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.
J Nutr Biochem ; 89: 108564, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33321184

RESUMO

Scavenger receptor BI (SR-BI) has been suggested to modulate adipocyte function. To uncover the potential relevance of SR-BI for the development of obesity and associated metabolic complications, we compared the metabolic phenotype of wild-type and SR-BI deficient mice fed an obesogenic diet enriched in fat. Both male and female SR-BI knockout mice gained significantly more weight as compared to their wild-type counterparts in response to 12 weeks high fat diet feeding (1.5-fold; P < .01 for genotype). Plasma free cholesterol levels were ~2-fold higher (P < .001) in SR-BI knockout mice of both genders, whilst plasma cholesteryl ester and triglyceride concentrations were only significantly elevated in males. Strikingly, the exacerbated obesity in SR-BI knockout mice was paralleled by a better glucose handling. In contrast, only SR-BI knockout mice developed atherosclerotic lesions in the aortic root, with a higher predisposition in females. Biochemical and histological studies in male mice revealed that SR-BI deficiency was associated with a reduced hepatic steatosis degree as evident from the 29% lower (P < .05) liver triglyceride levels. Relative mRNA expression levels of the glucose uptake transporter GLUT4 were increased (+47%; P < .05), whilst expression levels of the metabolic PPARgamma target genes CD36, HSL, ADIPOQ and ATGL were reduced 39%-58% (P < .01) in the context of unchanged PPARgamma expression levels in SR-BI knockout gonadal white adipose tissue. In conclusion, we have shown that SR-BI deficiency is associated with a decrease in adipocyte PPARgamma activity and a concomitant uncoupling of obesity development from hepatic steatosis and glucose intolerance development in high fat diet-fed mice.


Assuntos
Antígenos CD36/deficiência , Fígado Gorduroso/metabolismo , Intolerância à Glucose/metabolismo , Obesidade/metabolismo , Adipócitos/metabolismo , Tecido Adiposo Branco/metabolismo , Animais , Aterosclerose/metabolismo , Antígenos CD36/metabolismo , Colesterol/sangue , Ésteres do Colesterol/sangue , Dieta Hiperlipídica/efeitos adversos , Feminino , Humanos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , PPAR gama/genética , PPAR gama/metabolismo , Receptores Depuradores Classe B/metabolismo , Triglicerídeos/sangue
2.
Biochim Biophys Acta Mol Basis Dis ; 1865(6): 1402-1409, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-30776415

RESUMO

The nuclear receptor liver X receptor (LXR) impacts on cholesterol metabolism as well as hepatic lipogenesis via transcriptional regulation. It is proposed that inhibition of the protein arginine methyltransferase 3 (PRMT3) uncouples these two transcriptional pathways in vivo by acting as a specific lipogenic coactivator of LXR. Here we validated the hypothesis that treatment with the allosteric PRMT3 inhibitor SGC707 will diminish the hepatic steatosis extent, while leaving global cholesterol metabolism, important in cholesterol-driven pathologies like atherosclerosis, untouched. For this purpose, 12-week old hyperlipidemic apolipoprotein E knockout mice were fed a Western-type diet for six weeks to induce both hepatic steatosis and atherosclerosis. The mice received 3 intraperitoneal injections with SGC707 or solvent control per week. Mice chronically treated with SGC707 developed less severe hepatic steatosis as exemplified by the 51% reduced (P < 0.05) liver triglyceride levels. In contrast, the extent of in vivo macrophage foam cell formation and aortic root atherosclerosis was not affected by SGC707 treatment. Interestingly, SGC707-treated mice gained 94% less body weight (P < 0.05), which was paralleled by changes in white adipose tissue morphology, i.e. reduction in adipocyte size and browning. In conclusion, we have shown that through PRMT3 inhibitor treatment specific functions of LXR involved in respectively the development of fatty liver disease and atherosclerosis can be uncoupled, resulting in an overall diminished hepatic steatosis extent without a negative impact on atherosclerosis susceptibility. As such, our studies highlight that PRMT3 inhibition may constitute a novel therapeutic approach to limit the development of fatty liver disease in humans.


Assuntos
Apolipoproteínas E/genética , Aterosclerose/genética , Inibidores Enzimáticos/farmacologia , Fígado Gorduroso/prevenção & controle , Isoquinolinas/farmacologia , Proteína-Arginina N-Metiltransferases/genética , Tecido Adiposo Branco/efeitos dos fármacos , Tecido Adiposo Branco/enzimologia , Tecido Adiposo Branco/patologia , Regulação Alostérica/efeitos dos fármacos , Animais , Aorta/efeitos dos fármacos , Aorta/metabolismo , Aorta/patologia , Apolipoproteínas E/deficiência , Aterosclerose/enzimologia , Aterosclerose/etiologia , Aterosclerose/patologia , Colesterol/metabolismo , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Suscetibilidade a Doenças , Fígado Gorduroso/etiologia , Fígado Gorduroso/genética , Fígado Gorduroso/patologia , Células Espumosas/efeitos dos fármacos , Células Espumosas/metabolismo , Células Espumosas/patologia , Regulação da Expressão Gênica , Fígado/efeitos dos fármacos , Fígado/enzimologia , Fígado/patologia , Receptores X do Fígado/genética , Receptores X do Fígado/metabolismo , Masculino , Camundongos , Camundongos Knockout para ApoE , Proteína-Arginina N-Metiltransferases/antagonistas & inibidores , Proteína-Arginina N-Metiltransferases/metabolismo , Transdução de Sinais , Triglicerídeos/metabolismo
3.
Atherosclerosis ; 281: 47-55, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30658191

RESUMO

BACKGROUND AND AIMS: Stabilin-1 (STAB1) is a scavenger receptor expressed on alternatively activated macrophages and sinusoidal endothelial cells. Its ligands include oxidized low-density lipoprotein (LDL) and the extracellular matrix glycoprotein SPARC and it is present in both human and murine atherosclerotic lesions. We aimed to investigate the effect of specific deletion of STAB1 in bone marrow-derived cells, including macrophages on atherosclerotic lesion formation in mice. METHODS: Lethally irradiated hypercholesterolemic LDL receptor knockout mice received either wildtype (WT) or STAB1 knockout (STAB1 KO) bone marrow. Bone marrow transplanted mice were fed a Western-type diet for 9 weeks to induce atherosclerotic lesion formation. RESULTS: Interestingly, LDL receptor knockout mice reconstituted with STAB1 KO bone marrow showed increased body weight gain (two-way ANOVA: p < 0.001) and larger white adipocyte cell sizes (43% increase in cell area; p < 0.05) as compared to WT bone marrow transplanted mice, which correlated positively (r = 0.82; p < 0.001). This was paralleled by a significant increase in white adipose tissue relative mRNA expression levels of the adipokine leptin (+94% p < 0.05). Despite these changes, no differences in serum lipid levels, the extent of in vivo macrophage foam cell formation or circulating leukocyte concentrations were observed. Moreover, the size and composition of atherosclerotic lesions was not different between the two experimental groups. CONCLUSIONS: Bone marrow-specific Stabilin-1 deletion does not affect the susceptibility for atherosclerosis in mice. However, the increased body weight gain and adipocyte cell size highlight a potential role for leukocyte STAB1 in the development of metabolic disorders.


Assuntos
Doenças da Aorta/metabolismo , Aterosclerose/metabolismo , Moléculas de Adesão Celular Neuronais/deficiência , Células Espumosas/metabolismo , Receptores de LDL/deficiência , Adipócitos Brancos/metabolismo , Adipócitos Brancos/patologia , Tecido Adiposo Branco/metabolismo , Tecido Adiposo Branco/patologia , Animais , Doenças da Aorta/etiologia , Doenças da Aorta/genética , Doenças da Aorta/patologia , Aterosclerose/etiologia , Aterosclerose/genética , Aterosclerose/patologia , Moléculas de Adesão Celular Neuronais/genética , Dieta Ocidental , Modelos Animais de Doenças , Células Espumosas/patologia , Hipercolesterolemia/complicações , Hipercolesterolemia/genética , Leptina/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Placa Aterosclerótica , Receptores de LDL/genética , Aumento de Peso
4.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1864(4): 443-451, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30633988

RESUMO

OBJECTIVE: Since cholesterol is the sole precursor for glucocorticoid synthesis, it is hypothesized that genetic defects in proteins that impact the cellular cholesterol pool may underlie glucocorticoid insufficiency in humans. In the current study, we specifically focused on the cholesterol efflux mediator ATP-binding cassette transporter G1 (ABCG1) as gene candidate. METHODS: The adrenal transcriptional response to fasting stress was measured in wild-type mice to identify putative novel gene candidates. Subsequently, the adrenal glucocorticoid function was compared between ABCG1 knockout mice and wild-type controls. RESULTS: Overnight food deprivation induced a change in relative mRNA expression levels of cholesterol metabolism-related proteins previously linked to steroidogenesis, i.e. scavenger receptor class B type I (+149%; P < 0.001), LDL receptor (-70%; P < 0.001) and apolipoprotein E (-41%; P < 0.01). Strikingly, ABCG1 transcript levels were also markedly decreased (-61%; P < 0.05). In contrast to our hypothesis that decreasing cholesterol efflux would increase the adrenal cholesterol pool and enhance glucocorticoid output, ABCG1 knockout mice as compared to wild-type mice exhibited a reduced ability to secrete corticosterone in response to an ACTH challenge (two-way ANOVA: P < 0.001 for genotype) or fasting stress. As a result, glucocorticoid target gene expression levels in liver and hypothalamus were reduced and blood lymphocyte concentrations and spleen weights increased in ABCG1 knockout mice under fasting stress conditions. This was paralleled by a 48% reduction in adrenal cholesteryl ester stores and stimulation of adrenal NPC intracellular cholesterol transporter 2 (+37%; P < 0.05) and apolipoprotein E (+59%; P < 0.01) mRNA expression. CONCLUSION: ABCG1 deficiency is associated with mild glucocorticoid insufficiency in mice.


Assuntos
Transportador 1 de Cassete de Ligação de ATP/deficiência , Apolipoproteínas E/genética , Glucocorticoides/deficiência , Receptores de LDL/genética , Receptores Depuradores Classe B/genética , Transportador 1 de Cassete de Ligação de ATP/genética , Animais , Ésteres do Colesterol/metabolismo , Modelos Animais de Doenças , Privação de Alimentos , Humanos , Metabolismo dos Lipídeos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
5.
Biochim Biophys Acta Mol Basis Dis ; 1865(2): 494-501, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30448542

RESUMO

OBJECTIVE: Proteoglycan 4 (Prg4) has emerged from human association studies as a possible factor contributing to weight gain, dyslipidemia and insulin resistance. In the current study, we investigated the causal role of Prg4 in controlling lipid and glucose metabolism in mice. METHODS: Prg4 knockout (KO) mice and wild-type (WT) littermates were challenged with an obesogenic high-fat diet (45% of total calories as fat) for 16 weeks. To further stimulate the development of metabolic alterations, 10% fructose water was provided starting from week 13. RESULTS: Prg4 deficiency only tended to reduce diet-induced body weight gain, but significantly improved glucose handling (AUC: -29%; p < 0.05), which was also reflected by a tendency towards a reduced HOMA-IR score (-49%; p = 0.06 as compared to WT mice). This coincided with lower hepatic expression of glycolysis (Gck: -30%; p < 0.05) and lipogenesis (Acc: -21%; p < 0.05 and Scd1: -38%; p < 0.001) genes, which translated in significantly lower hepatic triglyceride levels (-56%; p < 0.001) in Prg4 KO mice as compared to WT mice. Prg4 KO mice likely had lower glucose utilization by skeletal muscle as compared to WT mice, judged by a significant reduction in the genes Glut4 (-29%; p < 0.01), Pfkm (-21%; p < 0.05) and Hk2 (-39%; p < 0.001). Moreover, Prg4 KO mice showed a favorable white adipose tissue phenotype with lower uptake of triglyceride-derived fatty acids (-46%; p < 0.05) and lower gene expression of inflammatory markers Cd68, Mcp1 and Tnfα (-65%, -81% and -63%, respectively; p < 0.01) than WT mice. CONCLUSION: Prg4 KO mice are protected from high-fat diet-induced glucose intolerance and fatty liver disease.


Assuntos
Fígado Gorduroso/complicações , Fígado Gorduroso/prevenção & controle , Intolerância à Glucose/complicações , Intolerância à Glucose/prevenção & controle , Proteoglicanas/deficiência , Tecido Adiposo Branco/metabolismo , Animais , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Dieta Hiperlipídica , Fígado Gorduroso/patologia , Feminino , Glucose/metabolismo , Intolerância à Glucose/patologia , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Obesos , Músculos/metabolismo , Proteoglicanas/metabolismo , Gordura Subcutânea/metabolismo
6.
Atherosclerosis ; 280: 132-139, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30513408

RESUMO

BACKGROUND AND AIMS: The development of atherosclerosis is tightly regulated by the innate and adaptive immune system. Communication between these two compartments occurs, among others, upon presentation of lipid antigens to the NKT cell population by CD1d-expressing antigen-presenting cells. Recent evidence states that also mast cells express CD1d and can directly communicate with NKT cells. However, no such relationship has been reported in atherosclerosis. Here, we aimed to elucidate in vivo the CD1d-mediated interaction between mast cells and NKT cells upon atherosclerosis progression. METHODS: We adoptively transferred CD1d-/- or control mast cells to mast cell-deficient apoE-/-KitW-sh/W-sh mice and subsequently placed the animals on a Western-type diet for 10 weeks. RESULTS: At the end of the Western-type diet period, the aortic root of CD1d-/- mast cell-reconstituted mice displayed increased plaque size, with less collagen deposition and higher intraplaque CD4+ T cells, as compared to control mice. In addition, T cells inside the aortic arch showed higher pro-inflammatory cytokine production in the form of IFNγ, TNFα and IL-17. Finally, T-bet expression was found elevated in both CD4+ and CD8+ circulating T cells. CONCLUSIONS: This study is the first to illustrate that disruption of the CD1d communication pathway between mast cells and NKT cells aggravates atherosclerosis, through a shift towards pro-inflammatory T cell responses. This ability of mast cell action during plaque progression sheds new light on their role in atherosclerosis.


Assuntos
Antígenos CD1d/metabolismo , Aterosclerose/imunologia , Linfócitos T CD4-Positivos/citologia , Células Matadoras Naturais/citologia , Mastócitos/citologia , Células T Matadoras Naturais/citologia , Placa Aterosclerótica/imunologia , Animais , Aorta Torácica/metabolismo , Células da Medula Óssea/citologia , Linfócitos T CD8-Positivos/citologia , Feminino , Inflamação , Interferon gama/imunologia , Interleucina-17/imunologia , Ativação Linfocitária , Mastócitos/metabolismo , Camundongos , Fator de Necrose Tumoral alfa/imunologia
9.
Br J Pharmacol ; 175(15): 3175-3183, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29774529

RESUMO

BACKGROUND AND PURPOSE: Agonists for the liver X receptor (LXR) are considered promising therapeutic moieties in cholesterol-driven diseases by promoting cellular cholesterol efflux pathways. However, current clinical application of these agents is hampered by concomitant LXR-induced activation of a lipogenic transcriptional network, leading to hepatic steatosis. Recent studies have suggested that protein arginine methyltransferase 3 (PRMT3) may act as a selective co-activator of LXR activity. Here, we verified the hypothesis that PRMT3 inhibition selectively disrupts the ability of LXR to stimulate lipogenesis while maintaining its capacity to modulate macrophage cholesterol homeostasis. EXPERIMENTAL APPROACH: A combination of the LXR agonist T0901317 and palm oil was administered to C57BL/6 mice to maximally stimulate LXR and PRMT3 activity. PRMT3 activity was inhibited using the allosteric inhibitor SGC707. KEY RESULTS: Treatment with SGC707 did not negatively influence the T0901317/palm oil-induced up-regulation of the cholesterol efflux ATP-binding cassette transporter genes, ABCA1 and ABCG1, in peritoneal cells. In contrast, SGC707 treatment was associated with a significant decrease in the hepatic expression of the lipogenic gene fatty acid synthase (-64%). A similar trend was observed for stearoyl-coenzyme A desaturase and acetyl CoA carboxylase expression (-43%; -56%). This obstruction of lipogenic gene transcription coincided with a significant 2.3-fold decrease in liver triglyceride content as compared with the T0901317 and palm oil-treated control group. CONCLUSION AND IMPLICATIONS: We showed that inhibition of PRMT3 activity by SGC707 treatment selectively impairs LXR-driven transcription of hepatic lipogenic genes, while the positive effect of LXR stimulation on macrophage cholesterol efflux pathways is maintained.


Assuntos
Isoquinolinas/farmacologia , Lipogênese/efeitos dos fármacos , Receptores X do Fígado/genética , Fígado/efeitos dos fármacos , Proteína-Arginina N-Metiltransferases/antagonistas & inibidores , Transportador 1 de Cassete de Ligação de ATP/genética , Membro 1 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/genética , Animais , Colesterol/sangue , Hidrocarbonetos Fluorados/farmacologia , Lipogênese/genética , Fígado/metabolismo , Receptores X do Fígado/agonistas , Macrófagos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Óleo de Palmeira/farmacologia , Sulfonamidas/farmacologia , Transcrição Gênica/efeitos dos fármacos , Triglicerídeos/sangue
10.
Atherosclerosis ; 274: 120-127, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29772480

RESUMO

BACKGROUND AND AIMS: Proteoglycan 4 (Prg4) has a high structural similarity with the established atherosclerosis-modulating proteoglycan versican, but its role in atherogenesis is still unknown. Therefore, the impact of Prg4 deficiency on macrophage function in vitro and atherosclerosis susceptibility in vivo was investigated. METHODS: The presence and localization of Prg4 was studied in atherosclerotic lesions. Furthermore, the effect of Prg4 deficiency on macrophage foam cell formation, cholesterol efflux and lipopolysaccharide (LPS) response was determined. Finally, susceptibility for atherosclerotic lesion formation was investigated in bone marrow-specific Prg4 knockout (KO) mice. RESULTS: Prg4 mRNA expression was induced 91-fold (p<0.001) in murine initial atherosclerotic lesions and Prg4 protein co-localized with human lesional macrophages. Murine Prg4 KO macrophages showed increased foam cell formation (+2.1-fold, p<0.01). In parallel, the expression of the cholesterol efflux genes ATP-binding cassette transporter A1 and scavenger receptor type B1 was lower (-35%, p<0.05;-40%, p<0.05) in Prg4 KO macrophages. This translated into an impaired cholesterol efflux to high-density lipoprotein (-13%, p<0.001) and apolipoprotein A1 (-8%, p<0.05). Furthermore, Prg4 KO macrophages showed an impaired LPS-induced rise in TNFα secretion as compared to wild-type controls (-31%, p<0.001), indicating a reduced inflammatory response. Combined, these pro- and anti-atherogenic effects did not translate into a significant difference in atherosclerotic lesion formation upon bone marrow-specific deletion of Prg4 in low-density lipoprotein receptor KO mice. CONCLUSIONS: Prg4 is present in macrophages in both murine and human atherosclerotic lesions and critically influences macrophage function, but deletion of Prg4 in bone marrow-derived cells does not affect atherosclerotic lesion development.


Assuntos
Aterosclerose/metabolismo , Células da Medula Óssea/metabolismo , Artérias Carótidas/metabolismo , Doenças das Artérias Carótidas/metabolismo , Células Espumosas/metabolismo , Macrófagos Peritoneais/metabolismo , Placa Aterosclerótica , Proteoglicanas/metabolismo , Transportador 1 de Cassete de Ligação de ATP/metabolismo , Animais , Aterosclerose/genética , Aterosclerose/patologia , Células da Medula Óssea/patologia , Transplante de Medula Óssea , Artérias Carótidas/patologia , Doenças das Artérias Carótidas/genética , Doenças das Artérias Carótidas/patologia , Células Cultivadas , Colesterol/metabolismo , Modelos Animais de Doenças , Células Espumosas/efeitos dos fármacos , Células Espumosas/patologia , Células Espumosas/transplante , Humanos , Mediadores da Inflamação/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/patologia , Macrófagos Peritoneais/transplante , Masculino , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout para ApoE , Fenótipo , Proteoglicanas/deficiência , Proteoglicanas/genética , Receptores Depuradores Classe B/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
11.
Atherosclerosis ; 261: 99-104, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28242047

RESUMO

BACKGROUND AND AIMS: Statin treatment disrupts HMG-CoA reductase-mediated endogenous cholesterol synthesis and lowers plasma LDL-cholesterol levels. Although statin treatment can theoretically impair adrenal steroid hormone synthesis, thus far, no effect on glucocorticoid output has been described, as LDL-cholesterol levels usually remain within the physiological range. However, novel statin-based treatment regimens that dramatically decrease LDL-cholesterol levels are currently employed. Here, we assessed whether inhibition of cholesterol synthesis under these relatively hypocholesterolemic conditions may alter adrenal glucocorticoid output. METHODS: Hypocholesterolemic apolipoprotein A1 (apoA1) knockout mice were administered high dose simvastatin twice daily for 3 days. RESULTS: Simvastatin treatment did not change plasma cholesterol levels or modify the adrenal expression levels of genes involved in cholesterol metabolism. However, simvastatin treatment lowered basal plasma levels of the primary glucocorticoid corticosterone (-62%; p < 0.05). Upon injection with adrenocorticotropic hormone, control-treated apoA1 knockout mice already showed only a mild increase in plasma corticosterone levels, indicative of relative glucocorticoid insufficiency. Importantly, simvastatin treatment further diminished the adrenal glucocorticoid response to adrenocorticotropic hormone exposure (two-way ANOVA p < 0.05 for treatment). Peak corticosterone levels were 49% lower (p < 0.01) upon simvastatin treatment. CONCLUSIONS: We have shown that simvastatin treatment aggravates the glucocorticoid insufficiency associated with hypocholesterolemia in mice. Our data suggest that (1) HMG-CoA reductase activity controls the adrenal steroidogenic capacity under hypocholesterolemic conditions and (2) imply that it might be important to monitor adrenal function in humans subjected to statin-based treatments aimed at achieving sub-physiological LDL-cholesterol levels, as these may potentially execute a negative impact on the glucocorticoid function in humans.


Assuntos
Glândulas Suprarrenais/efeitos dos fármacos , Insuficiência Adrenal/induzido quimicamente , Colesterol/sangue , Corticosterona/deficiência , Inibidores de Hidroximetilglutaril-CoA Redutases/toxicidade , Hipercolesterolemia/tratamento farmacológico , Sinvastatina/toxicidade , Testes de Função do Córtex Suprarrenal , Glândulas Suprarrenais/metabolismo , Glândulas Suprarrenais/fisiopatologia , Insuficiência Adrenal/sangue , Insuficiência Adrenal/fisiopatologia , Hormônio Adrenocorticotrópico/administração & dosagem , Animais , Apolipoproteína A-I/deficiência , Apolipoproteína A-I/genética , Biomarcadores/sangue , Corticosterona/sangue , Modelos Animais de Doenças , Regulação da Expressão Gênica , Hipercolesterolemia/sangue , Hipercolesterolemia/genética , Hipercolesterolemia/fisiopatologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de LDL/deficiência , Receptores de LDL/genética
12.
Br J Pharmacol ; 172(9): 2397-405, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25572138

RESUMO

BACKGROUND AND PURPOSE: Antipsychotic drugs have been shown to modulate the expression of ATP-binding cassette transporter A1 (ABCA1), a key factor in the anti-atherogenic reverse cholesterol transport process, in vitro. Here we evaluated the potential of the typical antipsychotic drug haloperidol to modulate the cholesterol efflux function of macrophages in vitro and their susceptibility to atherosclerosis in vivo. EXPERIMENTAL APPROACH: Thioglycollate-elicited peritoneal macrophages were used for in vitro studies. Hyperlipidaemic low-density lipoprotein (LDL) receptor knockout mice were implanted with a haloperidol-containing pellet and subsequently fed a Western-type diet for 5 weeks to induce the development of atherosclerotic lesions in vivo. KEY RESULTS: Haloperidol induced a 54% decrease in the mRNA expression of ABCA1 in peritoneal macrophages. This coincided with a 30% decrease in the capacity of macrophages to efflux cholesterol to apolipoprotein A1. Haloperidol treatment stimulated the expression of ABCA1 (+51%) and other genes involved in reverse cholesterol transport, that is, CYP7A1 (+98%) in livers of LDL receptor knockout mice. No change in splenic ABCA1 expression was noted. However, the average size of the atherosclerotic size was significantly smaller (-31%) in the context of a mildly more atherogenic metabolic phenotype upon haloperidol treatment. More importantly, haloperidol markedly lowered MCP-1 expression (-70%) and secretion (-28%) by peritoneal macrophages. CONCLUSIONS AND IMPLICATIONS: Haloperidol treatment lowered the susceptibility of hyperlipidaemic LDL receptor knockout mice to develop atherosclerotic lesions. Our findings suggest that the beneficial effect of haloperidol on atherosclerosis susceptibility can be attributed to its ability to inhibit macrophage chemotaxis.


Assuntos
Transportador 1 de Cassete de Ligação de ATP/efeitos dos fármacos , Antipsicóticos/farmacologia , Aorta/efeitos dos fármacos , Doenças da Aorta/prevenção & controle , Aterosclerose/prevenção & controle , Colesterol/metabolismo , Haloperidol/farmacologia , Hiperlipidemias/tratamento farmacológico , Macrófagos Peritoneais/efeitos dos fármacos , Receptores de LDL/deficiência , Transportador 1 de Cassete de Ligação de ATP/genética , Transportador 1 de Cassete de Ligação de ATP/metabolismo , Animais , Aorta/metabolismo , Aorta/patologia , Doenças da Aorta/genética , Doenças da Aorta/metabolismo , Doenças da Aorta/patologia , Apolipoproteína A-I/metabolismo , Aterosclerose/genética , Aterosclerose/metabolismo , Aterosclerose/patologia , Células Cultivadas , Quimiocina CCL2/metabolismo , Quimiotaxia/efeitos dos fármacos , Dieta Ocidental , Modelos Animais de Doenças , Regulação para Baixo , Feminino , Hiperlipidemias/genética , Hiperlipidemias/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Macrófagos Peritoneais/metabolismo , Camundongos Knockout , Placa Aterosclerótica , Receptores de LDL/genética
13.
PLoS One ; 7(12): e52605, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23285108

RESUMO

Spinal muscular atrophy (SMA) is a leading genetic cause of infant mortality, resulting primarily from the degeneration and loss of lower motor neurons. Studies using mouse models of SMA have revealed widespread heterogeneity in the susceptibility of individual motor neurons to neurodegeneration, but the underlying reasons remain unclear. Data from related motor neuron diseases, such as amyotrophic lateral sclerosis (ALS), suggest that morphological properties of motor neurons may regulate susceptibility: in ALS larger motor units innervating fast-twitch muscles degenerate first. We therefore set out to determine whether intrinsic morphological characteristics of motor neurons influenced their relative vulnerability to SMA. Motor neuron vulnerability was mapped across 10 muscle groups in SMA mice. Neither the position of the muscle in the body, nor the fibre type of the muscle innervated, influenced susceptibility. Morphological properties of vulnerable and disease-resistant motor neurons were then determined from single motor units reconstructed in Thy.1-YFP-H mice. None of the parameters we investigated in healthy young adult mice - including motor unit size, motor unit arbor length, branching patterns, motor endplate size, developmental pruning and numbers of terminal Schwann cells at neuromuscular junctions - correlated with vulnerability. We conclude that morphological characteristics of motor neurons are not a major determinant of disease-susceptibility in SMA, in stark contrast to related forms of motor neuron disease such as ALS. This suggests that subtle molecular differences between motor neurons, or extrinsic factors arising from other cell types, are more likely to determine relative susceptibility in SMA.


Assuntos
Neurônios Motores/metabolismo , Neurônios Motores/patologia , Atrofia Muscular Espinal/metabolismo , Animais , Modelos Animais de Doenças , Suscetibilidade a Doenças , Humanos , Camundongos , Camundongos Knockout , Doença dos Neurônios Motores/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Atrofia Muscular Espinal/genética , Junção Neuromuscular/metabolismo , Plasticidade Neuronal , Células de Schwann/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...