Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Circ Res ; 122(10): 1369-1384, 2018 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-29523554

RESUMEN

RATIONALE: Macrophages face a substantial amount of cholesterol after the ingestion of apoptotic cells, and the LIPA (lysosomal acid lipase) has a major role in hydrolyzing cholesteryl esters in the endocytic compartment. OBJECTIVE: Here, we directly investigated the role of LIPA-mediated clearance of apoptotic cells both in vitro and in vivo. METHODS AND RESULTS: We show that LIPA inhibition causes a defective efferocytic response because of impaired generation of 25-hydroxycholesterol and 27-hydroxycholesterol. Reduced synthesis of 25-hydroxycholesterol after LIPA inhibition contributed to defective mitochondria-associated membrane leading to mitochondrial oxidative stress-induced NLRP3 (NOD-like receptor family, pyrin domain containing) inflammasome activation and caspase-1-dependent Rac1 (Ras-related C3 botulinum toxin substrate 1) degradation. A secondary event consisting of failure to appropriately activate liver X receptor-mediated pathways led to mitigation of cholesterol efflux and apoptotic cell clearance. In mice, LIPA inhibition caused defective clearance of apoptotic lymphocytes and stressed erythrocytes by hepatic and splenic macrophages, culminating in splenomegaly and splenic iron accumulation under hypercholesterolemia. CONCLUSIONS: Our findings position lysosomal cholesterol hydrolysis as a critical process that prevents metabolic inflammation by enabling efficient macrophage apoptotic cell clearance.


Asunto(s)
Colesterol/metabolismo , Inflamación/metabolismo , Lisosomas/metabolismo , Macrófagos/metabolismo , Oxiesteroles/metabolismo , Esterol Esterasa/metabolismo , Animales , Apoptosis , Transporte Biológico , Ésteres del Colesterol/metabolismo , Eritrocitos/metabolismo , Hidrólisis , Hipercolesterolemia/metabolismo , Inflamasomas/metabolismo , Receptores X del Hígado/metabolismo , Linfocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Neuropéptidos/metabolismo , Receptores de LDL/metabolismo , Esplenomegalia/metabolismo , Esterol Esterasa/antagonistas & inhibidores , Proteína de Unión al GTP rac1/metabolismo
2.
Cell Physiol Biochem ; 53(3): 573-586, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31529929

RESUMEN

BACKGROUND/AIMS: In our recent work, the importance of GSK3ß-mediated phosphorylation of presenilin-1 as crucial process to establish a Ca2+ leak in the endoplasmic reticulum and, subsequently, the pre-activation of resting mitochondrial activity in ß-cells was demonstrated. The present work is a follow-up and reveals the importance of GSK3ß-phosphorylated presenilin-1 for responsiveness of pancreatic islets and ß-cells to elevated glucose in terms of cytosolic Ca2+ spiking and insulin secretion. METHODS: Freshly isolated pancreatic islets and the two pancreatic ß-cell lines INS-1 and MIN-6 were used. Cytosolic Ca2+ was fluorometrically monitored using Fura-2/AM and cellular insulin content and secretion were measured by ELISA. RESULTS: Our data strengthened our previous findings of the existence of a presenilin-1-mediated ER-Ca2+ leak in ß-cells, since a reduction of presenilin-1 expression strongly counteracted the ER Ca2+ leak. Furthermore, our data revealed that cytosolic Ca2+ spiking upon administration of high D-glucose was delayed in onset time and strongly reduced in amplitude and frequency upon siRNA-mediated knock-down of presenilin-1 or the inhibition of GSK3ß in the pancreatic ß-cells. Moreover, glucose-triggered initial insulin secretion disappeared by depletion from presenilin-1 and inhibition of GSK3ß in the pancreatic ß-cells and isolated pancreatic islets, respectively. CONCLUSION: These data complement our previous work and demonstrate that the sensitivity of pancreatic islets and ß-cells to glucose illustrated as glucose-triggered cytosolic Ca2+ spiking and initial but not long-lasting insulin secretion crucially depends on a strong ER Ca2+ leak that is due to the phosphorylation of presenilin-1 by GSK3ß, a phenomenon that might be involved in the development of type 2 diabetes.


Asunto(s)
Retículo Endoplásmico/metabolismo , Glucosa/farmacología , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Presenilina-1/metabolismo , Animales , Antracenos/farmacología , Calcio/metabolismo , Línea Celular , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Retículo Endoplásmico/efectos de los fármacos , Humanos , Secreción de Insulina/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , Islotes Pancreáticos/efectos de los fármacos , MAP Quinasa Quinasa 4/antagonistas & inhibidores , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo
3.
Cell Physiol Biochem ; 52(1): 57-75, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30790505

RESUMEN

BACKGROUND/AIMS: In pancreatic ß-cells, the intracellular Ca²âº homeostasis is an essential regulator of the cells major functions. The endoplasmic reticulum (ER) as interactive intracellular Ca²âº store balances cellular Ca²âº. In this study basal ER Ca²âº homeostasis was evaluated in order to reveal potential ß-cell-specificity of ER Ca²âº handling and its consequences for mitochondrial Ca²âº, ATP and respiration. METHODS: The two pancreatic cell lines INS-1 and MIN-6, freshly isolated pancreatic islets, and the two non-pancreatic cell lines HeLA and EA.hy926 were used. Cytosolic, ER and mitochondrial Ca²âº and ATP measurements were performed using single cell fluorescence microscopy and respective (genetically-encoded) sensors/dyes. Mitochondrial respiration was monitored by respirometry. GSK3ß activity was measured with ELISA. RESULTS: An atypical ER Ca²âº leak was observed exclusively in pancreatic islets and ß-cells. This continuous ER Ca²âº efflux is directed to mitochondria and increases basal respiration and organellar ATP levels, is established by GSK3ß-mediated phosphorylation of presenilin-1, and is prevented by either knockdown of presenilin-1 or an inhibition/knockdown of GSK3ß. Expression of a presenlin-1 mutant that mimics GSK3ß-mediated phosphorylation established a ß-cell-like ER Ca²âº leak in HeLa and EA.hy926 cells. The ER Ca²âº loss in ß-cells was compensated at steady state by Ca²âº entry that is linked to the activity of TRPC3. CONCLUSION: Pancreatic ß-cells establish a cell-specific ER Ca²âº leak that is under the control of GSK3ß and directed to mitochondria, thus, reflecting a cell-specific intracellular Ca²âº handling for basal mitochondrial activity.


Asunto(s)
Señalización del Calcio , Calcio/metabolismo , Retículo Endoplásmico/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Células Secretoras de Insulina/metabolismo , Mitocondrias/metabolismo , Presenilina-1/metabolismo , Animales , Línea Celular Tumoral , Retículo Endoplásmico/genética , Glucógeno Sintasa Quinasa 3 beta/genética , Células HeLa , Humanos , Masculino , Ratones , Mitocondrias/genética , Fosforilación , Presenilina-1/genética , Ratas
4.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1863(4): 467-478, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29374543

RESUMEN

Lysosomal acid lipase (LAL) is the only known enzyme, which hydrolyzes cholesteryl esters and triacylglycerols in lysosomes of multiple cells and tissues. Here, we explored the role of LAL in brown adipose tissue (BAT). LAL-deficient (Lal-/-) mice exhibit markedly reduced UCP1 expression in BAT, modified BAT morphology with accumulation of lysosomes, and mitochondrial dysfunction, consequently leading to regular hypothermic events in mice kept at room temperature. Cold exposure resulted in reduced lipid uptake into BAT, thereby aggravating dyslipidemia and causing life threatening hypothermia in Lal-/- mice. Linking LAL as a potential regulator of lipoprotein lipase activity, we found Angptl4 mRNA expression upregulated in BAT. Our data demonstrate that LAL is critical for shuttling fatty acids derived from circulating lipoproteins to BAT during cold exposure. We conclude that inhibited lysosomal lipid hydrolysis in BAT leads to impaired thermogenesis in Lal-/- mice.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Ácidos Grasos/metabolismo , Esterol Esterasa/metabolismo , Termogénesis , Acetilcoenzima A/metabolismo , Adipocitos Marrones/metabolismo , Tejido Adiposo Pardo/ultraestructura , Animales , Autofagia , Temperatura Corporal , Carnitina/análogos & derivados , Carnitina/metabolismo , Frío , Progresión de la Enfermedad , Dislipidemias/metabolismo , Dislipidemias/patología , Metabolismo Energético , Glucosa/metabolismo , Hipotermia Inducida , Gotas Lipídicas/metabolismo , Lipólisis , Masculino , Ratones Endogámicos C57BL , Músculos/metabolismo , Oxidación-Reducción , Consumo de Oxígeno , Esterol Esterasa/deficiencia , Proteína Desacopladora 1/metabolismo
5.
Mol Metab ; 71: 101705, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36907508

RESUMEN

OBJECTIVE: In brown adipose tissue (iBAT), the balance between lipid/glucose uptake and lipolysis is tightly regulated by insulin signaling. Downstream of the insulin receptor, PDK1 and mTORC2 phosphorylate AKT, which activates glucose uptake and lysosomal mTORC1 signaling. The latter requires the late endosomal/lysosomal adaptor and MAPK and mTOR activator (LAMTOR/Ragulator) complex, which serves to translate the nutrient status of the cell to the respective kinase. However, the role of LAMTOR in metabolically active iBAT has been elusive. METHODS: Using an AdipoqCRE-transgenic mouse line, we deleted LAMTOR2 (and thereby the entire LAMTOR complex) in adipose tissue (LT2 AKO). To examine the metabolic consequences, we performed metabolic and biochemical studies in iBAT isolated from mice housed at different temperatures (30 °C, room temperature and 5 °C), after insulin treatment, or in fasted and refed condition. For mechanistic studies, mouse embryonic fibroblasts (MEFs) lacking LAMTOR 2 were analyzed. RESULTS: Deletion of the LAMTOR complex in mouse adipocytes resulted in insulin-independent AKT hyperphosphorylation in iBAT, causing increased glucose and fatty acid uptake, which led to massively enlarged lipid droplets. As LAMTOR2 was essential for the upregulation of de novo lipogenesis, LAMTOR2 deficiency triggered exogenous glucose storage as glycogen in iBAT. These effects are cell autonomous, since AKT hyperphosphorylation was abrogated by PI3K inhibition or by deletion of the mTORC2 component Rictor in LAMTOR2-deficient MEFs. CONCLUSIONS: We identified a homeostatic circuit for the maintenance of iBAT metabolism that links the LAMTOR-mTORC1 pathway to PI3K-mTORC2-AKT signaling downstream of the insulin receptor.


Asunto(s)
Proteínas Proto-Oncogénicas c-akt , Receptor de Insulina , Ratones , Animales , Receptor de Insulina/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Tejido Adiposo Pardo/metabolismo , Fibroblastos/metabolismo , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Insulina/metabolismo , Ratones Transgénicos , Lisosomas/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Nutrientes , Homeostasis , Glucosa/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas/metabolismo
6.
Cells ; 10(10)2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34685599

RESUMEN

Lysosomal acid lipase (LAL) is the sole enzyme known to be responsible for the hydrolysis of cholesteryl esters and triglycerides at an acidic pH in lysosomes, resulting in the release of unesterified cholesterol and free fatty acids. However, the role of LAL in diet-induced adaptations is largely unexplored. In this study, we demonstrate that feeding a Western-type diet to Lal-deficient (LAL-KO) mice triggers metabolic reprogramming that modulates gut-liver cholesterol homeostasis. Induction of ileal fibroblast growth factor 15 (three-fold), absence of hepatic cholesterol 7α-hydroxylase expression, and activation of the ERK phosphorylation cascade results in altered bile acid composition, substantial changes in the gut microbiome, reduced nutrient absorption by 40%, and two-fold increased fecal lipid excretion in LAL-KO mice. These metabolic adaptations lead to impaired bile acid synthesis, lipoprotein uptake, and cholesterol absorption and ultimately to the resistance of LAL-KO mice to diet-induced obesity. Our results indicate that LAL-derived lipolytic products might be important metabolic effectors in the maintenance of whole-body lipid homeostasis.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Disbiosis/metabolismo , Metabolismo de los Lípidos , Obesidad/metabolismo , Esterol Esterasa/fisiología , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Esterol Esterasa/genética
7.
Bone ; 148: 115946, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33838322

RESUMEN

Lysosomal acid lipase (LAL) is essential for cholesteryl ester (CE) and triacylglycerol (TAG) hydrolysis in the lysosome. Clinically, an autosomal recessive LIPA mutation causes LAL deficiency (LALD), previously described as Wolman Disease or Cholesteryl Ester Storage Disease (CESD). LAL-D is associated with ectopic lipid accumulation in the liver, small intestine, spleen, adrenal glands, and blood. Considering the importance of unesterified cholesterol and fatty acids in bone metabolism, we hypothesized that LAL is essential for bone formation, and ultimately, skeletal health. To investigate the role of LAL in skeletal homeostasis, we used LAL-deficient (-/-) mice, in vitro osteoblast cultures, and novel clinical data from LAL-D patients. Both male and female LAL-/- mice demonstarted lower trabecular and cortical bone parameters , which translated to reduced biomechanical properties. Further histological analyses revealed that LAL-/- mice had fewer osteoblasts, with no change in osteoclast or marrow adipocyte numbers. In studying the cell-autonomous role of LAL, we observed impaired differentiation of LAL-/- calvarial osteoblasts and in bone marrow stromal cells treated with the LAL inhibitor lalistat. Consistent with LAL's role in other tissues, lalistat resulted in profound lipid puncta accumulation and an altered intracellular lipid profile. Finally, we analyzed a large de-identified national insurance database (i.e. 2016/2017 Optum Clinformatics®) which revealed that adults (≥18 years) with CESD (n = 3076) had a higher odds ratio (OR = 1.21; 95% CI = 1.03-1.41) of all-cause fracture at any location compared to adults without CESD (n = 13.7 M) after adjusting for demographic variables and osteoporosis. These data demonstrate that alterations in LAL have significant clinical implications related to fracture risk and that LAL's modulation of lipid metabolism is a critical for osteoblast function.


Asunto(s)
Enfermedad de Acumulación de Colesterol Éster , Enfermedad de Wolman , Animales , Ésteres del Colesterol , Femenino , Humanos , Hígado , Masculino , Ratones , Esterol Esterasa/genética , Enfermedad de Wolman/genética
8.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1864(4): 500-511, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30639734

RESUMEN

Lysosomal acid lipase (LAL) hydrolyzes cholesteryl esters (CE) and triglycerides (TG) to generate fatty acids (FA) and cholesterol. LAL deficiency (LAL-D) in both humans and mice leads to hepatomegaly, hypercholesterolemia, and shortened life span. Despite its essential role in lysosomal neutral lipid catabolism, the cell type-specific contribution of LAL to disease progression is still elusive. To investigate the role of LAL in the liver in more detail and to exclude the contribution of LAL in macrophages, we generated hepatocyte-specific LAL-deficient mice (Liv-Lipa-/-) and fed them either chow or high fat/high cholesterol diets (HF/HCD). Comparable to systemic LAL-D, Liv-Lipa-/- mice were resistant to diet-induced obesity independent of food intake, movement, and energy expenditure. Reduced body weight gain was mainly due to reduced white adipose tissue depots. Furthermore, Liv-Lipa-/- mice exhibited improved glucose clearance during glucose and insulin tolerance tests compared to control mice. Analysis of hepatic lipid content revealed a massive reduction of TG, whereas CE concentrations were markedly increased, leading to CE crystal formation in the livers of Liv-Lipa-/- mice. Elevated plasma transaminase activities, increased pro-inflammatory cytokines and chemokines as well as hepatic macrophage infiltration indicated liver inflammation. Our data provide evidence that hepatocyte-specific LAL deficiency is sufficient to alter whole-body lipid and energy homeostasis in mice. We conclude that hepatic LAL plays a pivotal role by preventing liver damage and maintaining lipid and energy homeostasis, especially during high lipid availability.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Hepatitis/genética , Hepatocitos/enzimología , Obesidad/prevención & control , Esterol Esterasa/deficiencia , Animales , Modelos Animales de Enfermedad , Técnicas de Silenciamiento del Gen , Hepatocitos/inmunología , Homeostasis , Metabolismo de los Lípidos , Masculino , Ratones , Obesidad/inducido químicamente , Obesidad/genética , Esterol Esterasa/genética , Esterol Esterasa/metabolismo
9.
Biochim Biophys Acta Mol Cell Res ; 1866(3): 337-348, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30595160

RESUMEN

The discovery of significant amounts of metabolically active brown adipose tissue (BAT) in adult humans renders it a promising target for anti-obesity therapies by inducing weight loss through increased energy expenditure. The components of the N-acetylaspartate (NAA) pathway are highly abundant in BAT. Aspartate N-acetyltransferase (Asp-NAT, encoded by Nat8l) synthesizes NAA from acetyl-CoA and aspartate and increases energy expenditure in brown adipocytes. However, the exact mechanism how the NAA pathway contributes to accelerated mobilization and oxidation of lipids and the physiological regulation of the NAA pathway remained elusive. Here, we demonstrate that the expression of NAA pathway genes corresponds to nutrient availability and specifically responds to changes in exogenous glucose. NAA is preferentially produced from glucose-derived acetyl-CoA and aspartate and its concentration increases during adipogenesis. Overexpression of Nat8l drains glucose-derived acetyl-CoA into the NAA pool at the expense of cellular lipids and certain amino acids. Mechanistically, we elucidated that a combined activation of neutral and lysosomal (acid) lipolysis is responsible for the increased lipid degradation. Specifically, translocation of the transcription factor EB to the nucleus activates the biosynthesis of autophagosomes and lysosomes. Lipid degradation within lysosomes accompanied by adipose triglyceride lipase-mediated lipolysis delivers fatty acids for the support of elevated mitochondrial respiration. Together, our data suggest a crucial role of the NAA pathway in energy metabolism and metabolic adaptation in BAT.


Asunto(s)
Adipocitos Marrones/metabolismo , Ácido Aspártico/análogos & derivados , Nutrientes/metabolismo , Acetilcoenzima A/metabolismo , Acetiltransferasas/metabolismo , Adipocitos Marrones/fisiología , Adipogénesis/genética , Adipogénesis/fisiología , Tejido Adiposo Pardo/metabolismo , Animales , Ácido Aspártico/genética , Ácido Aspártico/metabolismo , Ácido Aspártico/fisiología , Metabolismo Energético/fisiología , Ácidos Grasos/metabolismo , Glucosa/metabolismo , Metabolismo de los Lípidos/fisiología , Lípidos/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Oxidación-Reducción
10.
Sci Rep ; 7(1): 12485, 2017 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-28970555

RESUMEN

Endothelial lipase (EL) is a potent modulator of the structural and functional properties of HDL. Impact of EL on cholesterol efflux capacity (CEC) of serum and isolated HDL is not well understood and apparently contradictory data were published. Here, we systematically examined the impact of EL on composition and CEC of serum and isolated HDL, in vitro and in vivo, using EL-overexpressing cells and EL-overexpressing mice. CEC was examined in a validated assay using 3H-cholesterol labelled J774 macrophages. In vitro EL-modification of serum resulted in complex alterations, including enrichment of serum with lipid-free/-poor apoA-I, decreased size of human (but not mouse) HDL and altered HDL lipid composition. EL-modification of serum increased CEC, in line with increased lipid-free/-poor apoA-I formation. In contrast, CEC of isolated HDL was decreased likely through altered lipid composition. In contrast to in vitro results, EL-overexpression in mice markedly decreased HDL-cholesterol and apolipoprotein A-I serum levels associated with a decreased CEC of serum. HDL lipid composition was altered, but HDL particle size and CEC were not affected. Our study highlights the multiple and complex effects of EL on HDL composition and function and may help to clarify the seemingly contradictory data found in published articles.


Asunto(s)
Apolipoproteína A-I/sangre , Apolipoproteína B-100/sangre , HDL-Colesterol/sangre , Lipasa/genética , Animales , Transporte Biológico , Línea Celular , Expresión Génica , Células Hep G2 , Humanos , Lipasa/sangre , Macrófagos/citología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Tamaño de la Partícula
11.
Oncotarget ; 8(25): 40037-40051, 2017 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-28402950

RESUMEN

Degradation of lysosomal lipids requires lysosomal acid lipase (LAL), the only intracellular lipase known to be active at acidic pH. We found LAL to be expressed in murine immune cells with highest mRNA expression in macrophages and neutrophils. Furthermore, we observed that loss of LAL in mice caused lipid accumulation in white blood cells in the peripheral circulation, which increased in response to an acute inflammatory stimulus. Lal-deficient (-/-) macrophages accumulate neutral lipids, mainly cholesteryl esters, within lysosomes. The cholesteryl ester fraction is particularly enriched in the PUFAs 18:2 and 20:4, important precursor molecules for lipid mediator synthesis. To investigate whether loss of LAL activity affects the generation of lipid mediators and to eliminate potential systemic effects from other cells and tissues involved in the pronounced phenotype of Lal-/- mice, we treated macrophages from Wt mice with the LAL-specific inhibitor LAListat-2. Acute inhibition of LAL resulted in reduced release of 18:2- and 20:4-derived mediators from macrophages, indicating that lipid hydrolysis by LAL is an important source for lipid mediator synthesis in macrophages. We conclude that lysosomes should be considered as organelles that provide precursor molecules for lipid mediators such as eicosanoids.


Asunto(s)
Metabolismo de los Lípidos , Lisosomas/metabolismo , Macrófagos/metabolismo , Esterol Esterasa/metabolismo , Animales , Carbamatos/farmacología , Ésteres del Colesterol/metabolismo , Eicosanoides/metabolismo , Femenino , Hidrólisis , Lípidos/análisis , Lípidos/sangre , Macrófagos/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Esterol Esterasa/antagonistas & inhibidores , Esterol Esterasa/genética , Especificidad por Sustrato , Tiadiazoles/farmacología
12.
Oncotarget ; 8(20): 33122-33136, 2017 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-28380440

RESUMEN

Monoglyceride lipase (MGL) hydrolyzes monoglycerides (MGs) to glycerol and fatty acids. Among various MG species MGL also degrades 2-arachidonoylglycerol (2-AG), the most abundant endocannabinoid and potent activator of cannabinoid receptors (CBR) 1 and 2. MGL-knockout (-/-) mice exhibit pronounced 2-AG accumulation, but lack central cannabimimetic effects due to CB1R desensitization. We have previously shown that MGL affects plaque stability in apolipoprotein E (ApoE)-/- mice, an established animal model for dyslipidemia and atherosclerosis. In the current study, we investigated functional consequences of MGL deficiency on lipid and energy metabolism in ApoE/MGL double knockout (DKO) mice. MGL deficiency affected hepatic cholesterol metabolism by causing increased cholesterol elimination via the biliary pathway. Moreover, DKO mice exhibit lipid-triggered delay in gastric emptying without major effects on overall triglyceride and cholesterol absorption. The observed phenotype of DKO mice is likely not a consequence of potentiated CB1R signaling but rather dependent on the activation of alternative signaling pathways. We conclude that MGL deficiency causes complex metabolic changes including cholesterol metabolism and regulation of gut transit independent of the endocannabinoid system.


Asunto(s)
Apolipoproteínas E/genética , Asialoglicoproteínas/genética , Aterosclerosis/metabolismo , Colesterol/metabolismo , Dislipidemias/metabolismo , Lectinas Tipo C/genética , Hígado/metabolismo , Proteínas de la Membrana/genética , Oxidorreductasas de Alcohol/metabolismo , Animales , Ácidos Araquidónicos/metabolismo , Asialoglicoproteínas/deficiencia , Modelos Animales de Enfermedad , Endocannabinoides/metabolismo , Técnicas de Inactivación de Genes , Glicéridos/metabolismo , Mucosa Intestinal/metabolismo , Lectinas Tipo C/deficiencia , Masculino , Proteínas de la Membrana/deficiencia , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA