Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Int J Mol Sci ; 24(24)2023 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-38139330

RESUMO

Serum amyloid A (SAA) is a family of proteins, the plasma levels of which may increase >1000-fold in acute inflammatory states. We investigated the role of SAA in sepsis using mice deficient in all three acute-phase SAA isoforms (SAA-TKO). SAA deficiency significantly increased mortality rates in the three experimental sepsis mouse models: cecal ligation and puncture (CLP), cecal slurry (CS) injection, and lipopolysaccharide (LPS) treatments. SAA-TKO mice had exacerbated lung pathology compared to wild-type (WT) mice after CLP. A bulk RNA sequencing performed on lung tissues excised 24 h after CLP indicated significant enrichment in the expression of genes associated with chemokine production, chemokine and cytokine-mediated signaling, neutrophil chemotaxis, and neutrophil migration in SAA-TKO compared to WT mice. Consistently, myeloperoxidase activity and neutrophil counts were significantly increased in the lungs of septic SAA-TKO mice compared to WT mice. The in vitro treatment of HL-60, neutrophil-like cells, with SAA or SAA bound to a high-density lipoprotein (SAA-HDL), significantly decreased cellular transmigration through laminin-coated membranes compared to untreated cells. Thus, SAA potentially prevents neutrophil transmigration into injured lungs, thus reducing exacerbated tissue injury and mortality. In conclusion, we demonstrate for the first time that endogenous SAA plays a protective role in sepsis, including ameliorating lung injury.


Assuntos
Lesão Pulmonar , Sepse , Animais , Camundongos , Lesão Pulmonar/patologia , Proteína Amiloide A Sérica/genética , Sepse/patologia , Pulmão/patologia , Quimiocinas , Camundongos Endogâmicos C57BL , Modelos Animais de Doenças
2.
J Lipid Res ; 64(5): 100365, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37004910

RESUMO

Serum amyloid A (SAA) is predictive of CVD in humans and causes atherosclerosis in mice. SAA has many proatherogenic effects in vitro. However, HDL, the major carrier of SAA in the circulation, masks these effects. The remodeling of HDL by cholesteryl ester transfer protein (CETP) liberates SAA restoring its proinflammatory activity. Here, we investigated whether deficiency of SAA suppresses the previously described proatherogenic effect of CETP. ApoE-/- mice and apoE-/- mice deficient in the three acute-phase isoforms of SAA (SAA1.1, SAA2.1, and SAA3; "apoE-/- SAA-TKO") with and without adeno-associated virus-mediated expression of CETP were studied. There was no effect of CETP expression or SAA genotype on plasma lipids or inflammatory markers. Atherosclerotic lesion area in the aortic arch of apoE-/- mice was 5.9 ± 1.2%; CETP expression significantly increased atherosclerosis in apoE-/- mice (13.1 ± 2.2%). However, atherosclerotic lesion area in the aortic arch of apoE-/- SAA-TKO mice (5.1 ± 1.1%) was not significantly increased by CETP expression (6.2 ± 0.9%). The increased atherosclerosis in apoE-/- mice expressing CETP was associated with markedly increased SAA immunostaining in aortic root sections. Thus, SAA augments the atherogenic effects of CETP, which suggests that inhibiting CETP may be of particular benefit in patients with high SAA.


Assuntos
Aterosclerose , Proteínas de Transferência de Ésteres de Colesterol , Humanos , Camundongos , Animais , Proteínas de Transferência de Ésteres de Colesterol/genética , Proteína Amiloide A Sérica/metabolismo , Aterosclerose/metabolismo , Apolipoproteínas E/metabolismo , Aorta/metabolismo
3.
Cell Mol Gastroenterol Hepatol ; 12(4): 1329-1341, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34217896

RESUMO

BACKGROUND & AIMS: Identifying new approaches to lessen inflammation, as well as the associated malignant consequences, remains crucial to improving the lives and prognosis of patients diagnosed with inflammatory bowel diseases. Although it previously has been suggested as a suitable biomarker for monitoring disease activity in patients diagnosed with Crohn's disease, the role of the acute-phase protein serum amyloid A (SAA) in inflammatory bowel disease remains unclear. In this study, we aimed to assess the role of SAA in colitis-associated cancer. METHODS: We established a model of colitis-associated cancer in wild-type and SAA double-knockout (Saa1/2-/-) mice by following the azoxymethane/dextran sulfate sodium protocol. Disease activity was monitored throughout the study while colon and tumor tissues were harvested for subsequent use in cytokine analyses, Western blot, and immunohistochemistry +experiments. RESULTS: We observed attenuated disease activity in mice deficient for Saa1/2 as evidenced by decreased weight loss, increased stool consistency, decreased rectal bleeding, and decreased colitis-associated tissue damage. Macrophage infiltration, including CD206+ M2-like macrophages, also was attenuated in SAA knockout mice, while levels of interleukin 4, interleukin 10, and tumor necrosis factor-ɑ were decreased in the distal colon. Mice deficient for SAA also showed a decreased tumor burden, and tumors were found to have increased apoptotic activity coupled with decreased expression for markers of proliferation. CONCLUSION: Based on these findings, we conclude that SAA has an active role in inflammatory bowel disease and that it could serve as a therapeutic target aimed at decreasing chronic inflammation and the associated risk of developing colitis-associated cancer.


Assuntos
Transformação Celular Neoplásica/metabolismo , Neoplasias Associadas a Colite/etiologia , Neoplasias Associadas a Colite/metabolismo , Suscetibilidade a Doenças , Proteína Amiloide A Sérica/metabolismo , Animais , Biomarcadores , Transformação Celular Neoplásica/genética , Neoplasias Associadas a Colite/patologia , Modelos Animais de Doenças , Imuno-Histoquímica , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Camundongos Knockout , Isoformas de Proteínas , Proteína Amiloide A Sérica/genética
4.
J Lipid Res ; 61(3): 328-337, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31915139

RESUMO

Liver-derived serum amyloid A (SAA) is present in plasma where it is mainly associated with HDL and from which it is cleared more rapidly than are the other major HDL-associated apolipoproteins. Although evidence suggests that lipid-free and HDL-associated forms of SAA have different activities, the pathways by which SAA associates and disassociates with HDL are poorly understood. In this study, we investigated SAA lipidation by hepatocytes and how this lipidation relates to the formation of nascent HDL particles. We also examined hepatocyte-mediated clearance of lipid-free and HDL-associated SAA. We prepared hepatocytes from mice injected with lipopolysaccharide or an SAA-expressing adenoviral vector. Alternatively, we incubated primary hepatocytes from SAA-deficient mice with purified SAA. We analyzed conditioned media to determine the lipidation status of endogenously produced and exogenously added SAA. Examining the migration of lipidated species, we found that SAA is lipidated and forms nascent particles that are distinct from apoA-I-containing particles and that apoA-I lipidation is unaltered when SAA is overexpressed or added to the cells, indicating that SAA is not incorporated into apoA-I-containing HDL during HDL biogenesis. Like apoA-I formation, generation of SAA-containing particles was dependent on ABCA1, but not on scavenger receptor class B type I. Hepatocytes degraded significantly more SAA than apoA-I. Taken together, our results indicate that SAA's lipidation and metabolism by the liver is independent of apoA-I and that SAA is not incorporated into HDL during HDL biogenesis.


Assuntos
Lipoproteínas HDL/metabolismo , Proteína Amiloide A Sérica/metabolismo , Animais , Apolipoproteína A-I/deficiência , Apolipoproteína A-I/metabolismo , Hepatócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Amiloide A Sérica/deficiência , Proteína Amiloide A Sérica/genética
5.
Nature ; 567(7747): 249-252, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30842658

RESUMO

The liver is the most common site of metastatic disease1. Although this metastatic tropism may reflect the mechanical trapping of circulating tumour cells, liver metastasis is also dependent, at least in part, on the formation of a 'pro-metastatic' niche that supports the spread of tumour cells to the liver2,3. The mechanisms that direct the formation of this niche are poorly understood. Here we show that hepatocytes coordinate myeloid cell accumulation and fibrosis within the liver and, in doing so, increase the susceptibility of the liver to metastatic seeding and outgrowth. During early pancreatic tumorigenesis in mice, hepatocytes show activation of signal transducer and activator of transcription 3 (STAT3) signalling and increased production of serum amyloid A1 and A2 (referred to collectively as SAA). Overexpression of SAA by hepatocytes also occurs in patients with pancreatic and colorectal cancers that have metastasized to the liver, and many patients with locally advanced and metastatic disease show increases in circulating SAA. Activation of STAT3 in hepatocytes and the subsequent production of SAA depend on the release of interleukin 6 (IL-6) into the circulation by non-malignant cells. Genetic ablation or blockade of components of IL-6-STAT3-SAA signalling prevents the establishment of a pro-metastatic niche and inhibits liver metastasis. Our data identify an intercellular network underpinned by hepatocytes that forms the basis of a pro-metastatic niche in the liver, and identify new therapeutic targets.


Assuntos
Hepatócitos/patologia , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/secundário , Fígado/patologia , Metástase Neoplásica , Neoplasias Pancreáticas/patologia , Microambiente Tumoral , Animais , Carcinoma Ductal Pancreático/patologia , Neoplasias Colorretais/patologia , Neoplasias Colorretais/secundário , Feminino , Interleucina-6/metabolismo , Masculino , Camundongos , Fator de Transcrição STAT3/metabolismo , Proteína Amiloide A Sérica/metabolismo
6.
Atherosclerosis ; 268: 32-35, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29175652

RESUMO

BACKGROUND AND AIMS: Serum amyloid A (SAA) predicts cardiovascular events. Overexpression of SAA increases atherosclerosis development; however, deficiency of two of the murine acute phase isoforms, SAA1.1 and SAA2.1, has no effect on atherosclerosis. SAA3 is a pseudogene in humans, but is an expressed acute phase isoform in mice. The goal of this study was to determine if SAA3 affects atherosclerosis in mice. METHODS: ApoE-/- mice were used as the model for all studies. SAA3 was overexpressed by an adeno-associated virus or suppressed using an anti-sense oligonucleotide approach. RESULTS: Over-expression of SAA3 led to a 4-fold increase in atherosclerosis lesion area compared to control mice (p = 0.01). Suppression of SAA3 decreased atherosclerosis in mice genetically deficient in SAA1.1 and SAA2.1 (p < 0.0001). CONCLUSIONS: SAA3 augments atherosclerosis in mice. Our results resolve a previous paradox in the literature and support extensive epidemiological data that SAA is pro-atherogenic.


Assuntos
Aorta/metabolismo , Doenças da Aorta/sangue , Aterosclerose/sangue , Placa Aterosclerótica , Proteína Amiloide A Sérica/metabolismo , Animais , Aorta/patologia , Doenças da Aorta/diagnóstico , Doenças da Aorta/patologia , Doenças da Aorta/prevenção & controle , Aterosclerose/genética , Aterosclerose/patologia , Aterosclerose/prevenção & controle , Modelos Animais de Doenças , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout para ApoE , Proteína Amiloide A Sérica/deficiência , Proteína Amiloide A Sérica/genética
7.
Arterioscler Thromb Vasc Biol ; 35(5): 1156-65, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25745063

RESUMO

OBJECTIVE: Rupture of abdominal aortic aneurysm (AAA), a major cause of death in the aged population, is characterized by vascular inflammation and matrix degradation. Serum amyloid A (SAA), an acute-phase reactant linked to inflammation and matrix metalloproteinase induction, correlates with aortic dimensions before aneurysm formation in humans. We investigated whether SAA deficiency in mice affects AAA formation during angiotensin II (Ang II) infusion. APPROACH AND RESULTS: Plasma SAA increased ≈60-fold in apoE(-/-) mice 24 hours after intraperitoneal Ang II injection (100 µg/kg; n=4) and ≈15-fold after chronic 28-day Ang II infusion (1000 ng/kg per minute; n=9). AAA incidence and severity after 28-day Ang II infusion was significantly reduced in apoE(-/-) mice lacking both acute-phase SAA isoforms (SAAKO; n=20) compared with apoE(-/-) mice (SAAWT; n=20) as assessed by in vivo ultrasound and ex vivo morphometric analyses, despite a significant increase in systolic blood pressure in SAAKO mice compared with SAAWT mice after Ang II infusion. Atherosclerotic lesion area of the aortic arch was similar in SAAKO and SAAWT mice after 28-day Ang II infusion. Immunostaining detected SAA in AAA tissues of Ang II-infused SAAWT mice that colocalized with macrophages, elastin breaks, and enhanced matrix metalloproteinase activity. Matrix metalloproteinase-2 activity was significantly lower in aortas of SAAKO mice compared with SAAWT mice after 10-day Ang II infusion. CONCLUSIONS: Lack of endogenous acute-phase SAA protects against experimental AAA through a mechanism that may involve reduced matrix metalloproteinase-2 activity.


Assuntos
Angiotensina II/farmacologia , Aneurisma da Aorta Abdominal/prevenção & controle , Apolipoproteínas E/deficiência , Metaloproteinase 2 da Matriz/metabolismo , Proteína Amiloide A Sérica/deficiência , Animais , Aneurisma da Aorta Abdominal/patologia , Biomarcadores/sangue , Modelos Animais de Doenças , Elastina/metabolismo , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Distribuição Aleatória , Sensibilidade e Especificidade , Proteína Amiloide A Sérica/metabolismo
8.
Arterioscler Thromb Vasc Biol ; 34(2): 255-61, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24265416

RESUMO

OBJECTIVE: Although elevated plasma concentrations of serum amyloid A (SAA) are associated strongly with increased risk for atherosclerotic cardiovascular disease in humans, the role of SAA in the pathogenesis of lesion formation remains obscure. Our goal was to determine the impact of SAA deficiency on atherosclerosis in hypercholesterolemic mice. APPROACH AND RESULTS: Apolipoprotein E-deficient (apoE(-/-)) mice, either wild type or deficient in both major acute phase SAA isoforms, SAA1.1 and SAA2.1, were fed a normal rodent diet for 50 weeks. Female mice, but not male apoE-/- mice deficient in SAA1.1 and SAA2.1, had a modest increase (22%; P≤0.05) in plasma cholesterol concentrations and a 53% increase in adipose mass compared with apoE-/- mice expressing SAA1.1 and SAA2.1 that did not affect the plasma cytokine levels or the expression of adipose tissue inflammatory markers. SAA deficiency did not affect lipoprotein cholesterol distributions or plasma triglyceride concentrations in either male or female mice. Atherosclerotic lesion areas measured on the intimal surfaces of the arch, thoracic, and abdominal regions were not significantly different between apoE-/- mice deficient in SAA1.1 and SAA2.1 and apoE-/- mice expressing SAA1.1 and SAA2.1 in either sex. To accelerate lesion formation, mice were fed a Western diet for 12 weeks. SAA deficiency had effect neither on diet-induced alterations in plasma cholesterol, triglyceride, or cytokine concentrations nor on aortic atherosclerotic lesion areas in either male or female mice. In addition, SAA deficiency in male mice had no effect on lesion areas or macrophage accumulation in the aortic roots. CONCLUSIONS: The absence of endogenous SAA1.1 and 2.1 does not affect atherosclerotic lipid deposition in apolipoprotein E-deficient mice fed either normal or Western diets.


Assuntos
Doenças da Aorta/metabolismo , Apolipoproteínas E/deficiência , Aterosclerose/metabolismo , Proteína Amiloide A Sérica/deficiência , Tecido Adiposo/metabolismo , Tecido Adiposo/fisiopatologia , Adiposidade , Animais , Aorta Abdominal/metabolismo , Aorta Abdominal/patologia , Aorta Torácica/metabolismo , Aorta Torácica/patologia , Doenças da Aorta/genética , Doenças da Aorta/patologia , Doenças da Aorta/fisiopatologia , Apolipoproteínas E/genética , Aterosclerose/genética , Aterosclerose/patologia , Aterosclerose/fisiopatologia , Colesterol/sangue , Citocinas/sangue , Modelos Animais de Doenças , Feminino , Hipercolesterolemia/complicações , Hipercolesterolemia/genética , Hipercolesterolemia/metabolismo , Mediadores da Inflamação/sangue , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Amiloide A Sérica/genética , Fatores de Tempo , Triglicerídeos/sangue
9.
Cytokine ; 61(2): 506-12, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23165195

RESUMO

SAA has been shown to have potential proinflammatory properties in inflammatory diseases such as atherosclerosis. These include induction of tumor necrosis factor α, interleukin-6, and monocyte chemoattractant protein 1 in vitro. However, concern has been raised that these effects might be due to use of recombinant SAA with low level of endotoxin contaminants or its non-native forms. Therefore, physiological relevance has not been fully elucidated. In this study, we investigated the role of SAA in the production of inflammatory cytokines. Stimulation of mouse monocyte J774 cells with lipid-poor recombinant human SAA and purified SAA derived from cardiac surgery patients, but not ApoA-I and ApoA-II, elicited pro-inflammatory cytokines like granulocyte colony stimulating factor (G-CSF). However, HDL-associated SAA failed to stimulate production of these cytokines. Using neutralizing antibodies against toll like receptor (TLR) 2 and 4, we could evaluate that TLR 2 is responsible for G-CSF production by lipid-poor SAA. To confirm these data in vivo, we expressed mouse SAA in SAA deficient C57BL/6 mice using an adenoviral vector. G-CSF was identically expressed in SAA-Adenoviral infected mice as well as in control null-Adenoviral mice at the early time points (4-8h) and could not be detected in plasma 24h after infection when plasma SAA levels were maximally elevated, indicating that adenoviral vector rather than SAA affected G-CSF levels. Taken together, our findings suggest that lipid-poor SAA, but not HDL-associated SAA, stimulates G-CSF production and this stimulation is mediated through TLR 2 in J774 cells. However, its physiological role in vivo remains ambiguous.


Assuntos
Citocinas/biossíntese , Proteína Amiloide A Sérica/metabolismo , Animais , Apolipoproteína A-I/metabolismo , Apolipoproteína A-II/metabolismo , Fator Estimulador de Colônias de Granulócitos/metabolismo , Humanos , Lipopolissacarídeos , Lipoproteínas HDL/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Substâncias Protetoras/farmacologia , Proteína Amiloide A Sérica/deficiência , Receptor 2 Toll-Like/metabolismo
10.
J Lipid Res ; 52(12): 2255-2261, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21957202

RESUMO

Inflammation is associated with significant decreases in plasma HDL-cholesterol (HDL-C) and apoA-I levels. Endothelial lipase (EL) is known to be an important determinant of HDL-C in mice and in humans and is upregulated during inflammation. In this study, we investigated whether serum amyloid A (SAA), an HDL apolipoprotein highly induced during inflammation, alters the ability of EL to metabolize HDL. We determined that EL hydrolyzes SAA-enriched HDL in vitro without liberating lipid-free apoA-I. Coexpression of SAA and EL in mice by adenoviral vector produced a significantly greater reduction in HDL-C and apoA-I than a corresponding level of expression of either SAA or EL alone. The loss of HDL occurred without any evidence of HDL remodeling to smaller particles that would be expected to have more rapid turnover. Studies with primary hepatocytes demonstrated that coexpression of SAA and EL markedly impeded ABCA1-mediated lipidation of apoA-I to form nascent HDL. Our findings suggest that a reduction in nascent HDL formation may be partly responsible for reduced HDL-C during inflammation when both EL and SAA are known to be upregulated.


Assuntos
Hepatócitos/metabolismo , Lipase/metabolismo , Lipoproteínas HDL/metabolismo , Proteína Amiloide A Sérica/metabolismo , Animais , Células COS , Chlorocebus aethiops , HDL-Colesterol/metabolismo , Humanos , Inflamação/metabolismo , Camundongos , Camundongos Endogâmicos C57BL
11.
J Lipid Res ; 52(2): 345-53, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21138980

RESUMO

ATP binding cassette transporter G1 (ABCG1) mediates the transport of cellular cholesterol to HDL, and it plays a key role in maintaining macrophage cholesterol homeostasis. During inflammation, HDL undergoes substantial remodeling, acquiring lipid changes and serum amyloid A (SAA) as a major apolipoprotein. In the current study, we investigated whether remodeling of HDL that occurs during acute inflammation impacts ABCG1-dependent efflux. Our data indicate that lipid free SAA acts similarly to apolipoprotein A-I (apoA-I) in mediating sequential efflux from ABCA1 and ABCG1. Compared with normal mouse HDL, acute phase (AP) mouse HDL containing SAA exhibited a modest but significant 17% increase in ABCG1-dependent efflux. Interestingly, AP HDL isolated from mice lacking SAA (SAAKO mice) was even more effective in promoting ABCG1 efflux. Hydrolysis with Group IIA secretory phospholipase A(2) (sPLA(2)-IIA) significantly reduced the ability of AP HDL from SAAKO mice to serve as a substrate for ABCG1-mediated cholesterol transfer, indicating that phospholipid (PL) enrichment, and not the presence of SAA, is responsible for alterations in efflux. AP human HDL, which is not PL-enriched, was somewhat less effective in mediating ABCG1-dependent efflux compared with normal human HDL. Our data indicate that inflammatory remodeling of HDL impacts ABCG1-dependent efflux independent of SAA.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Reação de Fase Aguda/fisiopatologia , Colesterol/metabolismo , Lipoproteínas/metabolismo , Proteína Amiloide A Sérica/metabolismo , Membro 1 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Animais , Apolipoproteína A-I/metabolismo , Células Cultivadas , Cricetinae , Humanos , Lipoproteínas HDL/metabolismo , Macrófagos/metabolismo , Camundongos
12.
BMC Gastroenterol ; 10: 133, 2010 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-21067563

RESUMO

BACKGROUND: Serum Amyloid A (SAA) is a major acute phase protein of unknown function. SAA is mostly expressed in the liver, but also in other tissues including the intestinal epithelium. SAA reportedly has anti-bacterial effects, and because inflammatory bowel diseases (IBD) result from a breakdown in homeostatic interactions between intestinal epithelia and bacteria, we hypothesized that SAA is protective during experimental colitis. METHODS: Intestinal SAA expression was measured in mouse and human samples. Dextran sodium sulfate (DSS) colitis was induced in SAA 1/2 double knockout (DKO) mice and in wildtype controls. Anti-bacterial effects of SAA1/2 were tested in intestinal epithelial cell lines transduced with adenoviral vectors encoding the CE/J SAA isoform or control vectors prior to exposure to live Escherichia coli. RESULTS: Significant levels of SAA1/SAA2 RNA and SAA protein were detected by in situ hybridization and immunohistochemistry in mouse colonic epithelium. SAA3 expression was weaker, but similarly distributed. SAA1/2 RNA was present in the ileum and colon of conventional mice and in the colon of germfree mice. Expression of SAA3 was strongly regulated by bacterial lipopolysaccharides in cultured epithelial cell lines, whereas SAA1/2 expression was constitutive and not LPS inducible. Overexpression of SAA1/2 in cultured epithelial cell lines reduced the viability of co-cultured E. coli. This might partially explain the observed increase in susceptibility of DKO mice to DSS colitis. SAA1/2 expression was increased in colon samples obtained from Crohn's Disease patients compared to controls. CONCLUSIONS: Intestinal epithelial SAA displays bactericidal properties in vitro and could play a protective role in experimental mouse colitis. Altered expression of SAA in intestinal biopsies from Crohn's Disease patients suggests that SAA is involved in the disease process..


Assuntos
Bactérias/crescimento & desenvolvimento , Colite/genética , DNA/genética , Regulação da Expressão Gênica , Proteína Amiloide A Sérica/genética , Animais , Bactérias/efeitos dos fármacos , Biópsia , Linhagem Celular , Colite/microbiologia , Colite/patologia , Modelos Animais de Doenças , Progressão da Doença , Humanos , Immunoblotting , Hibridização In Situ , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase , Proteína Amiloide A Sérica/biossíntese
13.
Circulation ; 122(19): 1919-27, 2010 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-20974999

RESUMO

BACKGROUND: High-density lipoprotein (HDL) protects the artery wall by removing cholesterol from lipid-laden macrophages. However, recent evidence suggests that HDL might also inhibit atherogenesis by combating inflammation. METHODS AND RESULTS: To identify potential antiinflammatory mechanisms, we challenged macrophages with lipopolysaccharide, an inflammatory microbial ligand for Toll-like receptor 4. HDL inhibited the expression of 30 (277 of 911) of the genes normally induced by lipopolysaccharide, microarray analysis revealed. One of its major targets was the type I interferon response pathway, a family of potent viral immunoregulators controlled by Toll-like receptor 4 and the TRAM/TRIF signaling pathway. Unexpectedly, the ability of HDL to inhibit gene expression was independent of macrophage cholesterol stores. Immunofluorescent studies suggested that HDL promoted TRAM translocation to intracellular compartments, which impaired subsequent signaling by Toll-like receptor 4 and TRIF. To examine the potential in vivo relevance of the pathway, we used mice deficient in apolipoprotein A-I, the major protein of HDL. After infection with Salmonella typhimurium, a Gram-negative bacterium that expresses lipopolysaccharide, apolipoprotein A-I-deficient mice had 6-fold higher plasma levels of interferon-ß, a key regulator of the type I interferon response, than did wild-type mice. CONCLUSIONS: HDL inhibits a subset of lipopolysaccharide-stimulated macrophage genes that regulate the type I interferon response, and its action is independent of sterol metabolism. These findings raise the possibility that regulation of macrophage genes by HDL might link innate immunity and cardioprotection.


Assuntos
Interferon Tipo I/imunologia , Lipopolissacarídeos/farmacologia , Lipoproteínas HDL/farmacologia , Macrófagos/imunologia , Animais , Quimiocina CXCL10/metabolismo , Quimiocinas/genética , Citocinas/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Terapia de Imunossupressão , Interferon beta/metabolismo , Interleucina-12/metabolismo , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/genética , Transdução de Sinais/fisiologia , Tioglicolatos/farmacologia , Receptor 4 Toll-Like/agonistas , Receptor 4 Toll-Like/genética , Receptores Toll-Like/genética
14.
Obesity (Silver Spring) ; 18(1): 35-41, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19498343

RESUMO

The epidemic of obesity sweeping developed nations is accompanied by an increase in atherosclerotic cardiovascular diseases. Dyslipidemia, diabetes, hypertension, and obesity are risk factors for cardiovascular disease. However, delineating the mechanism of obesity-accelerated atherosclerosis has been hampered by a paucity of animal models. Similar to humans, apolipoprotein E-deficient (apoE(-/-)) mice spontaneously develop atherosclerosis over their lifetime. To determine whether apoE(-/-) mice would develop obesity with accelerated atherosclerosis, we fed mice diets containing 10 (low fat (LF)) or 60 (high fat (HF)) kcal % from fat for 17 weeks. Mice fed the HF diet had a marked increase in body weight and atherosclerotic lesion formation compared to mice fed the LF diet. There were no significant differences between groups in serum total cholesterol, triglycerides, or leptin concentrations. Plasma concentrations of the acute-phase reactant serum amyloid A (SAA) are elevated in both obesity and cardiovascular disease. Accordingly, plasma SAA concentrations were increased fourfold (P < 0.01) in mice fed the HF diet. SAA was associated with both pro- and antiatherogenic lipoproteins in mice fed the HF diet compared to those fed the LF diet, in which SAA was primarily associated with the antiatherogenic lipoprotein high-density lipoprotein (HDL). Moreover, SAA was localized with apoB-containing lipoproteins and biglycan in the vascular wall. Taken together, these data suggest male apoE-deficient mice are a model of metabolic syndrome and that chronic low level inflammation associated with increased SAA concentrations may mediate atherosclerotic lesion formation.


Assuntos
Apolipoproteínas E/genética , Aterosclerose/genética , Peso Corporal/genética , Dieta , Obesidade/genética , Adiponectina/sangue , Análise de Variância , Animais , Apolipoproteínas E/metabolismo , Aterosclerose/metabolismo , Glicemia/metabolismo , Western Blotting , Quimiocina CCL2/sangue , Cromatografia Líquida de Alta Pressão , Gorduras na Dieta , Progressão da Doença , Ingestão de Energia/genética , Ensaio de Imunoadsorção Enzimática , Processamento de Imagem Assistida por Computador , Interleucina-6/sangue , Leptina/sangue , Lipídeos/sangue , Fígado/metabolismo , Masculino , Camundongos , Camundongos Knockout , Obesidade/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Necrose Tumoral alfa/sangue
15.
Arterioscler Thromb Vasc Biol ; 29(9): 1298-303, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19304574

RESUMO

OBJECTIVE: The purpose of this study was to investigate the interaction of SAA and SR-BI in remodeling of acute phase HDL (AP HDL). METHODS AND RESULTS: We used SAA and SR-BI adenoviral vector expression models to study the interaction between these entities. SR-BI processing of mouse AP HDL generated progressively smaller discreet HDL particles with distinct apolipoprotein compositions. SR-BI actions segregated apolipoproteins with the smallest particles containing only apoA-I. Larger remnants contained apoA-I, apoA-II, and SAA. Small apoA-I only particles failed to associate with preformed HDL, whereas larger remnants readily did. The presence of SAA on SR-BI-processed HDL particles propelled apoA-I to a small lipid-poor form and accelerated apoA-I catabolism. CONCLUSIONS: Data indicate that after core and surface HDL lipid perturbation by SR-BI, SAA propels apoA-I to a small lipid-poor form while accelerating HDL metabolism.


Assuntos
Reação de Fase Aguda/metabolismo , Lipoproteínas HDL/metabolismo , Receptores Depuradores Classe B/metabolismo , Reação de Fase Aguda/induzido quimicamente , Adenoviridae/genética , Animais , Apolipoproteína A-I/deficiência , Apolipoproteína A-I/genética , Apolipoproteína A-II/metabolismo , Transporte Biológico , Modelos Animais de Doenças , Vetores Genéticos , Lipopolissacarídeos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Tamanho da Partícula , Receptores Depuradores Classe B/genética , Proteína Amiloide A Sérica/genética , Proteína Amiloide A Sérica/metabolismo , Fatores de Tempo
16.
Arterioscler Thromb Vasc Biol ; 29(2): 261-7, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19008529

RESUMO

OBJECTIVE: The purpose of this study was to examine the interactive action of serum amyloid A (SAA), group IIA secretory phospholipase A(2) (sPLA(2)-IIA), and cholesteryl ester transfer protein (CETP) on HDL remodeling and cholesterol efflux during the acute phase (AP) response elicited in humans after cardiac surgery. METHODS AND RESULTS: Plasma was collected from patients before (pre-AP), 24 hours after (AP-1 d), and 5 days after cardiac surgery (AP-5 d). SAA levels were increased 16-fold in AP-1 d samples. The activity of sPLA(2)-IIA was increased from 77.7+/-38.3 U/mL (pre-AP) to 281.4+/-57.1 U/mL (AP-1 d; P<0.001). CETP mass and activity reduction was commensurate to the reduction of HDL cholesterol levels. The combined action of SAA, sPLA(2)-IIA, and CETP in vitro markedly remodeled HDL with the generation of lipid-poor apoA-I from both pre-AP and AP-1 d HDL. The net result of this remodeling was a relative preservation of ABCA1- and ABCG1-dependent cholesterol efflux during the acute phase response. CONCLUSIONS: Our results show that the many and complex changes in plasma proteins during the acute phase response markedly remodel HDL with functional implications, particularly the relative retention of cholesterol efflux capacity.


Assuntos
Reação de Fase Aguda/etiologia , Procedimentos Cirúrgicos Cardíacos/efeitos adversos , Colesterol/sangue , Lipoproteínas HDL/sangue , Macrófagos/metabolismo , Transportador 1 de Cassete de Ligação de ATP , Membro 1 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/sangue , Reação de Fase Aguda/sangue , Apolipoproteína A-I/sangue , Proteínas de Transferência de Ésteres de Colesterol/sangue , Cromatografia de Afinidade , Fosfolipases A2 do Grupo II/sangue , Humanos , Proteínas Recombinantes/metabolismo , Proteína Amiloide A Sérica/metabolismo , Fatores de Tempo
17.
Am J Pathol ; 173(6): 1902-10, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18974302

RESUMO

Inflammatory markers serum amyloid A (SAA) and C-reactive protein (CRP) are predictive of cardiac disease and are proposed to play causal roles in the development of atherosclerosis, in which the retention of lipoproteins by vascular wall proteoglycans is critical. The purpose of this study was to determine whether SAA and/or CRP alters vascular proteoglycan synthesis and lipoprotein retention in a pro-atherogenic manner. Vascular smooth muscle cells were stimulated with either SAA or CRP (1 to 100 mg/L) and proteoglycans were then isolated and characterized. SAA, but not CRP, increased proteoglycan sulfate incorporation by 50 to 100% in a dose-dependent manner (P < 0.0001), increased glycosaminoglycan chain length, and increased low-density lipoprotein (LDL) binding affinity (K(d), 29 microg/ml LDL versus 90 microg/ml LDL for SAA versus control proteoglycans; P < 0.005). Furthermore, SAA up-regulated biglycan via the induction of endogenous transforming growth factor (TGF)-beta. To determine whether SAA stimulated proteoglycan synthesis in vivo, ApoE(-/-) mice were injected with an adenovirus expressing human SAA-1, a null virus, or saline. Mice that received adenovirus expressing SAA had increased TGF-beta concentrations in plasma and increased aortic biglycan content compared with mice that received either null virus or saline. Thus, SAA alters vascular proteoglycans in a pro-atherogenic manner via the stimulation of TGF-beta and may play a causal role in the development of atherosclerosis.


Assuntos
Aterosclerose/metabolismo , Proteína C-Reativa/metabolismo , Proteoglicanas/metabolismo , Proteína Amiloide A Sérica/metabolismo , Animais , Aorta/metabolismo , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Biglicano , Células Cultivadas , Proteínas da Matriz Extracelular/metabolismo , Glicosaminoglicanos/química , Glicosaminoglicanos/metabolismo , Haplorrinos , Humanos , Lipoproteínas LDL/metabolismo , Masculino , Camundongos , Camundongos Knockout , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/metabolismo , Proteoglicanas/química , Receptores de Formil Peptídeo/genética , Receptores de Formil Peptídeo/metabolismo , Receptores de Lipoxinas/genética , Receptores de Lipoxinas/metabolismo , Fator de Crescimento Transformador beta/metabolismo
18.
J Biol Chem ; 280(43): 35890-5, 2005 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-16120612

RESUMO

Serum amyloid A (SAA) is an acute phase protein whose expression is markedly up-regulated during inflammation and infection. The physiological function of SAA is unclear. In this study, we reported that SAA promotes cellular cholesterol efflux mediated by scavenger receptor B-I (SR-BI). In Chinese hamster ovary cells, SAA promoted cellular cholesterol efflux in an SR-BI-dependent manner, whereas apoA-I did not. Similarly, SAA, but not apoA-I, promoted cholesterol efflux from HepG2 cells in an SR-BI-dependent manner as shown by using the SR-BI inhibitor BLT-1. When SAA was overexpressed in HepG2 cells using adenovirus-mediated gene transfer, the endogenously expressed SAA promoted SR-BI-dependent efflux. To assess the effect of SAA on SR-BI-mediated efflux to high density lipoprotein (HDL), we compared normal HDL, acute phase HDL (AP-HDL, prepared from mice injected with lipopolysaccharide), and AdSAA-HDL (HDL prepared from mice overexpressing SAA). Both AP-HDL and AdSAA-HDL promoted 2-fold greater cholesterol efflux than normal HDL. Lipid-free SAA was shown to also stimulate ABCA1-dependent cholesterol efflux in fibroblasts, in line with an earlier report (Stonik, J. A., Remaley, A. T., Demosky, S. J., Neufeld, E. B., Bocharov, A., and Brewer, H. B. (2004) Biochem. Biophys. Res. Commun. 321, 936-941). When added to cells together, SAA and HDL exerted a synergistic effect in promoting ABCA1-dependent efflux, suggesting that SAA may remodel HDL in a manner that releases apoA-I or other efficient ABCA1 ligands from HDL. SAA also facilitated efflux by a process that was independent of SR-BI and ABCA1. We conclude that the acute phase protein SAA plays an important role in HDL cholesterol metabolism by promoting cellular cholesterol efflux through a number of different efflux pathways.


Assuntos
Colesterol/metabolismo , Técnicas de Transferência de Genes , Receptores Depuradores Classe B/fisiologia , Proteína Amiloide A Sérica/fisiologia , Transportador 1 de Cassete de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/metabolismo , Adenoviridae/genética , Animais , Células CHO , Linhagem Celular , Cricetinae , Relação Dose-Resposta a Droga , Humanos , Inflamação , Ligantes , Lipoproteínas HDL/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Receptores do Leucotrieno B4/metabolismo , Receptores Purinérgicos P2/metabolismo , Proteínas Recombinantes/metabolismo , Receptores Depuradores Classe B/metabolismo
19.
J Biol Chem ; 280(4): 2954-61, 2005 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-15561721

RESUMO

Serum amyloid A is an acute phase protein that is carried in the plasma largely as an apolipoprotein of high density lipoprotein (HDL). In this study we investigated whether SAA is a ligand for the HDL receptor, scavenger receptor class B type I (SR-BI), and how SAA may influence SR-BI-mediated HDL binding and selective cholesteryl ester uptake. Studies using Chinese hamster ovary cells expressing SR-BI showed that (125)I-labeled SAA, both in lipid-free form and in reconstituted HDL particles, functions as a high affinity ligand for SR-BI. SAA also bound with high affinity to the hepatocyte cell line, HepG2. Alexa-labeled SAA was shown by fluorescence confocal microscopy to be internalized by cells in a SR-BI-dependent manner. To assess how SAA association with HDL influences HDL interaction with SR-BI, SAA-containing HDL was isolated from mice overexpressing SAA through adenoviral gene transfer. SAA presence on HDL had little effect on HDL binding to SR-BI but decreased (30-50%) selective cholesteryl ester uptake. Lipid-free SAA, unlike lipid-free apoA-I, was an effective inhibitor of both SR-BI-dependent binding and selective cholesteryl ester uptake of HDL. We have concluded that SR-BI plays a key role in SAA metabolism through its ability to interact with and internalize SAA and, further, that SAA influences HDL cholesterol metabolism through its inhibitory effects on SR-BI-mediated selective lipid uptake.


Assuntos
Metabolismo dos Lipídeos , Lipoproteínas HDL/metabolismo , Receptores Imunológicos/fisiologia , Proteína Amiloide A Sérica/biossíntese , Adenoviridae/genética , Animais , Antígenos CD36 , Células CHO , Linhagem Celular , Ésteres do Colesterol/metabolismo , Cricetinae , Relação Dose-Resposta a Droga , Técnicas de Transferência de Genes , Humanos , Ligantes , Camundongos , Microscopia Confocal , Ligação Proteica , Isoformas de Proteínas , Receptores Imunológicos/metabolismo , Receptores Depuradores , Receptores Depuradores Classe B , Temperatura , Fatores de Tempo
20.
Arterioscler Thromb Vasc Biol ; 25(1): 168-73, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15514202

RESUMO

OBJECTIVE: To investigate the potential of circulating low-density lipoprotein (LDL), isolated from apolipoprotein E (apoE)-deficient mice (E-/-LDL) and from LDL receptor-deficient mice (Lr-/-LDL), to induce foam cell formation. METHODS AND RESULTS: Binding studies using COS-7 cells overexpressing CD36, J774 cells, and mouse peritoneal macrophages (MPMs) unexpectedly showed for the first time that E-/-LDL, which is enriched in cholesterol, is a high-affinity ligand for CD36 and exhibited greater macrophage uptake than Lr-/-LDL or normal LDL. Minimal copper-mediated oxidization of Lr-/-LDL or C57LDL in vitro resulted in increased ligand internalization, although cell uptake of these oxidized LDLs was lower than that of E-/-LDL, even at oxidation levels similar to that found in E-/-LDL. Treatment of MPMs with E-/-LDL and Lr-/-LDL (to a 2- to 3-fold lesser extent), but not normal LDL, resulted in significant cellular cholesteryl ester accumulation and foam cell formation. Experiments using MPMs lacking CD36, scavenger receptor class A (SR-A), or both, indicated a major contribution of CD36 ( approximately 50%), and to a lesser extent, SR-A (24% to 30%), to E-/-LDL uptake. CONCLUSIONS: Because of its increased state of oxidation and high cholesterol content, LDL in apoE-deficient mice acts in a proatherogenic manner, without requiring further modification in the vascular wall, to induce foam cell formation through its uptake by scavenger receptors.


Assuntos
Apolipoproteínas E/deficiência , Antígenos CD36/metabolismo , Metabolismo dos Lipídeos , Macrófagos Peritoneais/metabolismo , Receptores Imunológicos/metabolismo , Adenoviridae/genética , Animais , Apolipoproteínas/metabolismo , Antígenos CD36/genética , Células COS/química , Células COS/metabolismo , Células COS/virologia , Linhagem Celular , Chlorocebus aethiops , Ésteres do Colesterol/metabolismo , Células Espumosas/metabolismo , Técnicas de Transferência de Genes , Lipoproteínas LDL/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Receptores de LDL/deficiência , Receptores Depuradores , Receptores Depuradores Classe A , Transdução Genética/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA