protrusion formation via disrupting actin and microtubule reorganization and Golgi reorientation. Inhibition of the p38 MAPK pathway abolished the suppression of SAA on astrocyte migration and polarization. CONCLUSIONS: These results suggest that increased SAA in the brain of APP/PS1 mice inhibits the migration of astrocytes to amyloid plaques by activating the p38 MAPK pathway.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Astrocitos/efectos de los fármacos , Encéfalo/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proteína Amiloide A Sérica/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Astrocitos/metabolismo , Encéfalo/metabolismo , Línea Celular Tumoral , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Transgénicos , Presenilina-1/genética , Presenilina-1/metabolismo
9.
Front Immunol ; 11: 843, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32477346

RESUMEN

Infection, sterile injury, and chronic inflammation trigger the acute phase response in order to re-establish homeostasis. This response includes production of positive acute phase proteins in the liver, such as members of the serum amyloid A (SAA) family. In humans the major acute phase SAAs comprise a group of closely related variants of SAA1 and SAA2. SAA1 was proven to be chemotactic for several leukocyte subtypes through activation of the G protein-coupled receptor FPRL1/FPR2. Several other biological activities of SAA1, such as cytokine induction, reported to be mediated via TLRs, have been debated recently. Especially commercial SAA1, recombinantly produced in Escherichia coli, was found to be contaminated with bacterial products confounding biological assays performed with this rSAA1. We purified rSAA1 by RP-HPLC to homogeneity, removing contaminants such as lipopolysaccharides, lipoproteins and formylated peptides, and re-assessed several biological activities attributed to SAA1 (chemotaxis, cytokine induction, MMP-9 release, ROS generation, and macrophage differentiation). The homogeneous rSAA1 (hrSAA1) lacked most cell-activating properties, but its leukocyte-recruiting capacity in vivo and it's in vitro synergy with other leukocyte attractants remained preserved. Furthermore, hrSAA1 maintained the ability to promote monocyte survival. This indicates that pure hrSAA1 retains its potential to activate FPR2, whereas TLR-mediated effects seem to be related to traces of bacterial TLR ligands in the E. coli-produced human rSAA1.


Asunto(s)
Leucocitos/efectos de los fármacos , Leucocitos/inmunología , Proteína Amiloide A Sérica/farmacología , Donantes de Sangre , Diferenciación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Quimiotaxis/efectos de los fármacos , Citocinas/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Células HEK293 , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptores de Formil Péptido/genética , Receptores de Lipoxina/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/farmacología , Proteína Amiloide A Sérica/genética , Proteína Amiloide A Sérica/aislamiento & purificación , Transducción de Señal/efectos de los fármacos , Receptores Toll-Like/metabolismo , Transfección
10.
Lipids ; 55(6): 615-626, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32558932

RESUMEN

Cellular lipid metabolism, lipoprotein interactions, and liver X receptor (LXR) activation have been implicated in the pathophysiology and treatment of cancer, although findings vary across cancer models and by lipoprotein profiles. In this study, we investigated the effects of human-derived low-density lipoproteins (LDL), high-density lipoproteins (HDL), and HDL-associated proteins apolipoprotein A1 (apoA1) and serum amyloid A (SAA) on markers of viability, cholesterol flux, and differentiation in K562 cells-a bone marrow-derived, stem-like erythroleukemia cell model of chronic myelogenous leukemia (CML). We further evaluated whether lipoprotein-mediated effects were altered by concomitant LXR activation. We observed that LDL promoted higher K562 cell viability in a dose- and time-dependent manner and increased cellular cholesterol concentrations, while LXR activation by the agonist TO901317 ablated these effects. LXR activation in the presence of HDL, apoA1 and SAA-rich HDL suppressed K562 cell viability, while robustly inducing mRNA expression of ATP-binding cassette transporter A1 (ABCA1). HDL and its associated proteins additionally suppressed mRNA expression of anti-apoptotic B-cell lymphoma-extra large (BCL-xL), and the erythroid lineage marker 5'-aminolevulinate synthase 2 (ALAS2), while SAA-rich HDL induced mRNA expression of the megakaryocytic lineage marker integrin subunit alpha 2b (ITGA2B). Together, these findings suggest that lipoproteins and LXR may impact the viability and characteristics of CML cells.


Asunto(s)
Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Lipoproteínas HDL/farmacología , Lipoproteínas LDL/farmacología , Transportador 1 de Casete de Unión a ATP/genética , Transportador 1 de Casete de Unión a ATP/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1/genética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1/metabolismo , Apolipoproteína A-I/metabolismo , Apolipoproteína A-I/farmacología , Diferenciación Celular/genética , Supervivencia Celular/efectos de los fármacos , Colesterol/metabolismo , Relación Dosis-Respuesta a Droga , Regulación Leucémica de la Expresión Génica , Humanos , Células K562 , Leucemia Mielógena Crónica BCR-ABL Positiva/sangre , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Lipoproteínas HDL/administración & dosificación , Lipoproteínas HDL/metabolismo , Lipoproteínas LDL/administración & dosificación , Lipoproteínas LDL/metabolismo , Receptores X del Hígado/metabolismo , Proteína Amiloide A Sérica/metabolismo , Proteína Amiloide A Sérica/farmacología , Factores de Tiempo , Proteína bcl-X/genética
11.
J Biol Chem ; 295(9): 2570-2581, 2020 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-31819008

RESUMEN

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.


Asunto(s)
Antibacterianos/metabolismo , Inmunidad Innata , Proteína Amiloide A Sérica/metabolismo , Infecciones Estafilocócicas/inmunología , Infecciones Cutáneas Estafilocócicas/inmunología , Proteínas de Fase Aguda/inmunología , Proteínas de Fase Aguda/metabolismo , Animales , Antibacterianos/farmacología , Adhesión Bacteriana , Membrana Celular/efectos de los fármacos , Concentración de Iones de Hidrógeno , Interleucina-6/fisiología , Ratones , Proteína Amiloide A Sérica/inmunología , Proteína Amiloide A Sérica/farmacología , Staphylococcus aureus/citología , Staphylococcus aureus/ultraestructura
12.
Arthritis Res Ther ; 21(1): 250, 2019 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-31775905

RESUMEN

BACKGROUND: Hydroxychloroquine (HCQ) is used for the treatment of patients with rheumatic diseases. We tested the hypothesis that HCQ affects the NLRP3 inflammasome, which is involved in autoinflammation. METHODS: Human neutrophils were stimulated with serum amyloid A (SAA) in vitro and measured for IL-1ß and caspase-1 (p20) secretion by ELISA. Pro-IL-1ß mRNA expression in human neutrophils was quantified by real-time RT-PCR. RESULTS: SAA stimulation induced significant production of IL-1ß in human neutrophils. SAA stimulation also induced NF-κB activation, pro-IL-1ß mRNA expression, and NLRP3 protein expression in human neutrophils. HCQ pretreatment significantly inhibited the SAA-induced IL-1ß production in human neutrophils, but did not affect the SAA-induced NF-κB activation, pro-IL-1ß mRNA expression, and NLRP3 protein expression. Furthermore, SAA stimulation induced cleaved caspase-1 (p20) secretion from human neutrophils, and this release was suppressed by HCQ pretreatment. CONCLUSIONS: Treatment with HCQ was associated with impaired production of IL-1ß in SAA-stimulated human neutrophils without affecting the priming process of the NLRP3 inflammasome such as pro-IL-1ß or NLRP3 induction. These findings suggest that HCQ affects the NLRP3 activation process, resulting in the impaired IL-1ß production in human neutrophils, as representative innate immune cells.


Asunto(s)
Hidroxicloroquina/farmacología , Interleucina-1beta/metabolismo , Neutrófilos/efectos de los fármacos , Proteína Amiloide A Sérica/farmacología , Adulto , Animales , Caspasa 1/metabolismo , Células Cultivadas , Relación Dosis-Respuesta a Droga , Femenino , Expresión Génica/efectos de los fármacos , Humanos , Inflamasomas/metabolismo , Interleucina-1beta/genética , Masculino , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Neutrófilos/citología , Neutrófilos/metabolismo
13.
Atherosclerosis ; 291: 1-8, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31629987

RESUMEN

BACKGROUND AND AIMS: Patients with rheumatic diseases have an increased risk of atherosclerosis with up-regulated serum amyloid A (SAA), tumor necrosis factor-α (TNF-α) and interleukin-1ß (IL-1ß), which were reported to activate human coronary artery endothelial cells (HCAEC). We aimed to investigate the effects of TNF-α inhibitor infliximab and anti-infliximab antibodies on the TNF-α/IL-1ß/SAA activated HCAEC. METHODS: HCAEC were incubated with TNF-α, IL-1ß, SAA, infliximab, anti-infliximab antibodies and their combinations. The protein levels of pro- and anti-atherogenic analytes were measured in supernatants using ELISA and multiplex assays, while mRNA expression was determined by RT-PCR. Anti-infliximab antibodies were purified from sera samples by affinity chromatography. RESULTS: IL-6, IL-8, GM-CSF and GRO-α were synergistically up-regulated in triple stimulation with TNF-α, IL-1ß and SAA, while their levels in solely SAA- or TNF-α-stimulated HCAEC did not increase. IL-1Ra, IL-1α, VCAM-1, MCP-1, IL-10 and IL-17A were increased, but no synergistic responses were observed in triple stimulation. Infliximab was effective in lowering the synergistic effect of IL-6, IL-8, GM-CSF and GRO-α in triple stimulation, while anti-infliximab antibodies restored the levels. The changes were confirmed at the mRNA expression level for IL-6, IL-8 and GM-CSF. CONCLUSIONS: Triple stimulation with TNF-α, IL-1ß and SAA synergistically elevated IL-6, IL-8, GM-CSF and GRO-α release in supernatants of HCAEC, with infliximab substantially inhibiting their levels. An isolated, enriched fraction of polyclonal anti-infliximab antibodies was capable of neutralizing infliximab, in the presence of TNF-α/IL-1ß/SAA. The long-term presence of anti-infliximab antibodies in the circulation of patients with chronic rheumatic diseases is potentially important for promoting the atherosclerotic process.


Asunto(s)
Anticuerpos Neutralizantes/metabolismo , Vasos Coronarios/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Infliximab/inmunología , Inhibidores del Factor de Necrosis Tumoral/inmunología , Anticuerpos Neutralizantes/sangre , Células Cultivadas , Vasos Coronarios/inmunología , Vasos Coronarios/metabolismo , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Humanos , Mediadores de Inflamación/metabolismo , Interleucina-1beta/farmacología , Proteína Amiloide A Sérica/farmacología , Factor de Necrosis Tumoral alfa/farmacología
14.
BMC Mol Cell Biol ; 20(1): 15, 2019 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-31216990

RESUMEN

BACKGROUND: To investigate the effects of serum amyloid A1 (SAA1) on lipopolysaccharide (LPS) -induced inflammation in vascular smooth muscle cells (VSMCs). SAA1 expression was detected in LPS induced VSMCs at different concentrations for different time by using Western blotting. After pre-incubation with recombinant SAA1 protein, VSMCs were treated with 1 µg/ml LPS for 24 h. The VSMCs were then divided into Control, SAA1 siRNA, Nox4 siRNA, LPS, LPS + SAA1 siRNA, LPS + Nox4 siRNA and LPS + SAA1 siRNA + Nox4 groups. MTT was performed to observe the toxicity of VSMCs. Lucigenin-enhanced chemiluminescence method was used to detect superoxide anion (O2-) production and NADPH oxidase activity. Quantitative real-time polymerase chain reaction (qRT-PCR) was used to determine expressions of inflammatory factors. Western blotting was used to determine expressions of NOX-4 and p38MAPK/NF-κB pathway related proteins. RESULTS: LPS promoted SAA1 protein expression in a concentration-/time-dependent manner. Recombinant SAA1 protein could increase NOX4/ROS production and promote the release of inflammatory factors (IL-1ß, IL-6, IL-8, IL-17, TNF-α and MCP-1) in LPS (1 µg/ml) - induced VSMCs. Besides, both SAA1 siRNA and NOX-4 siRNA could not only enhance the O2- production and NADPH oxidase activity, but also up-regulate the protein expression of NOX4, the release of inflammatory factors, and the levels of p-p38 and p-NF-κB p65 in LPS-induced VSMCs. However, no significant differences in each index were observed between LPS group and LPS + SAA1 siRNA + Nox4 group. CONCLUSION: SAA1-mediated NOX4/ROS pathway could activate p38MAPK/NF-κB pathway, thereby contributing to the release of inflammatory factors in LPS-induced VSMCs.


Asunto(s)
Músculo Liso Vascular/citología , Miocitos del Músculo Liso/metabolismo , NADPH Oxidasa 4/metabolismo , FN-kappa B/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteína Amiloide A Sérica/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Citocinas/metabolismo , Inflamación/inducido químicamente , Lipopolisacáridos/administración & dosificación , Lipopolisacáridos/farmacología , Masculino , NADPH Oxidasa 4/genética , NADPH Oxidasas/metabolismo , ARN Interferente Pequeño/genética , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacología , Proteína Amiloide A Sérica/administración & dosificación , Proteína Amiloide A Sérica/genética , Proteína Amiloide A Sérica/farmacología , Transfección
15.
Am J Reprod Immunol ; 82(2): e13150, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31131948

RESUMEN

PROBLEM: Cortisol, which is regenerated from biologically inactive cortisone by 11ß-hydroxysteroid dehydrogenase type 1 (11ß-HSD1) in human fetal membranes, may play an important role in human parturition. Recently, we have demonstrated that human fetal membranes are capable of de novo synthesis of serum amyloid A1 (SAA1), an acute-phase protein of inflammation, and SAA1 may be engaged in multiple actions associated with human parturition. It remains to be determined whether SAA1 can interact with cortisol in the regulation of 11ß-HSD1 in the fetal membranes. METHOD OF STUDY: In the current study, we examined the regulation of 11ß-HSD1 expression by SAA1, and the interaction between SAA1 and cortisol in the regulation of 11ß-HSD1 expression in primary human amnion fibroblasts and amnion tissue. RESULTS: Either SAA1 or cortisol induced 11ß-HSD1 expression in a concentration-dependent manner. Combination of SAA1 and cortisol synergistically enhanced 11ß-HSD1 expression. Mechanism studies revealed that SAA1 and cortisol induced the phosphorylation of the transcription factor STAT3 in a sequential order with the induction by SAA1 preceding the induction by cortisol. Furthermore, the induction of 11ß-HSD1 expression by either SAA1 or cortisol or combination of SAA1 and cortisol was blocked by STAT3 inhibition with its antagonist S3I-201 or siRNA-mediated knockdown. CONCLUSION: This study has demonstrated that SAA1 and cortisol can reinforce each other in the induction of 11ß-HSD1 expression through sequential phosphorylation of STAT3. The synergistic enhancement of 11ß-HSD1 expression by SAA1 and cortisol may lead to excessive cortisol accumulation in the fetal membranes at parturition.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/metabolismo , Amnios/metabolismo , Fibroblastos/metabolismo , Hidrocortisona/farmacología , Parto/metabolismo , Factor de Transcripción STAT3/metabolismo , Proteína Amiloide A Sérica/farmacología , Amnios/efectos de los fármacos , Células Cultivadas , Sinergismo Farmacológico , Membranas Extraembrionarias/metabolismo , Femenino , Fibroblastos/efectos de los fármacos , Humanos , Parto/fisiología , Fosforilación , Embarazo , ARN Interferente Pequeño , Factor de Transcripción STAT3/antagonistas & inhibidores , Factor de Transcripción STAT3/química , Factor de Transcripción STAT3/genética , Proteína Amiloide A Sérica/genética
16.
J Neuroinflammation ; 15(1): 164, 2018 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-29803222

RESUMEN

BACKGROUND: Acute-phase response is a systemic reaction to environmental/inflammatory insults and involves production of acute-phase proteins, including serum amyloid A (SAA). Interleukin-1ß (IL-1ß), a master regulator of neuroinflammation produced by activated inflammatory cells of the myeloid lineage, in particular microglia, plays a key role in the pathogenesis of acute and chronic diseases of the peripheral nervous system and CNS. IL-1ß release is promoted by ATP acting at the purinergic P2X7 receptor (P2X7R) in cells primed with toll-like receptor (TLR) ligands. METHODS: Purified (> 99%) microglia cultured from neonatal rat cortex and cerebellum were first primed with the putative TLR4/TLR2 agonist SAA (recombinant human Apo-SAA) or the established TLR4 agonist lipopolysaccharide (LPS) followed by addition of ATP. Expression of genes for the NLRP3 inflammasome, IL-1ß, tumor necrosis factor-α (TNF-α), and SAA1 was measured by quantitative real-time polymerase chain reaction (q-PCR). Intracellular and extracellular amounts of IL-1ß were determined by ELISA. RESULTS: Apo-SAA stimulated, in a time-dependent manner, the expression of NLRP3, IL-1ß, and TNF-α in cortical microglia, and produced a concentration-dependent increase in the intracellular content of IL-1ß in these cells. A 2-h 'priming' of the microglia with Apo-SAA followed by addition of ATP for 1 h, resulting in a robust release of IL-1ß into the culture medium, with a concomitant reduction in its intracellular content. The selective P2X7R antagonist A740003 blocked ATP-dependent release of IL-1ß. Microglia prepared from rat cerebellum displayed similar behaviors. As with LPS, Apo-SAA upregulated SAA1 and TLR2 mRNA, and downregulated that of TLR4. LPS was less efficacious than Apo-SAA, perhaps reflecting an action of the latter at TLR4 and TLR2. The TLR4 antagonist CLI-095 fully blocked the action of LPS, but only partially that of Apo-SAA. Although the TLR2 antagonist CU-CPT22 was inactive against Apo-SAA, it also failed to block the TLR2 agonist Pam3CSK4. CONCLUSIONS: Microglia are central to the inflammatory process and a major source of IL-1ß when activated. P2X7R-triggered IL-1ß maturation and export is thus likely to represent an important contributor to this cytokine pool. Given that SAA is detected in Alzheimer disease and multiple sclerosis brain, together with IL-1ß-immunopositive microglia, these findings propose a link between P2X7R, SAA, and IL-1ß in CNS pathophysiology.


Asunto(s)
Adenosina Trifosfato/farmacología , Interleucina-1beta/metabolismo , Microglía/efectos de los fármacos , Proteína Amiloide A Sérica/farmacología , Animales , Animales Recién Nacidos , Encéfalo/citología , Células Cultivadas , Interleucina-1beta/genética , Lipopolisacáridos/farmacología , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , ARN Mensajero/metabolismo , Ratas , Factores de Tiempo
17.
Mediators Inflamm ; 2018: 8237209, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29670468

RESUMEN

BACKGROUND: RA patients have a higher incidence of cardiovascular diseases compared to the general population. Serum amyloid A (SAA) is an acute-phase protein, upregulated in sera of RA patients. AIM: To determine the effects of medications on SAA-stimulated human coronary artery endothelial cells (HCAEC). METHODS: HCAEC were preincubated for 2 h with medications from sterile ampules (dexamethasone, methotrexate, certolizumab pegol, and etanercept), dissolved in medium (captopril) or DMSO (etoricoxib, rosiglitazone, meloxicam, fluvastatin, and diclofenac). Human recombinant apo-SAA was used to stimulate HCAEC at a final 1000 nM concentration for 24 hours. IL-6, IL-8, sVCAM-1, and PAI-1 were measured by ELISA. The number of viable cells was determined colorimetrically. RESULTS: SAA-stimulated levels of released IL-6, IL-8, and sVCAM-1 from HCAEC were significantly attenuated by methotrexate, fluvastatin, and etoricoxib. Both certolizumab pegol and etanercept significantly decreased PAI-1 by an average of 43%. Rosiglitazone significantly inhibited sVCAM-1 by 58%. CONCLUSION: We observed marked influence of fluvastatin on lowering cytokine production in SAA-activated HCAEC. Methotrexate showed strong beneficial effects for lowering released Il-6, IL-8, and sVCAM-1. Interesting duality was observed for NSAIDs, with meloxicam exhibiting opposite-trend effects from diclofenac and etoricoxib. This represents unique insight into specific responsiveness of inflammatory-driven HCAEC relevant to atherosclerosis.


Asunto(s)
Vasos Coronarios/citología , Células Endoteliales/metabolismo , Proteína Amiloide A Sérica/farmacología , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Humanos , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Inhibidor 1 de Activador Plasminogénico/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo
18.
Blood ; 131(4): 439-449, 2018 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-29371208

RESUMEN

A natural leukocyte chemoattractant was isolated from bovine serum by an established 4-step purification procedure. Based on its relative molecular mass of 7287 and NH2-terminal sequence, the protein was identified as a carboxy-terminal peptide of the acute phase protein serum amyloid A1 (SAA1). This SAA1(46-112) fragment and its human equivalent SAA1(47-104) were chemically synthesized. Unlike intact SAA1α, these SAA fragments failed to directly chemoattract neutrophils and monocytes, to induce chemokines, and to stimulate downstream extracellular signal-regulated kinase signaling in monocytes. However, the SAA fragments potently synergized with CCL3 to induce monocyte migration and with CXCL8 to stimulate neutrophil shape changes and chemotaxis. Unlike intact SAA1α, SAA1(46-112) did not induce CXCL6 ex vivo but provoked a cooperative intraperitoneal neutrophil recruitment in mice when coinjected with CXCL6 into the peritoneal cavity. Moreover, SAA1(47-104) desensitized the synergy between intact SAA1α and CXCL8 in neutrophil chemotaxis, suggesting that this peptide binds formyl peptide receptor 2 (FPR2). This was evidenced by a complete blockade of synergy between the COOH-terminal SAA1 fragments and CXCL8 or CCL3 in neutrophil and monocyte chemotaxis, respectively, by the FPR2 antagonist WRW4 Thus, SAA1 is degraded into fragments lacking chemokine-inducing capacity, while keeping synergy with cytokine-induced chemokines to sustain limited inflammation.


Asunto(s)
Quimiocina CCL3/inmunología , Quimiocinas/inmunología , Interleucina-8/inmunología , Leucocitos/efectos de los fármacos , Receptores de Formil Péptido/inmunología , Receptores de Lipoxina/inmunología , Proteína Amiloide A Sérica/química , Proteína Amiloide A Sérica/farmacología , Animales , Bovinos , Quimiotaxis/efectos de los fármacos , Femenino , Humanos , Leucocitos/inmunología , Ratones , Péptidos/síntesis química , Péptidos/química , Péptidos/farmacología , Proteína Amiloide A Sérica/síntesis química
19.
J Immunol ; 199(3): 1105-1112, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28674180

RESUMEN

Serum amyloid A (SAA) is known as an acute-phase protein and a biomarker for inflammatory diseases. Published studies have shown that SAA possesses proinflammatory cytokine-like activity and is chemotactic for phagocytes, but the structural basis for these activities remains unidentified. In this article, we report that truncated SAA1 proteins lacking N- and C-terminal sequences exhibit reduced proinflammatory activity and strongly suppress LPS-induced expression of IL-1ß, IL-6, and TNF-α in macrophages. A truncated SAA1 containing aa 11-58 was examined further and found to facilitate p38 MAPK phosphorylation while reducing LPS-stimulated phosphorylation of ERK and JNK. In LPS-challenged mice, aa 11-58 reduced the severity of acute lung injury, with significantly less neutrophil infiltration in the lungs and attenuated pulmonary expression of IL-1ß, IL-6, and TNF-α. Coadministration of aa 11-58 markedly improved mouse survival in response to a lethal dose of LPS. A potent induction of IL-10 was observed in a TLR2-dependent, but TLR4-independent, manner in macrophages stimulated with aa 11-58. However, the aa 11-58 fragment of SAA1 was unable to induce chemotaxis or calcium flux through formyl peptide receptor 2. These results indicate that the N- and C-terminal sequences contain structural determinants for the proinflammatory and chemotactic activities of SAA1, and their removal switches SAA1 to an anti-inflammatory role. Given that proteolytic processing of SAA is associated with the pathological changes in several diseases, including secondary amyloidosis, our findings may shed light on the structure-function relationship of SAA1 with respect to its role in inflammation.


Asunto(s)
Inflamación/inmunología , Lipopolisacáridos/inmunología , Macrófagos/inmunología , Proteína Amiloide A Sérica/química , Proteína Amiloide A Sérica/inmunología , Animales , Quimiotaxis/efectos de los fármacos , Interleucina-10/biosíntesis , Interleucina-10/genética , Interleucina-10/inmunología , Interleucina-1beta/biosíntesis , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Interleucina-6/biosíntesis , Interleucina-6/genética , Interleucina-6/inmunología , Lipopolisacáridos/administración & dosificación , Ratones , Receptores de Formil Péptido/inmunología , Receptores de Formil Péptido/metabolismo , Proteína Amiloide A Sérica/metabolismo , Proteína Amiloide A Sérica/farmacología , Transducción de Señal , Receptor Toll-Like 2/inmunología , Receptor Toll-Like 3/inmunología , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
20.
PLoS One ; 12(4): e0175824, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28423002

RESUMEN

Serum amyloid A (SAA) is an acute phase protein with cytokine-like and chemotactic properties, that is markedly up-regulated during various inflammatory conditions. Several receptors, including FPRL-1, TLR2, TLR4, RAGE, class B scavenger receptors, SR-BI and CD36, have been identified as SAA receptors. This study provides new evidence that SR-BII, splice variant of SR-BI, could function as an SAA receptor mediating its uptake and pro-inflammatory signaling. The uptake of Alexa Fluor488 SAA was markedly (~3 fold) increased in hSR-BII-expressing HeLa cells when compared with mock-transfected cells. The levels of SAA-induced interleukin-8 secretion by hSR-BII-expressing HEK293 cells were also significantly (~3-3.5 fold) higher than those detected in control cells. Moderately enhanced levels of phosphorylation of all three mitogen-activated protein kinases, ERK1/2, and p38 and JNK, were observed in hSR-BII-expressing cells following SAA stimulation when compared with control wild type cells. Transgenic mice with pLiv-11-directed liver/kidney overexpression of hSR-BI or hSR-BII were used to assess the in vivo role of each receptor in SAA-induced pro-inflammatory response in these organs. Six hours after intraperitoneal SAA injection both groups of transgenic mice demonstrated markedly higher (~2-5-fold) expression levels of inflammatory mediators in the liver and kidney compared to wild type mice. Histological examinations of hepatic and renal tissue from SAA-treated mice revealed moderate level of damage in the liver of both transgenic but not in the wild type mice. Activities of plasma transaminases, biomarkers of liver injury, were also moderately higher in hSR-B transgenic mice when compared to wild type mice. Our findings identify hSR-BII as a functional SAA receptor that mediates SAA uptake and contributes to its pro-inflammatory signaling via the MAPKs-mediated signaling pathways.


Asunto(s)
Riñón/metabolismo , Hígado/metabolismo , Proteínas de Membrana de los Lisosomas/metabolismo , Receptores Depuradores/metabolismo , Proteína Amiloide A Sérica/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Transporte Biológico , Colorantes Fluorescentes/metabolismo , Fluorobencenos/metabolismo , Regulación de la Expresión Génica , Células HEK293 , Células HeLa , Humanos , Inflamación/inducido químicamente , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Riñón/efectos de los fármacos , Riñón/patología , Hígado/efectos de los fármacos , Hígado/patología , Proteínas de Membrana de los Lisosomas/genética , MAP Quinasa Quinasa 4/genética , MAP Quinasa Quinasa 4/metabolismo , Ratones , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Receptores Depuradores/genética , Proteína Amiloide A Sérica/genética , Proteína Amiloide A Sérica/farmacología , Transducción de Señal , Transfección , Transgenes , Proteínas Quinasas p38 Activadas por Mitógenos/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA