Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cardiovasc Res ; 119(2): 450-464, 2023 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-35576489

RESUMO

AIMS: The adenylate cyclase type 9 (ADCY9) gene appears to determine atherosclerotic outcomes in patients treated with dalcetrapib. In mice, we recently demonstrated that Adcy9 inactivation potentiates endothelial function and inhibits atherogenesis. The objective of this study was to characterize the contribution of ADCY9 to the regulation of endothelial signalling pathways involved in atherosclerosis. METHODS AND RESULTS: We show that ADCY9 is expressed in the endothelium of mouse aorta and femoral arteries. We demonstrate that ADCY9 inactivation in cultured endothelial cells paradoxically increases cAMP accumulation in response to the adenylate cyclase activators forskolin and vasoactive intestinal peptide (VIP). Reciprocally, ADCY9 overexpression decreases cAMP production. Using mouse femoral artery arteriography, we show that Adcy9 inactivation potentiates VIP-induced endothelial-dependent vasodilation. Moreover, Adcy9 inactivation reduces mouse atheroma endothelial permeability in different vascular beds. ADCY9 overexpression reduces forskolin-induced phosphorylation of Ser157-vasodilator-stimulated phosphoprotein (VASP) and worsens thrombin-induced fall of RAP1 activity, both leading to increased endothelial permeability. ADCY9 inactivation in thrombin-stimulated human coronary artery endothelial cells results in cAMP accumulation, increases p-Ser157-VASP, and inhibits endothelial permeability. MLC2 phosphorylation and actin stress fibre increases in response to thrombin were reduced by ADCY9 inactivation, suggesting actin cytoskeleton regulation. Finally, using the Miles assay, we demonstrate that Adcy9 regulates thrombin-induced endothelial permeability in vivo in normal and atherosclerotic animals. CONCLUSION: Adcy9 is expressed in endothelial cells and regulates local cAMP and endothelial functions including permeability relevant to atherogenesis.


Assuntos
Adenilil Ciclases , Aterosclerose , Animais , Humanos , Camundongos , Adenilil Ciclases/genética , Adenilil Ciclases/metabolismo , Aterosclerose/genética , Aterosclerose/prevenção & controle , Aterosclerose/metabolismo , Colforsina/farmacologia , Colforsina/metabolismo , Células Endoteliais/metabolismo , Endotélio/metabolismo , Trombina/metabolismo , AMP Cíclico/metabolismo
2.
Chemphyschem ; 21(9): 863-869, 2020 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-32092218

RESUMO

Paramagnetic restraints have been used in biomolecular NMR for the last three decades to elucidate and refine biomolecular structures, but also to characterize protein-ligand interactions. A common technique to generate such restraints in proteins, which do not naturally contain a (paramagnetic) metal, consists in the attachment to the protein of a lanthanide-binding-tag (LBT). In order to design such LBTs, it is important to consider the efficiency and stability of the conjugation, the geometry of the complex (conformational exchanges and coordination) and the chemical inertness of the ligand. Here we describe a photo-catalyzed thiol-ene reaction for the cysteine-selective paramagnetic tagging of proteins. As a model, we designed an LBT with a vinyl-pyridine moiety which was used to attach our tag to the protein GB1 in fast and irreversible fashion. Our tag T1 yields magnetic susceptibility tensors of significant size with different lanthanides and has been characterized using NMR and relaxometry measurements.


Assuntos
Proteínas/química , Compostos de Sulfidrila/química , Catálise , Cisteína/química , Elementos da Série dos Lantanídeos/química , Ligantes , Espectroscopia de Ressonância Magnética/métodos , Ressonância Magnética Nuclear Biomolecular/métodos , Processos Fotoquímicos , Piridinas/química
3.
Angiology ; 67(2): 157-67, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25904765

RESUMO

BACKGROUND: Given the link between cholesterol and activation of inflammation via interleukin 1ß (IL-1ß), we tested the effects of IL-1ß inhibition on atherosclerotic calcification in mice. Patients with familial hypercholesterolemia develop extensive aortic calcification and calcific aortic stenosis. Although statins delay this process, low-density lipoprotein (LDL) cholesterol lowering alone is not enough to avert it. Data suggest that vascular inflammation initiated by hypercholesterolemia is followed by unchecked mineralization at sites of atherosclerotic plaques. The LDL-receptor (LDLR)-deficient (Ldlr(-/-)) and LDLR-attenuated Pcsk9(Tg) mice are available animal models for pharmacological testing. METHODS: A mouse monoclonal antibody (mAb) against IL-1ß or placebo was administered subcutaneously in Ldlr(-/-) and Pcsk9(Tg) models fed a Western diet. Drug level, anthropometric, lipid, and glucose profiles were determined. Expressions of proprotein convertase subtilisin/kexin type 9 (PCSK9), serum amyloid A1, and cytokine were measured by enzyme-linked immunosorbent assay. Aortic calcification was determined by microcomputerized tomography (micro-CT) and X-ray densitometry, and aortic flow velocity was assessed by ultrasound. RESULTS: Circulating levels of IL-1ß in Ldlr(-/-) mice were significantly greater (2-fold) than observed in Pcsk9(Tg) mice. Placebo- and mAb-treated mice did not differ in their growth, lipid, glucose profiles, and other cytokines. Calcifications were significantly diminished in mAb-treatment Ldlr(-/-) mice (a reduction of ∼ 75% by X-ray and ∼ 90% by micro-CT) and reduced insignificantly in mAb-treatment Pcsk9(Tg) mice, whereas aortic flow velocity was unchanged in both models. CONCLUSIONS: Herein, we demonstrate that aortic calcifications can be inhibited by an IL-1ß mAb in LDLR-deficient mice. These results have a translational component to prevent vascular calcification in human and represent new evidence to rationalize targeting inflammation in cardiovascular disease.


Assuntos
Anti-Inflamatórios/farmacologia , Anticorpos Monoclonais/farmacologia , Aorta/efeitos dos fármacos , Doenças da Aorta/prevenção & controle , Hiperlipoproteinemia Tipo II/tratamento farmacológico , Interleucina-1beta/antagonistas & inibidores , Calcificação Vascular/prevenção & controle , Animais , Aorta/diagnóstico por imagem , Aorta/metabolismo , Aorta/fisiopatologia , Doenças da Aorta/sangue , Doenças da Aorta/diagnóstico , Doenças da Aorta/genética , Doenças da Aorta/imunologia , Doenças da Aorta/fisiopatologia , Aortografia/métodos , Biomarcadores/sangue , Velocidade do Fluxo Sanguíneo , Modelos Animais de Doenças , Predisposição Genética para Doença , Hiperlipoproteinemia Tipo II/sangue , Hiperlipoproteinemia Tipo II/genética , Hiperlipoproteinemia Tipo II/imunologia , Interleucina-1beta/sangue , Interleucina-1beta/imunologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Pró-Proteína Convertases/genética , Pró-Proteína Convertases/metabolismo , Receptores de LDL/deficiência , Receptores de LDL/genética , Fluxo Sanguíneo Regional , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo , Ultrassonografia , Calcificação Vascular/diagnóstico , Calcificação Vascular/genética , Calcificação Vascular/imunologia , Calcificação Vascular/metabolismo , Calcificação Vascular/fisiopatologia , Microtomografia por Raio-X
4.
PLoS One ; 7(7): e41865, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22848640

RESUMO

Proprotein convertase subtilisin/kexin-9 (PCSK9) enhances the degradation of hepatic low-density lipoprotein receptor (LDLR). Deletion of PCSK9, and loss-of-function mutants in humans result in lower levels of circulating LDL-cholesterol and a strong protection against coronary heart disease. Accordingly, the quest for PCSK9 inhibitors has major clinical implications. We have previously identified annexin A2 (AnxA2) as an endogenous binding partner and functional inhibitor of PCSK9. Herein, we studied the relevance of AnxA2 in PCSK9 inhibition and lipid metabolism in vivo. Plasma analyses of AnxA2(-/-) mice revealed: i) a ∼1.4-fold increase in LDL-cholesterol without significant changes in VLDLs or HDLs, and ii) a ∼2-fold increase in circulating PCSK9 levels. Western blotting and immunohistochemistry of AnxA2(-/-) tissues revealed that the LDLR was decreased by ∼50% in extrahepatic tissues, such as adrenals and colon. We also show that AnxA2-derived synthetic peptides block the PCSK9≡LDLR interaction in vitro, and adenoviral overexpression of AnxA2 in mouse liver increases LDLR protein levels in vivo. These results suggest that AnxA2 acts as an endogenous regulator of LDLR degradation, mostly in extrahepatic tissues. Finally, we identified an AnxA2 coding polymorphism, V98L, that correlates with lower circulating levels of PCSK9 thereby extending our results on the physiological role of AnxA2 in humans.


Assuntos
Anexina A2/metabolismo , Fígado/metabolismo , Pró-Proteína Convertases/metabolismo , Proteólise , Receptores de LDL/metabolismo , Serina Endopeptidases/metabolismo , Adenoviridae/genética , Sequência de Aminoácidos , Animais , Anexina A2/química , Anexina A2/deficiência , Anexina A2/genética , Linhagem Celular , LDL-Colesterol/sangue , Cricetinae , Éxons/genética , Espaço Extracelular/metabolismo , Humanos , Fígado/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Polimorfismo de Nucleotídeo Único , Pró-Proteína Convertase 9 , Pró-Proteína Convertases/sangue , Estrutura Terciária de Proteína , Serina Endopeptidases/sangue
5.
J Biol Chem ; 286(5): 3370-8, 2011 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-21106522

RESUMO

Macrophage foam cell is the predominant cell type in atherosclerotic lesions. Removal of excess cholesterol from macrophages thus offers effective protection against atherosclerosis. Here we report that a protein kinase A (PKA)-anchoring inhibitor, st-Ht31, induces robust cholesterol/phospholipid efflux, and ATP-binding cassette transporter A1 (ABCA1) greatly facilitates this process. Remarkably, we found that st-Ht31 completely reverses foam cell formation, and this process is ABCA1-dependent. The reversal is also accompanied by the restoration of well modulated inflammatory response to LPS. There is no detectable toxicity associated with st-Ht31, even when cells export up to 20% cellular cholesterol per hour. Using FRET-based PKA biosensors in live cells, we provide evidence that st-Ht31 drives cholesterol efflux by elevating PKA activity specifically in the cytoplasm. Furthermore, ABCA1 facilitates st-Ht31 uptake. This allows st-Ht31 to effectively remove cholesterol from ABCA1-expressing cells. We speculate that de-anchoring of PKA offers a novel therapeutic strategy to remove excess cholesterol from lipid-laden lesion macrophages.


Assuntos
Transportadores de Cassetes de Ligação de ATP/fisiologia , Colesterol/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Células Espumosas/efeitos dos fármacos , Proteínas/farmacologia , Transportador 1 de Cassete de Ligação de ATP , Animais , Transporte Biológico/efeitos dos fármacos , Linhagem Celular , Células Espumosas/citologia , Macrófagos/citologia , Camundongos
6.
J Lipid Res ; 50(3): 456-466, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18941142

RESUMO

ATP-binding cassette transporter A1 (ABCA1) mediates cholesterol efflux to lipid-poor apolipoprotein A-I (apoA-I) and generates HDL. Here, we demonstrate that ABCA1 also directly mediates the production of apoA-I free microparticles. In baby hamster kidney (BHK) cells and RAW macrophages, ABCA1 expression led to lipid efflux in the absence of apoA-I and released large microparticles devoid of apoB and apoE. We provide evidence that these microparticles are an integral component of the classical cholesterol efflux pathway when apoA-I is present and accounted for approximately 30% of the total cholesterol released to the medium. Furthermore, microparticle release required similar ABCA1 activities as was required for HDL production. For instance, a nucleotide binding domain mutation in ABCA1 (A937V) that impaired HDL generation also abolished microparticle release. Similarly, inhibition of protein kinase A (PKA) prevented the release of both types of particles. Interestingly, physical modulation of membrane dynamics affected HDL and microparticle production, rigidifying the plasma membrane with wheat germ agglutinin inhibited HDL and microparticle release, whereas increasing the fluidity promoted the production of these particles. Given the established role of ABCA1 in expending nonraft or more fluid-like membrane domains, our results suggest that both HDL and microparticle release is favored by a more fluid plasma membrane. We speculate that ABCA1 enhances the dynamic movement of the plasma membrane, which is required for apoA-I lipidation and microparticle formation.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Micropartículas Derivadas de Células/metabolismo , Colesterol/metabolismo , Lipoproteínas HDL/metabolismo , Transportador 1 de Cassete de Ligação de ATP , Animais , Apolipoproteína A-I/metabolismo , Transporte Biológico Ativo/efeitos dos fármacos , Linhagem Celular , Micropartículas Derivadas de Células/efeitos dos fármacos , Cricetinae , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Humanos , Rim/citologia , Rim/efeitos dos fármacos , Rim/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Fluidez de Membrana/efeitos dos fármacos , Fluidez de Membrana/fisiologia , Camundongos , Mifepristona/farmacologia , Proteínas Recombinantes/metabolismo , Aglutininas do Germe de Trigo/farmacologia
7.
J Biol Chem ; 281(47): 36091-101, 2006 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-16984907

RESUMO

ATP-binding cassette transporter A1 (ABCA1) is known to mediate cholesterol efflux to lipid-poor apolipoprotein A-I. In addition, ABCA1 has been shown to influence functions of the plasma membrane, such as endocytosis and phagocytosis. Here, we report that ABCA1 expression results in a significant redistribution of cholesterol and sphingomyelin from rafts to non-rafts. Caveolin, a raft/caveolae marker also redistributes from punctate caveolae-like structures to the general area of the plasma membrane upon ABCA1 expression. Furthermore, we observed significant reduction of Akt activation in ABCA1-expressing cells, consistent with raft disruption. Cholesterol content in the plasma membrane is, however, not altered. Moreover, we provide evidence that a non-functional ABCA1 with mutation in an ATP-binding domain, A937V, fails to redistribute cholesterol, sphingomyelin, or caveolin. A937V also fails to influence Akt activation. Finally, we show that apolipoprotein A-I preferentially associates with non-raft membranes in ABCA1-expressing cells. Our results thus demonstrate that ABCA1 causes a change in overall lipid packing of the plasma membrane, likely through its ATPase-related functions. Such reorganization by ABCA1 effectively expands the non-raft membrane fractions and, consequentially, pre-conditions cells for cholesterol efflux.


Assuntos
Transportadores de Cassetes de Ligação de ATP/fisiologia , Microdomínios da Membrana/química , Transportador 1 de Cassete de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/química , Animais , Caveolina 1/biossíntese , Membrana Celular/metabolismo , Colesterol/metabolismo , Cricetinae , Endocitose , Proteínas de Membrana Transportadoras/química , Microscopia de Fluorescência , Fosfolipídeos/química , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transferrina/metabolismo
8.
Mol Biol Cell ; 17(4): 1593-605, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16452637

RESUMO

Although cholesterol is synthesized in the endoplasmic reticulum (ER), compared with other cellular membranes, ER membrane has low cholesterol (3-6%). Most of the molecular machinery that regulates cellular cholesterol homeostasis also resides in the ER. Little is known about how cholesterol itself affects the ER membrane. Here, we demonstrate that acute cholesterol depletion in ER membranes impairs ER-to-Golgi transport of secretory membrane proteins. Cholesterol depletion is achieved by a brief inhibition of cholesterol synthesis with statins in cells grown in cholesterol-depleted medium. We provide evidence that secretory membrane proteins vesicular stomatitis virus glycoprotein and scavenger receptor A failed to be efficiently transported from the ER upon cholesterol depletion. Fluorescence photobleaching recovery experiments indicated that cholesterol depletion by statins leads to a severe loss of lateral mobility on the ER membrane of these transmembrane proteins, but not loss of mobility of proteins in the ER lumen. This impaired lateral mobility is correlated with impaired ER-to-Golgi transport. These results provide evidence for the first time that cholesterol is required in the ER membrane to maintain mobility of membrane proteins and thus protein secretion.


Assuntos
Colesterol/deficiência , Retículo Endoplasmático/metabolismo , Complexo de Golgi/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Células CHO , Cricetinae , Cricetulus , Retículo Endoplasmático/efeitos dos fármacos , Recuperação de Fluorescência Após Fotodegradação , Complexo de Golgi/ultraestrutura , Hidroximetilglutaril-CoA Redutases/efeitos dos fármacos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Glicoproteínas de Membrana/metabolismo , Ácido Mevalônico/farmacologia , Transporte Proteico , Vesículas Secretórias/efeitos dos fármacos , Vesículas Secretórias/metabolismo , Proteínas do Envelope Viral/metabolismo
9.
J Lipid Res ; 46(7): 1457-65, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15654121

RESUMO

It has been suggested that ABCA1 interacts preferentially with lipid-poor apolipoprotein A-I (apoA-I). Here, we show that treatment of plasma with dimyristoyl phosphatidylcholine (DMPC) multilamellar vesicles generates prebeta(1)-apoA-I-containing lipoproteins (LpA-I)-like particles similar to those of native plasma. Isolated prebeta(1)-LpA-I-like particles inhibited the binding of (125)I-apoA-I to ABCA1 more efficiently than HDL(3) (IC(50) = 2.20 +/- 0.35 vs. 37.60 +/- 4.78 microg/ml). We next investigated the ability of DMPC-treated plasma to promote phospholipid and unesterified (free) cholesterol efflux from J774 macrophages stimulated or not with cAMP. At 2 mg DMPC/ml plasma, both phospholipid and free cholesterol efflux were increased ( approximately 50% and 40%, respectively) in cAMP-stimulated cells compared with unstimulated cells. Similarly, both phospholipid and free cholesterol efflux to either isolated native prebeta(1)-LpA-I and prebeta(1)-LpA-I-like particles were increased significantly in stimulated cells. Furthermore, glyburide significantly inhibited phospholipid and free cholesterol efflux to DMPC-treated plasma. Removal of apoA-I-containing lipoproteins from normolipidemic plasma drastically reduced free cholesterol efflux mediated by DMPC-treated plasma. Finally, treatment of Tangier disease plasma with DMPC affected the amount of neither prebeta(1)-LpA-I nor free cholesterol efflux. These results indicate that DMPC enrichment of normal plasma resulted in the redistribution of apoA-I from alpha-HDL to prebeta-HDL, allowing for more efficient ABCA1-mediated cellular lipid release. Increasing the plasma prebeta(1)-LpA-I level by either pharmacological agents or direct infusions might prevent foam cell formation and reduce atherosclerotic vascular disease.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Colesterol/metabolismo , Lipoproteínas HDL/sangue , Lipoproteínas HDL/química , Fosfolipídeos/química , Transportador 1 de Cassete de Ligação de ATP , Animais , Apolipoproteína A-I/sangue , Apolipoproteína A-I/efeitos dos fármacos , Células Cultivadas , Dimiristoilfosfatidilcolina/química , Humanos , Lipoproteínas HDL3 , Lipossomos/química , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Camundongos , Doença de Tangier/sangue
10.
Circulation ; 110(18): 2881-8, 2004 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-15492319

RESUMO

BACKGROUND: Of the cells that compose the atherosclerotic plaque, vascular endothelial cells are the most resistant to cholesterol accumulation. Cholesterol efflux pathways may play an important role in endothelial cholesterol homeostasis. METHODS AND RESULTS: We examined the global genetic response of endothelial cells to cholesterol and in particular the contribution of the cholesterol efflux proteins ATP-binding cassette transporter A1 (ABCA1), ATP-binding cassette transporter G1 (ABCG1), and scavenger receptor B-I (SR-BI) to endothelial cell cholesterol efflux. The ABCG1 gene is induced in endothelial cells by cholesterol, whereas ABCA1 is not. Using specific chemical inhibitors of ABC transporters and SR-BI, we have shown that neither ABC transporters nor SR-BI is required for apolipoprotein A-1-mediated endothelial cholesterol efflux. CONCLUSIONS: Endothelial cells may use nontraditional pathways for cholesterol efflux.


Assuntos
Colesterol/metabolismo , Células Endoteliais/metabolismo , Endotélio Vascular/fisiologia , 8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , 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/antagonistas & inibidores , Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Aorta , Apolipoproteína A-I/farmacologia , Antígenos CD36 , Células COS , Bovinos , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/metabolismo , Chlorocebus aethiops , Colesterol/farmacologia , Células Endoteliais/efeitos dos fármacos , Endotélio Vascular/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/patologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Glibureto/farmacologia , Humanos , Hidroxicolesteróis/farmacologia , Lipoproteínas HDL/farmacologia , Lipoproteínas HDL3 , Análise de Sequência com Séries de Oligonucleotídeos , Receptores Imunológicos/antagonistas & inibidores , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo , Receptores Depuradores , Proteínas Recombinantes de Fusão/fisiologia , Soroalbumina Bovina/farmacologia , Doença de Tangier/genética , Doença de Tangier/patologia , Transfecção , Veias Umbilicais
12.
Mol Genet Metab ; 78(4): 265-74, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12706378

RESUMO

Mutations in the ATP-binding cassette transporter A1 (ABCA1) gene cause familial high-density lipoprotein deficiency and Tangier disease. ABCA1 plays a crucial role in active apolipoprotein A-I (apoA-I) lipidation, a key step in reverse cholesterol transport. We compared ABCA1 transcriptional regulation and cholesterol efflux in human skin fibroblasts, monocyte-derived macrophages and hepatocytes (HepG2). 8-Br-cAMP did not increase ABCA1 transcription in these tissues compared to mouse macrophages. We found that ABCA1 is differentially regulated among tissues. While transcription in HepG2 appears to be constitutive, sterols stimulate ABCA1 transcription in fibroblasts and monocyte-derived macrophages. ApoA-I promoted cholesterol efflux in fibroblasts, macrophages, and HepG2. Cholesterol homeostasis in fibroblasts is tightly regulated, and ABCA1 mRNA closely follows the cellular mass of free cholesterol (dose- and time-dependent manner). To further determine the mechanism used by fibroblasts to maintain sterol balance, we used a competitive inhibition approach with geranylgeranyl pyrophosphate (GGPP) to block the LXR induction pathway. GGPP blocked basal, 22-(R)-hydroxycholesterol- and cholesterol-induced ABCA1 expression. Taken together, these results demonstrate that: (1) ABCA1 expression varies among tissues, and (2) cholesterol conversion to hydroxycholesterol is an important mechanism for the maintenance of cholesterol homeostasis in fibroblasts.


Assuntos
Transportadores de Cassetes de Ligação de ATP/biossíntese , Transportadores de Cassetes de Ligação de ATP/metabolismo , 8-Bromo Monofosfato de Adenosina Cíclica/metabolismo , Transportador 1 de Cassete de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/genética , Transporte Biológico , Northern Blotting , Linhagem Celular , Colesterol/metabolismo , Relação Dose-Resposta a Droga , Regulação para Baixo , Fibroblastos/metabolismo , Regulação da Expressão Gênica , Humanos , Immunoblotting , Macrófagos/metabolismo , Monócitos/metabolismo , Fosfatos de Poli-Isoprenil/metabolismo , RNA Mensageiro/metabolismo , Fatores de Tempo , Transcrição Gênica
13.
J Lipid Res ; 43(12): 2087-94, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12454270

RESUMO

ATP-binding cassette transporter A1 (ABCA1) plays a crucial role in apoA-I lipidation, a key step in reverse cholesterol transport. cAMP induces apoA-I binding activity and promotes cellular cholesterol efflux. We investigated the role of the cAMP/protein kinase A (PKA) dependent pathway in the regulation of cellular cholesterol efflux. Treatment of normal fibroblasts with 8-bromo-cAMP (8-Br-cAMP) increased significantly apoA-I-mediated cholesterol efflux, with specificity for apoA-I, but not for cyclodextrin. Concomitantly, 8-Br-cAMP increased ABCA1 phosphorylation in a time-dependent manner. Maximum phosphorylation was reached in <10 min, representing a 260% increase compared to basal ABCA1 phosphorylation level. Forskolin, a known cAMP regulator, increased both cellular cholesterol efflux and ABCA1 phosphorylation. In contrast, H-89 PKA inhibitor reduced cellular cholesterol efflux by 70% in a dose-dependent manner and inhibited almost completely ABCA1 phosphorylation. To determine whether naturally occurring mutants of ABCA1 may affect its phosphorylation activity, fibroblasts from subjects with familial HDL deficiency (FHD, heterozygous ABCA1 defect) and Tangier disease (TD, homozygous/compound heterozygous ABCA1 defect) were treated with 8-Br-cAMP or forskolin. Cellular cholesterol efflux and ABCA1 phosphorylation were increased in FHD but not in TD cells. Taken together, these findings provide evidence for a link between the cAMP/PKA-dependent pathway, ABCA1 phosphorylation, and apoA-I mediated cellular cholesterol efflux.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Colesterol/metabolismo , AMP Cíclico/metabolismo , Fibroblastos/metabolismo , Transportador 1 de Cassete de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/biossíntese , Transportadores de Cassetes de Ligação de ATP/genética , Apolipoproteína A-I/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Humanos , Mutação , Fosforilação , RNA Mensageiro/metabolismo , Transdução de Sinais/fisiologia , Doença de Tangier/genética , Doença de Tangier/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA