Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
1.
Eur J Obstet Gynecol Reprod Biol ; 292: 8-16, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37948929

RESUMO

OBJECTIVE: Tibolone is a synthetic steroid with estrogenic, androgenic and progestogenic properties that is used as hormone replacement therapy (HRT) in postmenopausal women. Treatment with tibolone has been demonstrated to lead to changes of the lipid profile, including alterations in lipoprotein (a) and apolipoprotein levels. Hence, we conducted the present meta-analysis of randomized controlled trials (RCTs) to assess the effect of tibolone treatment on apolipoproteins and lipoprotein (a) values in postmenopausal women. METHODS: Several databases (Cochrane Library, PubMed/Medline, Scopus, and Google Scholar) were searched for English-language manuscripts published up to September 2023 that scrutinized the effects of tibolone administration on apolipoprotein A-I (ApoA-I), apolipoprotein A-II (ApoA-II), apolipoprotein B (ApoB), and lipoprotein (a) in postmenopausal women. The results were reported as the weighted mean difference (WMD) with a 95% confidence interval (CI), generated using a random-effects model. RESULTS: Finally, 12 publications with 13 RCT arms were included in the current meta-analysis. The overall results from the random-effects model demonstrated a notable reduction in ApoA-I (n = 9 RCT arms, WMD: -34.96 mg/dL, 95 % CI: -42.44, -27.48, P < 0.001) and lipoprotein (a) (n = 12 RCT arms, WMD: -7.49 mg/dl, 95 % CI: -12.17, -2.81, P = 0.002) after tibolone administration in postmenopausal women. However, treatment with tibolone did not impact ApoA- II (n = 4 RCT arms, WMD: 1.32 mg/dL, 95 % CI: -4.39, 7.05, P = 0.64) and ApoB (n = 9 RCT arms, WMD: -2.68 mg/dL, 95 % CI: -20.98, 15.61, P = 0.77) values. In the subgroup analyses, we noticed a notable decrease in lipoprotein (a) levels when tibolone was prescribed to females aged < 60 years (WMD: -10.78 mg/dl) and when it was prescribed for ≤ 6 months (WMD: -15.69 mg/dl). CONCLUSION: The present meta-analysis of RCTs highlighted that treatment with tibolone reduces lipoprotein (a) and apolipoprotein A-I levels in postmenopausal women. As the decrease in serum lipids' concentrations is associated with a decrease in the risk of cardiovascular disease (CVD), treatment with tibolone could be a suitable therapy for postmenopausal women with elevated CVD risk.


Assuntos
Apolipoproteína A-I , Doenças Cardiovasculares , Feminino , Humanos , Apolipoproteína A-I/farmacologia , Lipoproteína(a)/farmacologia , Pós-Menopausa , Ensaios Clínicos Controlados Aleatórios como Assunto , Apolipoproteínas/farmacologia , Apolipoproteínas B/farmacologia , Doenças Cardiovasculares/prevenção & controle
2.
Exp Gerontol ; 172: 112055, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36521567

RESUMO

BACKGROUND AND AIM: The administration of 17ß-estradiol plus norethisterone acetate seems to confer women cardioprotection, however, its impact on lipoprotein (a) and apolipoproteins' concentrations remains unclear. Thus, we conducted a meta-analysis of randomized controlled trials (RCTs) to investigate the effect of 17ß-estradiol plus norethisterone acetate treatment on lipoprotein (a) and apolipoproteins' values in females. METHODS: We systematically searched four databases (PubMed/MEDLINE, Scopus, Embase, and Web of Science) to identify relevant publications published until March 9th, 2022. No language restrictions were applied. The random-effects model (the DerSimonian and Laird methods) was employed to calculate the weighted mean difference (WMD). RESULTS: The administration of 17ß-estradiol plus norethisterone acetate resulted in a significant decrease of lipoprotein (a) (WMD: -67.59 mg/L, 95 % CI: -106.39 to -28.80; P < 0.001) and apolipoprotein B concentrations (WMD: -3.71 mg/dL, 95 % CI: -6.68 to -0.75; P = 0.014), respectively. No effect of 17ß-estradiol plus norethisterone acetate on apolipoprotein AI (WMD: 0.23 mg/dL, 95 % CI: -3.99 to 4.46; P = 0.91) or AII (WMD: 0.21 mg/dL, 95 % CI: -2.24 to 2.68; P = 0.86) concentrations was detected. In the stratified analysis, there was a notable reduction in lipoprotein (a) levels in the RCTs with a duration of ≥6 months (WMD: -73.34 mg/L), in postmenopausal women with a BMI ≥25 kg/m2 (WMD: -69.85 mg/L) and in postmenopausal women aged ˂60 years (WMD: -61.93 mg/L). CONCLUSION: The present meta-analysis of RCTs demonstrates that 17ß-estradiol plus norethisterone acetate treatment reduces lipoprotein (a) and apolipoprotein B levels in postmenopausal women.


Assuntos
Lipoproteína(a) , Noretindrona , Feminino , Humanos , Apolipoproteínas/farmacologia , Estradiol/farmacologia , Lipídeos , Lipoproteína(a)/farmacologia , Noretindrona/farmacologia , Acetato de Noretindrona/farmacologia , Pós-Menopausa , Ensaios Clínicos Controlados Aleatórios como Assunto , Apolipoproteínas B
4.
Int J Mol Sci ; 21(21)2020 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-33138028

RESUMO

Rhizoctonia solani (Rs) is a soil-borne pathogen with a broad host range. This pathogen incites a wide range of disease symptoms. Knowledge regarding its infection process is fragmented, a typical feature for basidiomycetes. In this study, we aimed at identifying potential fungal effectors and their function. From a group of 11 predicted single gene effectors, a rare lipoprotein A (RsRlpA), from a strain attacking sugar beet was analyzed. The RsRlpA gene was highly induced upon early-stage infection of sugar beet seedlings, and heterologous expression in Cercospora beticola demonstrated involvement in virulence. It was also able to suppress the hypersensitive response (HR) induced by the Avr4/Cf4 complex in transgenic Nicotiana benthamiana plants and functioned as an active protease inhibitor able to suppress Reactive Oxygen Species (ROS) burst. This effector contains a double-psi beta-barrel (DPBB) fold domain, and a conserved serine at position 120 in the DPBB fold domain was found to be crucial for HR suppression. Overall, R. solani seems to be capable of inducing an initial biotrophic stage upon infection, suppressing basal immune responses, followed by a switch to necrotrophic growth. However, regulatory mechanisms between the different lifestyles are still unknown.


Assuntos
Beta vulgaris/imunologia , Lipoproteína(a)/farmacologia , Doenças das Plantas/imunologia , Proteínas de Plantas/farmacologia , Inibidores de Proteases/farmacologia , Rhizoctonia/fisiologia , Virulência , Beta vulgaris/efeitos dos fármacos , Beta vulgaris/crescimento & desenvolvimento , Beta vulgaris/microbiologia , Doenças das Plantas/microbiologia , Microbiologia do Solo
5.
J Clin Endocrinol Metab ; 104(10): 4793-4803, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31220285

RESUMO

CONTEXT: Elevated serum lipoprotein(a) [Lp(a)] levels are associated with increased cardiovascular disease risk. ABCA1-mediated cholesterol efflux from macrophages may be an antiatherogenic process. Plasminogen (PLG) is a driver of ABCA1-mediated cholesterol efflux, and its action is inhibited by purified human Lp(a). OBJECTIVE: To determine the effects of Lp(a) in human serum on ABCA1 cholesterol efflux. METHODS: Cholesterol efflux capacity (CEC) was measured with two different cell-culture models using serum from 76 patients with either low (<50 mg/dL) or high (>50 mg/dL) Lp(a) levels. RESULTS: Using cAMP-stimulated J774 macrophages or baby hamster kidney fibroblasts overexpressing human ABCA1, we show that CEC was lower in patients with high Lp(a) levels compared with patients with low levels (-30.6%, P = 0.002 vs -24.1%, P < 0.001, respectively). Total-serum CEC negatively correlated with Lp(a) levels (r = -0.433, P = 0.0007 vs r = -0.505, P = 0.0011, respectively). These negative associations persisted after adjusting for serum cholesterol, age, sex, and statin use in a multiple linear regression model (adjusted R2 = 0.413 or 0.405, respectively) and were strengthened when further adjusting for the interaction between Lp(a) and PLG levels (adjusted R2 = 0.465 and 0.409, respectively). Total-serum and isolated Lp(a) from patients with high Lp(a) inhibited PLG-mediated ABCA1 cholesterol efflux. CONCLUSION: Total-serum CEC is reduced in patients with high Lp(a) levels. This is in part due to the inhibition of PLG-mediated ABCA1 cholesterol efflux by Lp(a). Our findings suggest an atherogenic role for Lp(a) through its ability to inhibit CEC.


Assuntos
Transportador 1 de Cassete de Ligação de ATP/fisiologia , Colesterol/metabolismo , Lipoproteína(a)/sangue , Lipoproteína(a)/farmacologia , Transportador 1 de Cassete de Ligação de ATP/metabolismo , Adulto , Animais , Transporte Biológico/efeitos dos fármacos , Células Cultivadas , Cricetinae , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Humanos , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Camundongos , Pessoa de Meia-Idade
6.
Hepatology ; 65(6): 1851-1864, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28152568

RESUMO

The development of different cell culture models has greatly contributed to increased understanding of the hepatitis C virus (HCV) life cycle. However, it is still challenging to grow HCV clinical isolates in cell culture. If overcome, this would open new perspectives to study HCV biology, including drug-resistant variants emerging with new antiviral therapies. In this study we hypothesized that this hurdle could be due to the presence of inhibitory factors in patient serum. Combining polyethylene glycol precipitation, iodixanol gradient, and size-exclusion chromatography, we obtained from HCV-seronegative sera a purified fraction enriched in inhibitory factors. Mass spectrometric analysis identified apolipoprotein(a) (apo[a]) as a potential inhibitor of HCV entry. Apo(a) consists of 10 kringle IV domains (KIVs), one kringle V domain, and an inactive protease domain. The 10 KIVs are present in a single copy with the exception of KIV type 2 (KIV2 ), which is encoded in a variable number of tandemly repeated copies, giving rise to numerous apo(a) size isoforms. In addition, apo(a) covalently links to the apolipoprotein B component of a low-density lipoprotein through a disulfide bridge to form lipoprotein(a). Using a recombinant virus derived from the JFH1 strain, we confirmed that plasma-derived and recombinant lipoprotein(a) as well as purified recombinant apo(a) variants were able to specifically inhibit HCV by interacting with infectious particles. Our results also suggest that small isoforms are less inhibitory than the large ones. Finally, we observed that the lipoprotein moiety of HCV lipoviroparticles was essential for inhibition, whereas functional lysine-binding sites in KIV7 , KIV8 , and KIV10 were not required. CONCLUSIONS: Our results identify apo(a) as an additional component of the lipid metabolism modulating HCV infection. (Hepatology 2017;65:1851-1864).


Assuntos
Hepacivirus/metabolismo , Hepatite C/terapia , Lipoproteína(a)/farmacologia , Western Blotting , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Cromatografia Líquida/métodos , Hepacivirus/efeitos dos fármacos , Hepatite C/sangue , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Humanos , Imunoprecipitação , Lisina/metabolismo , Ligação Proteica , Sensibilidade e Especificidade , Relação Estrutura-Atividade
8.
J Lipid Res ; 56(7): 1318-28, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25852127

RESUMO

Elevated levels of lipoprotein (a) [Lp(a)] are a well-established risk factor for developing CVD. While Lp(a) levels are thought to be independent of other plasma lipoproteins, some trials have reported a positive association between Lp(a) and HDL. Whether Lp(a) has a direct effect on HDL is not known. Here we investigated to determine whether Lp(a) had any effect on the ABCA1 pathway of HDL production in liver cells. Incubation of HepG2 cells with Lp(a) upregulated the PPARγ protein by 1.7-fold and the liver X receptor α protein by 3-fold. This was accompanied by a 1.8-fold increase in ABCA1 protein and a 1.5-fold increase in cholesterol efflux onto apoA1. We showed that Lp(a) was internalized by HepG2 cells, however, the ABCA1 response to Lp(a) was mediated by the selective uptake of oxidized phospholipids (oxPLs) from Lp(a) via the scavenger receptor-B1 and not by Lp(a) internalization per se. We conclude that there is a biological connection between Lp(a) and HDL through the ability of Lp(a)'s oxPLs to upregulate HDL biosynthesis.


Assuntos
Transportador 1 de Cassete de Ligação de ATP/genética , Antígenos CD36/metabolismo , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Lipoproteína(a)/farmacologia , Fosfolipídeos/metabolismo , Regulação para Cima/efeitos dos fármacos , Células Hep G2 , Humanos , Oxirredução/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos
9.
Biochem Biophys Res Commun ; 423(1): 79-84, 2012 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-22634007

RESUMO

BACKGROUND: Lipoprotein (a) (Lp(a)) is one of the risk factors for peripheral artery disease (PAD). Our previous report demonstrated that hepatocyte growth factor (HGF) gene therapy attenuated the impairment of collateral formation in Lp(a) transgenic mice. Since risk factors for atherosclerosis accelerate endothelial senescence and impair angiogenesis, we examined the role of Lp(a) in dysfunction and senescence of endothelial progenitor cells (EPC) and endothelial cells. METHODS: In vitro and in vivo incorporation assays were performed using ex-vivo expanded DiI-labeled human EPC. Senescence of cultured endothelial cells, production of oxidative stress and angiogenesis function were evaluated by SA-ß-galactosidase staining, dihydroethidium (DHE) staining and Matrigel assay, respectively. RESULTS: EPC transplantation significantly stimulated recovery of ischemic limb perfusion, while EPC pre-treated with Lp(a) did not increase ischemic limb perfusion. Impairment of angiogenesis by EPC with Lp(a) was associated with a significant decrease in CD31-positive capillaries and DiI-labeled EPC. Importantly, Lp(a) significantly accelerated the onset of senescence and production of reactive oxygen species (ROS) in human aortic endothelial cells, accompanied by a significant increase in the protein expression of p53 and p21. On the other hand, HGF significantly attenuated EPC dysfunction, senescence, ROS production, and p53 and p21 expression induced by Lp(a). CONCLUSION: Lp(a) might affect atherosclerosis via acceleration of senescence, ROS production, and functional impairment of the endothelial cell lineage. HGF might have inhibitory effects on these atherogenic actions of Lp(a).


Assuntos
Células Endoteliais/metabolismo , Fator de Crescimento de Hepatócito/metabolismo , Lipoproteína(a)/antagonistas & inibidores , Neovascularização Fisiológica , Células-Tronco/metabolismo , Animais , Aterosclerose/metabolismo , Aterosclerose/terapia , Células Cultivadas , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Fator de Crescimento de Hepatócito/farmacologia , Membro Posterior/irrigação sanguínea , Humanos , Isquemia/metabolismo , Isquemia/terapia , Lipoproteína(a)/metabolismo , Lipoproteína(a)/farmacologia , Camundongos , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Transplante de Células-Tronco , Células-Tronco/efeitos dos fármacos , Proteína Supressora de Tumor p53/antagonistas & inibidores , Proteína Supressora de Tumor p53/metabolismo
10.
Clin Chim Acta ; 411(23-24): 1915-9, 2010 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-20705065

RESUMO

BACKGROUND: High blood lipoprotein (a) [Lp(a)] concentration is a risk factor for a thrombotic event. Annexin A5 is involved in anticoagulation on the endothelial surface. How Lp(a) affects the annexin A5 function is not clear. This study investigates annexin A5 binding on the cell membrane in the presence of Lp(a). METHODS: Lp(a) was isolated from human blood plasma by ultracentrifugation and annexin A5 protein was purchased commercially. The cell membrane was prepared from primary human umbilical vein endothelial cells (HUVEC) and cultured cell line HepG2 by sucrose density gradient centrifugation. Enzyme-linked immunosorbent assays (ELISA) were used to examine annexin A5 binding to the cell membrane in the presence of Lp(a). Flow cytometry was used to analyze the binding of fluorescence-labeled annexin A5 to phosphatidylserine (PS)-translocated intact cells in the presence of Lp(a). RESULTS: Annexin A5 binding to the cell membrane was attenuated by a high concentration of Lp(a) in both HUVEC and HepG2 membrane surfaces. The phenomenon was also observed with annexin A5 surface labeling of HepG2 cells and flow cytometry analysis. CONCLUSIONS: The results imply that Lp(a) interferes with annexin A5 binding to the procoagulant PS which translocates to the membrane surface under stress condition and therefore may increase the risk for thrombosis.


Assuntos
Anexina A5/metabolismo , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Lipoproteína(a)/farmacologia , Anexina A2/metabolismo , Transporte Biológico/efeitos dos fármacos , Relação Dose-Resposta a Droga , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Citometria de Fluxo , Células Hep G2 , Humanos , Ligantes , Lipoproteína(a)/metabolismo , Fosfatidilserinas/metabolismo , Ligação Proteica/efeitos dos fármacos , Estresse Fisiológico/efeitos dos fármacos
11.
Clín. investig. arterioscler. (Ed. impr.) ; 22(supl.1): 3-8, abr. 2010. graf
Artigo em Espanhol | IBECS | ID: ibc-145465

RESUMO

Las lipoproteínas de alta densidad (HDL) transportan colesterol desde la periferia hasta el hígado. Los estudios transversales relacionando las concentraciones bajas de colesterol unido a las HDL (cHDL) con una mayor prevalencia de enfermedad coronaria (EC) datan de los años cincuenta del siglo pasado. Posteriores estudios poblacionales establecieron que el cHDL bajo es un predictor independiente de EC, y así se reconoce en las guías clínicas de prevención cardiovascular. Muchas publicaciones, pero no todas, han establecido una correlación inversa entre incidencia de ictus isquémicos, mortales o no. La proteína transferidora de ésteres de colesterol (CETP) intercambia cHDL por triglicéridos de lipoproteínas de muy baja densidad. Algunas familias con trastornos genéticos de CETP tienen cHDL elevados y menor incidencia de EC. Posteriores estudios observacionales, aunque no todos, han mostrado que sujetos con las anomalías funcionales de CETP tienen cHDL elevado y menor incidencia de EC. Eso ha despertado interés por la inhibición de CETP como intervención para reducir la enfermedad coronaria (AU)


High-density lipoproteins (HDL) transport cholesterol from the periphery to the liver. Cross-sectional studies relating low HDL-cholesterol (HDL-c) concentrations to a higher prevalence of cardiovascular disease (CVD) date back to the 1950s. Subsequent populationbased studies established that low HDL-c levels are an independent predictor of CVD, a finding that is recognized in clinical guidelines for cardiovascular prevention. Many publications, although not all, have established an inverse correlation between the incidence of ischemic stroke, whether fatal or non-fatal, and HDL-c. Cholesteryl ester transfer protein (CETP) facilitates the exchange of triglyceride (for cholesteryl ester) from very low density lipoprotein (VLDL) particles to HDL particles. Some families with genetic CETP alterations have high HDL-c concentrations and a lower incidence of CVD. Some observational studies, but not all, have shown that persons with functional CETP anomalies have high HDL-c levels and a lower incidence of CVD. This observation has prompted interest in CETP inhibition as an intervention to reduce coronary heart disease (AU)


Assuntos
Feminino , Humanos , Masculino , Lipoproteína(a)/deficiência , Lipoproteína(a) , Doenças Cardiovasculares/metabolismo , Doenças Cardiovasculares/patologia , Colesterol/deficiência , Estudos Transversais/métodos , Acidente Vascular Cerebral/metabolismo , Abetalipoproteinemia/metabolismo , Lipoproteína(a)/farmacologia , Lipoproteína(a)/normas , Doenças Cardiovasculares/diagnóstico , Colesterol/metabolismo , Estudos Transversais/instrumentação , Acidente Vascular Cerebral/líquido cefalorraquidiano , Abetalipoproteinemia/patologia
12.
Chin Med J (Engl) ; 123(2): 216-20, 2010 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-20137374

RESUMO

BACKGROUND: Mesangial hypercellularity is a critical early histopathological finding in human and experimental glomerular diseases. Hyperlipidemia and the glomerular deposition of lipoproteins are commonly associated with mesangial hypercellularity and play an important pathobiological role in the development of glomerular diseases. The activated cytoplasmic mitogen-activated protein kinase (MAPK), including mainly extracellular-signal regulated protein kinase (ERK), c-Jun amino-terminal kinase (JNK), and p38, has been thought to translocate into the nucleus and activate various transcription factors and protooncogenes associated with cell growth and proliferation. Lipoprotein (a) (Lp(a)) has been shown to stimulate proliferation of mesangial cells, but the events of Lp(a) signaling have not yet been characterized. The purpose of this study was to investigate the signal transduction pathways involved in Lp(a)-induced cell proliferation and provide an evidence for the participation of Lp(a) in intracellular signaling pathways for mesangial cell proliferation. METHODS: Lp(a) was isolated from a patient who was being treated with low density lipoprotein (LDL)-apheresis by density gradient ultracentrifugation and then chromatography. Human mesangial cells (HMCs) were isolated by the sequential sieving technique and stimulated with Lp(a) in different concentration and time course. The DNA synthesis of the cells was measured by [3H] thymidine incorporation for detecting the proliferation. The expression of all the three members of MAPK family, including ERK1/ERK2, JNK, and p38, and their phosphorylation were detected by Western blotting. RESULTS: Lp(a) could induce a significant dose-dependent proliferation of HMCs. The 3H-TdR incorporation was 1.64+/-0.31, 1.69+/-0.48, 3.59+/-0.68 (P<0.01), 4.14+/-0.78 (P<0.01), and 4.05+/-0.55 (P<0.01) (10(3) cpm) at the Lp(a) concentration of 0, 5, 10, 25, and 50 microg/ml, respectively. Lp(a) induced an increase in ERK1/ERK2 phosphorylation between 5 and 60 minutes, and in JNK phosphorylation between 15 and 30 minutes after incubating with HMCs, whereas the level of p38 and its phosphorylation was not changed. CONCLUSIONS: Lp(a) could stimulate the proliferation of HMCs by activiating the phosphorylation of ERK1/ERK2 and JNK MAPK signaling pathway, whereas p38 pathway had no effect on the Lp(a)-induced HMC proliferation, which indicated that three MAPKs seem to be distinctly involved in the effect. In particular, it also provides the evidence that Lp(a) may act as one of the major endogenous modulators for mitogenic signaling response and cell proliferation within the glomerulus.


Assuntos
Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Lipoproteína(a)/farmacologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Western Blotting , Células Cultivadas , Humanos , Células Mesangiais , Fosforilação/efeitos dos fármacos
13.
FASEB J ; 20(3): 559-61, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16403785

RESUMO

Lipoprotein(a) [Lp(a)], consisting of LDL and the unique constituent apolipoprotein(a) [apo(a)], which contains multiple repeats resembling plasminogen kringle 4, is considered a risk factor for the development of atherosclerotic disorders. However, the underlying mechanisms for the atherogenicity of Lp(a) are not completely understood. Here, we define a novel function of Lp(a) in promoting inflammatory cell recruitment that may contribute to its atherogenicity. Through its apo(a) moiety Lp(a) specifically interacts with the beta2-integrin Mac-1, thereby promoting the adhesion of monocytes and their transendothelial migration in a Mac-1-dependent manner. Interestingly, the interaction between Mac-1 and Lp(a) was strengthened in the presence of proatherogenic homocysteine and was blocked by plasminogen/angiostatin kringle 4. Through its interaction with Mac-1, Lp(a) induced activation of the proinflammatory transcription factor NFkappaB, as well as the NFkappaB-related expression of prothrombotic tissue factor. In atherosclerotic coronary arteries Lp(a) was found to be localized in close proximity to Mac-1 on infiltrating mononuclear cells. Taken together, our data demonstrate that Lp(a), via its apo(a) moiety, is a ligand for the beta2-integrin Mac-1, thereby facilitating inflammatory cell recruitment to atherosclerotic plaques. These observations suggest a novel mechanism for the atherogenic properties of Lp(a).


Assuntos
Aterosclerose/fisiopatologia , Quimiotaxia de Leucócito/fisiologia , Lipoproteína(a)/fisiologia , Antígeno de Macrófago 1/fisiologia , Monócitos/metabolismo , Idoso , Idoso de 80 Anos ou mais , Ácido Aminocaproico/farmacologia , Angiostatinas/farmacologia , Apolipoproteínas A/metabolismo , Aspirina/farmacologia , Movimento Celular , Células Cultivadas/citologia , Células Cultivadas/efeitos dos fármacos , Doença da Artéria Coronariana/metabolismo , Doença da Artéria Coronariana/patologia , Doença da Artéria Coronariana/fisiopatologia , Vasos Coronários/química , Vasos Coronários/patologia , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Endotélio Vascular/citologia , Feminino , Regulação da Expressão Gênica , Homocisteína/farmacologia , Humanos , Molécula 1 de Adesão Intercelular/metabolismo , Lipoproteína(a)/farmacologia , Antígeno-1 Associado à Função Linfocitária/metabolismo , Antígeno de Macrófago 1/química , Masculino , Pessoa de Meia-Idade , Monócitos/citologia , NF-kappa B/metabolismo , Plasminogênio/farmacologia , Ligação Proteica , Estrutura Terciária de Proteína , Transfecção
14.
Clin Chem Lab Med ; 42(9): 1013-9, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15497466

RESUMO

Lipoprotein(a) (Lp(a)) is considered an independent risk factor for atherosclerotic heart and circulatory diseases. The unique, polymorphic character of Lp(a) is based on its apolipoprotein(a) (apo(a)), which has remarkable structural analogies with plasminogen, an important protein for fibrinolysis. The formation of plasmin from plasminogen is a fundamental step in the dissolution of fibrin. Repression of this step may lead to a deceleration of fibrinolysis. It has been suggested that Lp(a) has antifibrinolytic properties through apo(a) and that the apo(a)-size polymorphism has a distinct influence on the prothrombotic properties of Lp(a). However, the results on this topic are controversial. Therefore we used a standardized in vitro fibrinolysis model to provide further information on the influence of Lp(a) on plasmin formation. Monitoring the time-course of plasmin formation, we investigated the inhibition of plasmin formation through dependence on Lp(a), respectively, free apo(a) concentration. Furthermore, we investigated the influence of three Lp(a)/apo(a) phenotypes ((22K)Lp(a), 22 kringle-4 repeats; (30K)Lp(a), 30 kringle-4 repeats; (35K)Lp(a), 35 kringle-4 repeats). Adding varying amounts of Lp(a) to our model, we observed that the rate of plasmin formation was inversely related to the Lp(a) concentration. At 0.1 micromol/l (30K)Lp(a), for example, the plasmin formation was reduced by 12.7% and decreased further by 40.7% at 0.25 micromol/l Lp(a). A similar but more distinct effect was observed when free (30K)apo(a) was added to the model (25.3% at 0.1 micromol/l vs. 59.3% at 0.25 micromol/l). Comparing the antifibrinolytic influence of different apo(a) phenotypes we found that the reduction of plasmin generation advanced with the size of apo(a). At 0.1 micromol/l Lp(a) the reduction of the plasmin formation increased in the order (22K)Lp(a), (30K)Lp(a) and (35K)Lp(a) from 3.7% to 10.7% and 22.3%, respectively. Experiments with different phenotypes of free apo(a) showed similar results (0.5 micromol/l: (22K)apo(a), 56.4% vs. (30K)Lp(a), 80.4%). Summarizing these results, our study indicates a distinct interrelation of Lp(a)/apo(a) phenotype and concentration with the formation of plasmin. From the antifibrinolytic Lp(a)/apo(a) effect in vitro it may be hypothesized that Lp(a)/apo(a) also has an inhibitory influence on in vivo fibrinolysis.


Assuntos
Antifibrinolíticos/farmacologia , Apolipoproteínas/farmacologia , Fibrinolisina/análise , Fibrinólise/fisiologia , Lipoproteína(a)/farmacologia , Apoproteína(a) , Humanos , Técnicas In Vitro , Fenótipo , Plasminogênio/metabolismo
15.
FASEB J ; 18(11): 1306-8, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15208269

RESUMO

Angiogenesis, the growth of new blood vessels, is a complex biological process that is orchestrated by several growth factors and components of the extracellular matrix, including fibronectin (FN) and its receptor the integrin alpha5beta1. Angiogenesis is a critical part of inflammation and wound repair, but the mechanism by which vascular proliferation and migration is regulated by inflammatory cells is not completely understood. We have previously shown that human neutrophil peptides (HNPs), also known as alpha-defensins, which are secreted in high concentrations when neutrophils are activated, bind specifically to FN in the extracellular matrix and inhibit plasminogen activation. Therefore, we asked whether HNPs act as a link between inflammation and angiogenesis. Alpha5beta1-mediated endothelial cell adhesion and migration to FN, both under control conditions and under stimulation by vascular endothelial growth factor (VEGF), were inhibited specifically and in a dose-dependent manner by HNPs, whereas endothelial cell adhesion and migration to other components of the extracellular matrix, such as vitronectin, collagen, or fibrinogen/fibrin were not. Consistent with this finding, HNPs bound to and promoted the binding of fibronectin to alpha5beta1 integrin in arginine-glycine-aspartic acid (RGD)-independent manner. HNPs also completely inhibited VEGF-induced proliferation and induced apoptosis of endothelial cells in a dose-dependent manner. Moreover, HNPs inhibited capillary tube formation in three-dimensional fibrin-matrices as well as neovascularization in vivo in the chicken chorioallantoic membrane assay. Taken together, these data indicate that HNPs can regulate angiogenesis by affecting endothelial cell adhesion and migration in an FN-dependent manner as well as endothelial cell proliferation. These findings provide new insight into the role of inflammatory cells in angiogenesis and might provide a platform for developing a novel class of anti-angiogenesis drugs.


Assuntos
Membrana Corioalantoide/irrigação sanguínea , Endotélio Vascular/efeitos dos fármacos , Neovascularização Fisiológica/efeitos dos fármacos , Esfingosina/análogos & derivados , alfa-Defensinas/farmacologia , Animais , Bovinos , Adesão Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Células Cultivadas/citologia , Células Cultivadas/efeitos dos fármacos , Quimiotaxia/efeitos dos fármacos , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Endotélio Vascular/citologia , Células Epiteliais/efeitos dos fármacos , Fibronectinas/metabolismo , Humanos , Integrina alfa5beta1/metabolismo , Lipoproteína(a)/farmacologia , Lisofosfolipídeos/farmacologia , Ligação Proteica/efeitos dos fármacos , Retina/citologia , Esfingosina/farmacologia , Veias Umbilicais , Fator A de Crescimento do Endotélio Vascular/farmacologia , alfa-Defensinas/fisiologia
16.
Cardiovasc Res ; 63(1): 130-8, 2004 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-15194469

RESUMO

OBJECTIVE: Lipoprotein (a) [Lp(a)] is considered an atherogenic lipoprotein, which is also implicated in thrombosis. Lp(a) binds to platelets and modulates the effects of various platelet agonists. Platelet activating factor (PAF) is a potent platelet agonist degraded and inactivated by PAF-acetylhydrolase (PAF-AH), which in plasma is associated with lipoproteins. Lp(a) is enriched in PAF-AH, thus a functional characteristic of this lipoprotein is its capability to hydrolyze and inactivate PAF. In the present study, we investigated the effect of Lp(a) on PAF-induced platelet activation. The potential roles of the apo(a) moiety and especially of the PAF-AH content of Lp(a) on the above effect were also addressed. METHODS: Lp(a) was isolated by affinity chromatography from plasma of apparently healthy fasting donors with serum Lp(a) concentrations >/=20 mg/dl. Reduced Lp(a) [Lp(a-)] was prepared by incubation of Lp(a) with dithiothreitol (DTT), whereas inactivation of Lp(a)-associated PAF-AH was performed by incubation of Lp(a) with pefabloc [pefa-Lp(a)]. Platelet-rich plasma (PRP) or washed platelets were prepared from peripheral venous blood samples of normolipidemic, apparently healthy fasting donors with their serum Lp(a) levels lower than 0.8 mg/dl. The surface expression of the platelet integrin-receptor alpha(IIb)beta3 and the fibrinogen binding to the activated alpha(IIb)beta3 was studied by flow cytometry. RESULTS: Lp(a), at doses higher than 20 microg/ml, inhibits PAF-induced platelet activation in a dose-dependent manner. Pefa-Lp(a), lacking PAF-AH activity, exhibited a similar to Lp(a) inhibitory effect. Importantly, the Lp(a) inhibitory effect was not influenced by the apo(a) isoform size, whereas Lp(a-) was a more potent inhibitor compared to Lp(a). Similarly to PAF, Lp(a) inhibits platelet aggregation induced by ADP or Calcium ionophore A23187. Lp(a), pefa-Lp(a) or Lp(a-) effectively inhibited PAF- or ADP-induced surface expression of alphaIIbbeta3, the Lp(a-) being more potent compared to Lp(a) or to pefa-Lp(a). Finally, Lp(a) significantly inhibited fibrinogen binding to platelets activated with PAF. CONCLUSIONS: Lp(a) inhibits PAF-induced platelet activation in a non-specific manner. The Lp(a)-associated PAF-AH does not play any important role in this effect, whereas the apo(a) moiety of Lp(a) significantly reduces its inhibitory effect. The inhibition of alpha(IIb)beta3 activation and fibrinogen binding to the activated platelets may represent the major mechanism by which Lp(a) inhibits PAF-induced platelet aggregation.


Assuntos
1-Alquil-2-acetilglicerofosfocolina Esterase/metabolismo , Lipoproteína(a)/farmacologia , Fator de Ativação de Plaquetas/metabolismo , Ativação Plaquetária/efeitos dos fármacos , Plaquetas/efeitos dos fármacos , Depressão Química , Fibrinogênio/metabolismo , Humanos , Técnicas In Vitro
17.
Zhonghua Er Ke Za Zhi ; 42(10): 734-6, 2004 Oct.
Artigo em Chinês | MEDLINE | ID: mdl-16221338

RESUMO

OBJECTIVE: The renal disease is commonly associated with hyperlipidemia and correlates with glomerular accumulation of atherogenic lipoproteins and mesangial hypercellularity. Therefore, in this study, the authors investigated a possible growth stimulatory effect and mode of action of lipoprotein(a) [Lp(a)] in human mesangial cells HMC, and the effect of Lp(a) on adhesion and migration in human mesangial cells. METHODS: The DNA synthesis of HMC was measured by (3)H-thymidine incorporation. The cell adhesion was detected by the expression of vinculin by means of indirect immunofluorescence. The cell migration was observed under the microscope. RESULTS: The incubation of HMC with Lp(a) for 24 hours induced a significant dose-dependent proliferation of HMC [Lp(a): 5 microg, 10 microg, 25 microg, 50 microg/ml vs. control 0 microg/ml; (3)H-TdR incorporation (x 10(3)cpm): 1.69 +/- 0.48, 3.59 +/- 0.68, 4.14 +/- 0.78, 4.05 +/- 0.55 vs. 1.64 +/- 0.31, P < 0.01]. The vinculin staining by indirect immunofluorescence showed positive result when HMC was incubated with 10 microg/ml Lp(a) for 24 hours, while vinculin was negative when HMC was incubated with 0 microg/ml Lp(a) as the control of the study. The incubation of HMC with 10 microg/ml Lp(a) for 72 hours demonstrated significant cell migration effect compared to the control of 0 microg/ml. (16.2/LP vs. 2.4/LP, P < 0.01). CONCLUSION: Lp(a) could stimulate a proliferation, adhesion and migration effect on human mesangial cells.


Assuntos
Adesão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Lipoproteína(a)/farmacologia , Células Mesangiais/efeitos dos fármacos , Humanos
18.
Biochim Biophys Acta ; 1642(1-2): 25-31, 2003 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-12972290

RESUMO

The association of elevated lipoprotein (a) (Lp(a)) with an increased risk for coronary events is clearly established. This increased risk may in part be due to the activation of monocytes as major cells involved in atherogenesis. High concentrations of plasma Lp(a) were shown to influence the gene expression of human blood monocytes and in the present study we demonstrate a reduced abundance of the lysosomal acid lipase (LAL) mRNA in monocytes of patients with coronary disease and selective Lp(a) hyperlipidemia. This is also supported by in vitro studies where purified Lp(a) but not low-density lipoprotein (LDL) was shown to downregulate mRNA levels of the LAL in control monocytes. A correlation of Lp(a) serum levels and the proinflammatory cytokine IL-6 was recently also described. Therefore, we investigated whether Lp(a) is capable to enhance the release of this acute phase cytokine from human blood monocytes. Purified Lp(a) led to an increased secretion of IL-6, but not TNF-alpha arguing against a general activation of these cells. The association of reduced LAL activity with the premature development of coronary artery disease has been demonstrated in patients with hypercholesterolemia, and in the present study we show for the first time that LAL expression is suppressed in monocytes from patients with Lp(a) hyperlipidemia and by purified Lp(a). In addition, increased levels of IL-6 also predict future cardiovascular events and IL-6 secretion was also induced by purified Lp(a).


Assuntos
Regulação para Baixo/imunologia , Predisposição Genética para Doença/genética , Interleucina-6/metabolismo , Lipase/metabolismo , Lipoproteína(a)/metabolismo , Lisossomos/metabolismo , Monócitos/enzimologia , Células Cultivadas , Doença da Artéria Coronariana/enzimologia , Doença da Artéria Coronariana/genética , Regulação para Baixo/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/genética , Humanos , Hiperlipidemias/enzimologia , Hiperlipidemias/genética , Lipase/genética , Lipoproteína(a)/farmacologia , Monócitos/efeitos dos fármacos , Monócitos/imunologia , Análise de Sequência com Séries de Oligonucleotídeos , Valor Preditivo dos Testes , RNA Mensageiro/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
19.
Arterioscler Thromb Vasc Biol ; 23(7): 1283-8, 2003 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-12775579

RESUMO

OBJECTIVE: Plasma high-density lipoprotein (HDL) level is inversely correlated with the risk of atherosclerosis. However, the cellular mechanism by which HDL exerts antiatherogenic actions is not well understood. In this study, we focus on the lipid components of HDL as mediators of the lipoprotein-induced antiatherogenic actions. METHODS AND RESULTS: HDL and sphingosine 1-phosphate (S1P) stimulated the migration and survival of human umbilical vein endothelial cells. These responses to HDL and S1P were almost completely inhibited by pertussis toxin and other specific inhibitors for intracellular signaling pathways, although the inhibition profiles of migration and survival were different. The HDL-stimulated migration and survival of the cells were markedly inhibited by antisense oligonucleotides against the S1P receptors EDG-1/S1P1 and EDG-3/S1P3. Cell migration was sensitive to both receptors, but cell survival was exclusively sensitive to S1P1. The S1P-rich fraction and chromatographically purified S1P from HDL stimulated cell migration, but the rest of the fraction did not, as was the case of the cell survival. CONCLUSIONS: HDL-induced endothelial cell migration and survival may be mediated by the lipoprotein component S1P and the lipid receptors S1P1 and S1P3.


Assuntos
Movimento Celular/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Lipoproteínas HDL/farmacologia , Esfingosina/análogos & derivados , Esfingosina/farmacologia , Adesão Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Células Cultivadas , Relação Dose-Resposta a Droga , Células Endoteliais/fisiologia , Humanos , Lipoproteína(a)/farmacologia , Lipoproteínas LDL/farmacologia , Lisofosfolipídeos/farmacologia , Oligonucleotídeos Antissenso/farmacologia , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Lisofosfolipídeos , Transdução de Sinais/efeitos dos fármacos
20.
Thromb Res ; 112(5-6): 321-4, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-15041277

RESUMO

Lipoproteins are known to influence platelet cyclic adenosine monophosphate (c-AMP) levels. Lipoprotein (a) (Lp(a))'s impact on platelet c-AMP levels has never been assessed. Increasing levels of purified human Lp(a) (1-100) mg/dl were incubated with washed human platelets. Lp(a) concentrations of 1-25 mg/dl resulted an initial statistically significant increase of platelet c-AMP above basal levels and decreased collagen-stimulated platelet aggregation levels. Higher concentrations progressively returned the platelet c-AMP concentrations to basal levels accompanied by further decreases in platelet aggregation. Increasing concentrations of purified apolipoprotein (a) (apo(a)) also resulted in a similar biphasic c-AMP response while Lp(a) without apo(a) was without impact. One antibody directed against apo(a) in intact Lp(a) removed the biphasic c-AMP pattern and eliminated Lp(a) platelet aggregation. Antibodies directed against apo B in intact Lp(a) gave results similar to intact Lp(a) in terms of the biphasic response of c-AMP upon platelet exposure to increasing levels of Lp(a). It is concluded that apo(a) mediates the Lp(a)-induced biphasic response in platelet c-AMP as the result of platelet exposure to increasing levels of Lp(a). The biphasic response in c-AMP assists in platelet aggregation decreases up to a concentration of 25 mg/dl Lp(a), such assistance being lost at higher Lp(a) concentrations.


Assuntos
Apolipoproteínas A/farmacologia , Plaquetas/química , AMP Cíclico/análise , Lipoproteína(a)/farmacologia , Anticorpos Monoclonais/farmacologia , Apolipoproteínas A/imunologia , Colágeno/farmacologia , Relação Dose-Resposta a Droga , Humanos , Agregação Plaquetária/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...