Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Hepatology ; 72(6): 2149-2164, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32170749

RESUMEN

BACKGROUND AND AIMS: Liver regeneration requires the organized and sequential activation of events that lead to restoration of hepatic mass. During this process, other vital liver functions need to be preserved, such as maintenance of blood glucose homeostasis, balancing the degradation of hepatic glycogen stores, and gluconeogenesis (GNG). Under metabolic stress, alanine is the main hepatic gluconeogenic substrate, and its availability is the rate-limiting step in this pathway. Na+ -coupled neutral amino acid transporters (SNATs) 2 and 4 are believed to facilitate hepatic alanine uptake. In previous studies, we demonstrated that a member of the Ca2+ -dependent phospholipid binding annexins, Annexin A6 (AnxA6), regulates membrane trafficking along endo- and exocytic pathways. Yet, although AnxA6 is abundantly expressed in the liver, its function in hepatic physiology remains unknown. In this study, we investigated the potential contribution of AnxA6 in liver regeneration. APPROACH AND RESULTS: Utilizing AnxA6 knockout mice (AnxA6-/- ), we challenged liver function after partial hepatectomy (PHx), inducing acute proliferative and metabolic stress. Biochemical and immunofluorescent approaches were used to dissect AnxA6-/- mice liver proliferation and energetic metabolism. Most strikingly, AnxA6-/- mice exhibited low survival after PHx. This was associated with an irreversible and progressive drop of blood glucose levels. Whereas exogenous glucose administration or restoration of hepatic AnxA6 expression rescued AnxA6-/- mice survival after PHx, the sustained hypoglycemia in partially hepatectomized AnxA6-/- mice was the consequence of an impaired alanine-dependent GNG in AnxA6-/- hepatocytes. Mechanistically, cytoplasmic SNAT4 failed to recycle to the sinusoidal plasma membrane of AnxA6-/- hepatocytes 48 hours after PHx, impairing alanine uptake and, consequently, glucose production. CONCLUSIONS: We conclude that the lack of AnxA6 compromises alanine-dependent GNG and liver regeneration in mice.


Asunto(s)
Anexina A6/metabolismo , Gluconeogénesis/fisiología , Regeneración Hepática/fisiología , Animales , Anexina A6/genética , Membrana Celular/metabolismo , Modelos Animales de Enfermedad , Glucosa/metabolismo , Glucólisis/fisiología , Hepatectomía , Hepatocitos/metabolismo , Humanos , Hígado/citología , Hígado/metabolismo , Hígado/cirugía , Masculino , Ratones , Ratones Noqueados
2.
Exp Cell Res ; 358(2): 397-410, 2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-28712927

RESUMEN

Annexin A6 (AnxA6) has been implicated in the regulation of endo-/exocytic pathways, cholesterol transport, and the formation of multifactorial signaling complexes in many different cell types. More recently, AnxA6 has also been linked to triglyceride storage in adipocytes. Here we investigated the potential role of AnxA6 in fatty acid (FA) - induced lipid droplet (LD) formation in hepatocytes. AnxA6 was associated with LD from rat liver and HuH7 hepatocytes. In oleic acid (OA) -loaded HuH7 cells, substantial amounts of AnxA6 bound to LD in a Ca2+-independent manner. Remarkably, stable or transient AnxA6 overexpression in HuH7 cells led to elevated LD numbers/size and neutral lipid staining under control conditions as well as after OA loading compared to controls. In contrast, overexpression of AnxA1, AnxA2 and AnxA8 did not impact on OA-induced lipid accumulation. On the other hand, incubation of AnxA6-depleted HuH7 cells or primary hepatocytes from AnxA6 KO-mice with OA led to reduced FA accumulation and LD numbers. Furthermore, morphological analysis of liver sections from A6-KO mice revealed significantly lower LD numbers compared to wildtype animals. Interestingly, pharmacological inhibition of cytoplasmic phospholipase A2α (cPLA2α)-dependent LD formation was ineffective in AnxA6-depleted HuH7 cells. We conclude that cPLA2α-dependent pathways contribute to the novel regulatory role of hepatic AnxA6 in LD formation.


Asunto(s)
Anexina A6/metabolismo , Hepatocitos/metabolismo , Gotas Lipídicas/metabolismo , Lipogénesis/fisiología , Animales , Transporte Biológico/fisiología , Línea Celular , Humanos , Hígado/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratas Sprague-Dawley
3.
J Biol Chem ; 291(3): 1320-35, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26578516

RESUMEN

Annexins are a family of proteins that bind to phospholipids in a calcium-dependent manner. Earlier studies implicated annexin A6 (AnxA6) to inhibit secretion and participate in the organization of the extracellular matrix. We recently showed that elevated AnxA6 levels significantly reduced secretion of the extracellular matrix protein fibronectin (FN). Because FN is directly linked to the ability of cells to migrate, this prompted us to investigate the role of AnxA6 in cell migration. Up-regulation of AnxA6 in several cell models was associated with reduced cell migration in wound healing, individual cell tracking and three-dimensional migration/invasion assays. The reduced ability of AnxA6-expressing cells to migrate was associated with decreased cell surface expression of αVß3 and α5ß1 integrins, both FN receptors. Mechanistically, we found that elevated AnxA6 levels interfered with syntaxin-6 (Stx6)-dependent recycling of integrins to the cell surface. AnxA6 overexpression caused mislocalization and accumulation of Stx6 and integrins in recycling endosomes, whereas siRNA-mediated AnxA6 knockdown did not modify the trafficking of integrins. Given our recent findings that inhibition of cholesterol export from late endosomes (LEs) inhibits Stx6-dependent integrin recycling and that elevated AnxA6 levels cause LE cholesterol accumulation, we propose that AnxA6 and blockage of LE cholesterol transport are critical for endosomal function required for Stx6-mediated recycling of integrins in cell migration.


Asunto(s)
Anexina A6/metabolismo , Colesterol/metabolismo , Endosomas/metabolismo , Integrina alfa5beta1/metabolismo , Integrina alfaVbeta3/metabolismo , Proteínas Qa-SNARE/metabolismo , Animales , Anexina A6/antagonistas & inhibidores , Anexina A6/genética , Células CHO , Línea Celular Tumoral , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Movimiento Celular , Células Cultivadas , Cricetulus , Endosomas/ultraestructura , Fibroblastos/citología , Fibroblastos/metabolismo , Fibroblastos/ultraestructura , Humanos , Integrina alfa5beta1/antagonistas & inhibidores , Integrina alfaVbeta3/antagonistas & inhibidores , Ratones , Microscopía Confocal , Microscopía por Video , Proteínas Qa-SNARE/antagonistas & inhibidores , Proteínas Qa-SNARE/genética , Interferencia de ARN , Ratas , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Imagen de Lapso de Tiempo
4.
Immunol Cell Biol ; 94(6): 543-53, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26853809

RESUMEN

Annexin A6 (AnxA6) has been implicated in cell signalling by contributing to the organisation of the plasma membrane. Here we examined whether AnxA6 regulates signalling and proliferation in T cells. We used a contact hypersensitivity model to immune challenge wild-type (WT) and AnxA6(-/-) mice and found that the in vivo proliferation of CD4(+) T cells, but not CD8(+) T cells, was impaired in AnxA6(-/-) relative to WT mice. However, T-cell migration and signalling through the T-cell receptor ex vivo was similar between T cells isolated from AnxA6(-/-) and WT mice. In contrast, interleukin-2 (IL-2) signalling was reduced in AnxA6(-/-) compared with WT T cells. Further, AnxA6-deficient T cells had reduced membrane order and cholesterol levels. Taken together, our data suggest that AnxA6 regulates IL-2 homeostasis and sensitivity in T cells by sustaining a lipid raft-like membrane environment.


Asunto(s)
Anexina A6/metabolismo , Interleucina-2/metabolismo , Linfocitos T/citología , Linfocitos T/metabolismo , Animales , Anexina A6/deficiencia , Membrana Celular/metabolismo , Movimiento Celular , Proliferación Celular , Colesterol/metabolismo , Dermatitis por Contacto/inmunología , Dermatitis por Contacto/patología , Interleucina-2/biosíntesis , Activación de Linfocitos , Ratones Endogámicos C57BL , Fosforilación , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores de Interleucina-2/metabolismo , Factor de Transcripción STAT5/metabolismo , Transducción de Señal
5.
Hepatology ; 60(4): 1367-77, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24700364

RESUMEN

UNLABELLED: Severe liver diseases are characterized by expansion of liver progenitor cells (LPC), which correlates with disease severity. However, the origin and role of LPC in liver physiology and in hepatic injury remains a contentious topic. We found that ductular reaction cells in human cirrhotic livers express hepatocyte nuclear factor 1 homeobox B (HNF1ß). However, HNF1ß expression was not present in newly generated epithelial cell adhesion molecule (EpCAM)-positive hepatocytes. In order to investigate the role of HNF1ß-expressing cells we used a tamoxifen-inducible Hnf1ßCreER/R26R(Yfp/LacZ) mouse to lineage-trace Hnf1ß(+) biliary duct cells and to assess their contribution to LPC expansion and hepatocyte generation. Lineage tracing demonstrated no contribution of HNF1ß(+) cells to hepatocytes during liver homeostasis in healthy mice or after loss of liver mass. After acute acetaminophen or carbon tetrachloride injury no contribution of HNF1ß(+) cells to hepatocyte was detected. We next assessed the contribution of Hnf1ß(+) -derived cells following two liver injury models with LPC expansion, a diethoxycarbonyl-1,4-dihydro-collidin (DDC)-diet and a choline-deficient ethionine-supplemented (CDE)-diet. The contribution of Hnf1ß(+) cells to liver regeneration was dependent on the liver injury model. While no contribution was observed after DDC-diet treatment, mice fed with a CDE-diet showed a small population of hepatocytes derived from Hnf1ß(+) cells that were expanded to 1.86% of total hepatocytes after injury recovery. Genome-wide expression profile of Hnf1ß(+) -derived cells from the DDC and CDE models indicated that no contribution of LPC to hepatocytes was associated with LPC expression of genes related to telomere maintenance, inflammation, and chemokine signaling pathways. CONCLUSION: HNF1ß(+) biliary duct cells are the origin of LPC. HNF1ß(+) cells do not contribute to hepatocyte turnover in the healthy liver, but after certain liver injury, they can differentiate to hepatocytes contributing to liver regeneration.


Asunto(s)
Conductos Biliares/patología , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Células Epiteliales/patología , Hepatocitos/patología , Regeneración Hepática/fisiología , Hígado/patología , Células Madre/patología , Acetaminofén/efectos adversos , Animales , Conductos Biliares/metabolismo , Tetracloruro de Carbono/efectos adversos , Diferenciación Celular/fisiología , Linaje de la Célula , Células Cultivadas , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/fisiopatología , Dieta/efectos adversos , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Femenino , Factor Nuclear 1-beta del Hepatocito/metabolismo , Hepatocitos/metabolismo , Homeostasis/fisiología , Humanos , Hígado/metabolismo , Masculino , Ratones , Ratones Transgénicos , Células Madre/metabolismo
6.
Mol Nutr Food Res ; : e2400020, 2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-38934352

RESUMEN

Sex hormone-binding globulin (SHBG) is a homodimeric glycoprotein produced by the human liver and secreted into the systemic circulation where it binds with high affinity sex steroids regulating their availability in blood and accessibility to target tissues. Plasma SHBG levels are altered in metabolic disorders such as obesity, anorexia, and insulin resistance. Several reports have shown that diets in terms of total calories or fat, fiber, or protein content can alter plasma SHBG levels. However, there are many components in a diet that can affect SHBG gene expression in the liver. In order to unravel the molecular mechanisms by which diets regulate SHBG production, it would be necessary to analyze single diet components and/or nutritional factors. This review summarizes the recent advances in identifying different nutritional factors regulating SHBG production and the related molecular mechanism, as well as the clinical implications.

7.
Elife ; 122023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36722855

RESUMEN

Mitochondrial dysfunction has been reported in obesity and insulin resistance, but primary genetic mitochondrial dysfunction is generally not associated with these, arguing against a straightforward causal relationship. A rare exception, recently identified in humans, is a syndrome of lower body adipose loss, leptin-deficient severe upper body adipose overgrowth, and insulin resistance caused by the p.Arg707Trp mutation in MFN2, encoding mitofusin 2. How the resulting selective form of mitochondrial dysfunction leads to tissue- and adipose depot-specific growth abnormalities and systemic biochemical perturbation is unknown. To address this, Mfn2R707W/R707W knock-in mice were generated and phenotyped on chow and high fat diets. Electron microscopy revealed adipose-specific mitochondrial morphological abnormalities. Oxidative phosphorylation measured in isolated mitochondria was unperturbed, but the cellular integrated stress response was activated in adipose tissue. Fat mass and distribution, body weight, and systemic glucose and lipid metabolism were unchanged, however serum leptin and adiponectin concentrations, and their secretion from adipose explants were reduced. Pharmacological induction of the integrated stress response in wild-type adipocytes also reduced secretion of leptin and adiponectin, suggesting an explanation for the in vivo findings. These data suggest that the p.Arg707Trp MFN2 mutation selectively perturbs mitochondrial morphology and activates the integrated stress response in adipose tissue. In mice, this does not disrupt most adipocyte functions or systemic metabolism, whereas in humans it is associated with pathological adipose remodelling and metabolic disease. In both species, disproportionate effects on leptin secretion may relate to cell autonomous induction of the integrated stress response.


Asunto(s)
Resistencia a la Insulina , Lipodistrofia , Humanos , Animales , Ratones , Leptina/metabolismo , Adiponectina/metabolismo , Tejido Adiposo/metabolismo , Obesidad/metabolismo , Hidrolasas/metabolismo , Lipodistrofia/genética , Lipodistrofia/metabolismo , Mitocondrias/metabolismo
8.
Mol Metab ; 65: 101589, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36064109

RESUMEN

OBJECTIVES: Obesity in humans and mice is associated with elevated levels of two hormones responsive to cellular stress, namely GDF15 and FGF21. Over-expression of each of these is associated with weight loss and beneficial metabolic changes but where they are secreted from and what they are required for physiologically in the context of overfeeding remains unclear. METHODS: Here we used tissue selective knockout mouse models and human transcriptomics to determine the source of circulating GDF15 in obesity. We then generated and characterized the metabolic phenotypes of GDF15/FGF21 double knockout mice. RESULTS: Circulating GDF15 and FGF21 are both largely derived from the liver, rather than adipose tissue or skeletal muscle, in obese states. Combined whole body deletion of FGF21 and GDF15 does not result in any additional weight gain in response to high fat feeding but it does result in significantly greater hepatic steatosis and insulin resistance than that seen in GDF15 single knockout mice. CONCLUSIONS: Collectively the data suggest that overfeeding activates a stress response in the liver which is the major source of systemic rises in GDF15 and FGF21. These hormones then activate pathways which reduce this metabolic stress.


Asunto(s)
Hígado Graso , Resistencia a la Insulina , Animales , Peso Corporal , Hígado Graso/genética , Hígado Graso/metabolismo , Factores de Crecimiento de Fibroblastos , Factor 15 de Diferenciación de Crecimiento/genética , Hormonas , Humanos , Resistencia a la Insulina/genética , Ratones , Ratones Noqueados , Obesidad/genética , Obesidad/metabolismo
9.
Cell Rep ; 34(10): 108810, 2021 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-33691105

RESUMEN

Adipogenin (Adig) is an adipocyte-enriched transmembrane protein. Its expression is induced during adipogenesis in rodent cells, and a recent genome-wide association study associated body mass index (BMI)-adjusted leptin levels with the ADIG locus. In order to begin to understand the biological function of Adig, we studied adipogenesis in Adig-deficient cultured adipocytes and phenotyped Adig null (Adig-/-) mice. Data from Adig-deficient cells suggest that Adig is required for adipogenesis. In vivo, Adig-/- mice are leaner than wild-type mice when fed a high-fat diet and when crossed with Ob/Ob hyperphagic mice. In addition to the impact on fat mass accrual, Adig deficiency also reduces fat-mass-adjusted plasma leptin levels and impairs leptin secretion from adipose explants, suggesting an additional impact on the regulation of leptin secretion.


Asunto(s)
Tejido Adiposo/metabolismo , Leptina/metabolismo , Proteínas Nucleares/genética , Adipocitos/citología , Adipocitos/metabolismo , Adipogénesis , Adiponectina/genética , Adiponectina/metabolismo , Animales , Peso Corporal , Dieta Alta en Grasa , Femenino , Prueba de Tolerancia a la Glucosa , Leptina/sangre , Leptina/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Obesos , Proteínas Nucleares/deficiencia , Fenotipo , Proteína Desacopladora 1/genética , Proteína Desacopladora 1/metabolismo
10.
Cell Metab ; 29(3): 707-718.e8, 2019 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-30639358

RESUMEN

GDF15 is an established biomarker of cellular stress. The fact that it signals via a specific hindbrain receptor, GFRAL, and that mice lacking GDF15 manifest diet-induced obesity suggest that GDF15 may play a physiological role in energy balance. We performed experiments in humans, mice, and cells to determine if and how nutritional perturbations modify GDF15 expression. Circulating GDF15 levels manifest very modest changes in response to moderate caloric surpluses or deficits in mice or humans, differentiating it from classical intestinally derived satiety hormones and leptin. However, GDF15 levels do increase following sustained high-fat feeding or dietary amino acid imbalance in mice. We demonstrate that GDF15 expression is regulated by the integrated stress response and is induced in selected tissues in mice in these settings. Finally, we show that pharmacological GDF15 administration to mice can trigger conditioned taste aversion, suggesting that GDF15 may induce an aversive response to nutritional stress.


Asunto(s)
Ingestión de Energía/fisiología , Factor 15 de Diferenciación de Crecimiento/metabolismo , Adulto , Animales , Línea Celular , Dieta Alta en Grasa/métodos , Factor 15 de Diferenciación de Crecimiento/farmacología , Humanos , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Adulto Joven
11.
PLoS One ; 13(8): e0201310, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30110341

RESUMEN

Annexin A6 (AnxA6) controls cholesterol and membrane transport in endo- and exocytosis, and modulates triglyceride accumulation and storage. In addition, AnxA6 acts as a scaffolding protein for negative regulators of growth factor receptors and their effector pathways in many different cell types. Here we investigated the role of AnxA6 in the regulation of whole body lipid metabolism and insulin-regulated glucose homeostasis. Therefore, wildtype (WT) and AnxA6-knockout (KO) mice were fed a high-fat diet (HFD) for 17 weeks. During the course of HFD feeding, AnxA6-KO mice gained less weight compared to controls, which correlated with reduced adiposity. Systemic triglyceride and cholesterol levels of HFD-fed control and AnxA6-KO mice were comparable, with slightly elevated high density lipoprotein (HDL) and reduced triglyceride-rich lipoprotein (TRL) levels in AnxA6-KO mice. AnxA6-KO mice displayed a trend towards improved insulin sensitivity in oral glucose and insulin tolerance tests (OGTT, ITT), which correlated with increased insulin-inducible phosphorylation of protein kinase B (Akt) and ribosomal protein S6 kinase (S6) in liver extracts. However, HFD-fed AnxA6-KO mice failed to downregulate hepatic gluconeogenesis, despite similar insulin levels and insulin signaling activity, as well as expression profiles of insulin-sensitive transcription factors to controls. In addition, increased glycogen storage in livers of HFD- and chow-fed AnxA6-KO animals was observed. Together with an inability to reduce glucose production upon insulin exposure in AnxA6-depleted HuH7 hepatocytes, this implicates AnxA6 contributing to the fine-tuning of hepatic glucose metabolism with potential consequences for the systemic control of glucose in health and disease.


Asunto(s)
Anexina A6/deficiencia , Grasas de la Dieta/farmacología , Gluconeogénesis/efectos de los fármacos , Glucosa/metabolismo , Glucógeno/metabolismo , Hígado/metabolismo , Adiposidad/efectos de los fármacos , Animales , Anexina A6/metabolismo , Grasas de la Dieta/efectos adversos , Gluconeogénesis/genética , Glucosa/genética , Glucógeno/genética , Resistencia a la Insulina , Lípidos/sangre , Hígado/patología , Masculino , Ratones , Ratones Noqueados , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Quinasas S6 Ribosómicas/genética , Proteínas Quinasas S6 Ribosómicas/metabolismo
12.
Dev Cell ; 45(4): 481-495.e8, 2018 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-29754800

RESUMEN

Cell and organelle membranes consist of a complex mixture of phospholipids (PLs) that determine their size, shape, and function. Phosphatidylcholine (PC) is the most abundant phospholipid in eukaryotic membranes, yet how cells sense and regulate its levels in vivo remains unclear. Here we show that PCYT1A, the rate-limiting enzyme of PC synthesis, is intranuclear and re-locates to the nuclear membrane in response to the need for membrane PL synthesis in yeast, fly, and mammalian cells. By aligning imaging with lipidomic analysis and data-driven modeling, we demonstrate that yeast PCYT1A membrane association correlates with membrane stored curvature elastic stress estimates. Furthermore, this process occurs inside the nucleus, although nuclear localization signal mutants can compensate for the loss of endogenous PCYT1A in yeast and in fly photoreceptors. These data suggest an ancient mechanism by which nucleoplasmic PCYT1A senses surface PL packing defects on the inner nuclear membrane to control PC homeostasis.


Asunto(s)
Membrana Celular/química , Núcleo Celular/química , Citidililtransferasa de Colina-Fosfato/metabolismo , Elasticidad , Membrana Nuclear/química , Fosfatidilcolinas/metabolismo , Estrés Fisiológico , Animales , Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Citidililtransferasa de Colina-Fosfato/genética , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Drosophila melanogaster/metabolismo , Homeostasis , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Membrana Nuclear/genética , Membrana Nuclear/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/crecimiento & desarrollo , Saccharomyces cerevisiae/metabolismo
13.
Mol Cell Endocrinol ; 439: 419-430, 2017 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-27702590

RESUMEN

Lipid storage and adipokine secretion are critical features of adipocytes. Annexin A6 (AnxA6) is a lipid-binding protein regulating secretory pathways and its role in adiponectin release was examined. The siRNA-mediated AnxA6 knock-down in 3T3-L1 preadipocytes impaired proliferation, and differentiation of AnxA6-depleted cells to mature adipocytes was associated with higher soluble adiponectin and increased triglyceride storage. The latter was partly attributed to reduced lipolysis. Accordingly, AnxA6 overexpression in 3T3-L1 adipocytes lowered cellular triglycerides and adiponectin secretion. Indeed, serum adiponectin was increased in AnxA6 deficient mice. Expression analysis identified AnxA6 protein to be more abundant in intra-abdominal compared to subcutaneous adipose tissues of mice and men. AnxA6 protein levels increased in white adipose tissues of obese mice and here, levels were highest in subcutaneous fat. AnxA6 protein in adipocytes was upregulated by oxidative stress which might trigger AnxA6 induction in adipose tissues and contribute to impaired fat storage and adiponectin release.


Asunto(s)
Adipocitos/metabolismo , Adiponectina/metabolismo , Anexina A6/metabolismo , Metabolismo de los Lípidos , Células 3T3-L1 , Adipocitos/efectos de los fármacos , Adipogénesis/efectos de los fármacos , Animales , Trióxido de Arsénico , Arsenicales/farmacología , Quimiocinas/sangre , Ácidos Grasos/farmacología , Técnicas de Silenciamiento del Gen , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Hipoglucemiantes/farmacología , Insulina/metabolismo , Péptidos y Proteínas de Señalización Intercelular/sangre , Grasa Intraabdominal/efectos de los fármacos , Grasa Intraabdominal/metabolismo , Grasa Intraabdominal/patología , Gotas Lipídicas/efectos de los fármacos , Gotas Lipídicas/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Lipólisis/efectos de los fármacos , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/metabolismo , Obesidad/patología , Óxidos/farmacología , Transducción de Señal/efectos de los fármacos , Triglicéridos/metabolismo
14.
Br J Pharmacol ; 172(7): 1677-90, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25409976

RESUMEN

BACKGROUND AND PURPOSE: Annexin A6 (AnxA6) is a calcium-dependent phospholipid-binding protein that can be recruited to the plasma membrane to function as a scaffolding protein to regulate signal complex formation, endo- and exocytic pathways as well as distribution of cellular cholesterol. Here, we have investigated how AnxA6 influences the membrane order. EXPERIMENTAL APPROACH: We used Laurdan and di-4-ANEPPDHQ staining in (i) artificial membranes; (ii) live cells to investigate membrane packing and ordered lipid phases; and (iii) a super-resolution imaging (photoactivated localization microscopy, PALM) and Ripley's K second-order point pattern analysis approach to assess how AnxA6 regulates plasma membrane order domains and protein clustering. KEY RESULTS: In artificial membranes, purified AnxA6 induced a global increase in membrane order. However, confocal microscopy using di-4-ANEPPDHQ in live cells showed that cells expressing AnxA6, which reduces plasma membrane cholesterol levels and modifies the actin cytoskeleton meshwork, displayed a decrease in membrane order (∼15 and 30% in A431 and MEF cells respectively). PALM data from Lck10 and Src15 membrane raft/non-raft markers revealed that AnxA6 expression induced clustering of both raft and non-raft markers. Altered clustering of Lck10 and Src15 in cells expressing AnxA6 was also observed after cholesterol extraction with methyl-ß-cyclodextrin or actin cytoskeleton disruption with latrunculin B. CONCLUSIONS AND IMPLICATIONS: AnxA6-induced plasma membrane remodelling indicated that elevated AnxA6 expression decreased membrane order through the regulation of cellular cholesterol homeostasis and the actin cytoskeleton. This study provides the first evidence from live cells that support current models of annexins as membrane organizers.


Asunto(s)
Anexina A6/metabolismo , Membrana Celular/metabolismo , Animales , Línea Celular , Línea Celular Tumoral , Membrana Celular/química , Humanos , Lípidos/química , Ratones Noqueados , Microscopía Fluorescente
15.
Cell Rep ; 7(3): 883-97, 2014 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-24746815

RESUMEN

Inhibition of cholesterol export from late endosomes causes cellular cholesterol imbalance, including cholesterol depletion in the trans-Golgi network (TGN). Here, using Chinese hamster ovary (CHO) Niemann-Pick type C1 (NPC1) mutant cell lines and human NPC1 mutant fibroblasts, we show that altered cholesterol levels at the TGN/endosome boundaries trigger Syntaxin 6 (Stx6) accumulation into VAMP3, transferrin, and Rab11-positive recycling endosomes (REs). This increases Stx6/VAMP3 interaction and interferes with the recycling of αVß3 and α5ß1 integrins and cell migration, possibly in a Stx6-dependent manner. In NPC1 mutant cells, restoration of cholesterol levels in the TGN, but not inhibition of VAMP3, restores the steady-state localization of Stx6 in the TGN. Furthermore, elevation of RE cholesterol is associated with increased amounts of Stx6 in RE. Hence, the fine-tuning of cholesterol levels at the TGN-RE boundaries together with a subset of cholesterol-sensitive SNARE proteins may play a regulatory role in cell migration and invasion.


Asunto(s)
Colesterol/metabolismo , Endosomas/metabolismo , Proteínas Qa-SNARE/metabolismo , Red trans-Golgi/metabolismo , Animales , Células CHO , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Movimiento Celular , Cricetinae , Cricetulus , Humanos , Integrina alfa5beta1/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Proteína Niemann-Pick C1 , Unión Proteica , Transporte de Proteínas , Proteínas Qa-SNARE/química , Receptores de Vitronectina/metabolismo , Proteínas SNARE/metabolismo , Proteína 3 de Membrana Asociada a Vesículas/química , Proteína 3 de Membrana Asociada a Vesículas/metabolismo , Proteínas de Unión al GTP rab/metabolismo
16.
PLoS One ; 8(4): e62667, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23634230

RESUMEN

Signal transduction modulates expression and activity of cholesterol transporters. We recently demonstrated that the Ras/mitogen-activated protein kinase (MAPK) signaling cascade regulates protein stability of Scavenger Receptor BI (SR-BI) through Proliferator Activator Receptor (PPARα) -dependent degradation pathways. In addition, MAPK (Mek/Erk 1/2) inhibition has been shown to influence liver X receptor (LXR) -inducible ATP Binding Cassette (ABC) transporter ABCA1 expression in macrophages. Here we investigated if Ras/MAPK signaling could alter expression and activity of ABCA1 and ABCG1 in steroidogenic and hepatic cell lines. We demonstrate that in Chinese Hamster Ovary (CHO) cells and human hepatic HuH7 cells, extracellular signal-regulated kinase 1/2 (Erk1/2) inhibition reduces PPARα-inducible ABCA1 protein levels, while ectopic expression of constitutively active H-Ras, K-Ras and MAPK/Erk kinase 1 (Mek1) increases ABCA1 protein expression, respectively. Furthermore, Mek1/2 inhibitors reduce ABCG1 protein levels in ABCG1 overexpressing CHO cells (CHO-ABCG1) and human embryonic kidney 293 (HEK293) cells treated with LXR agonist. This correlates with Mek1/2 inhibition reducing ABCG1 cell surface expression and decreasing cholesterol efflux onto High Density Lipoproteins (HDL). Real Time reverse transcriptase polymerase chain reaction (RT-PCR) and protein turnover studies reveal that Mek1/2 inhibitors do not target transcriptional regulation of ABCA1 and ABCG1, but promote ABCA1 and ABCG1 protein degradation in HuH7 and CHO cells, respectively. In line with published data from mouse macrophages, blocking Mek1/2 activity upregulates ABCA1 and ABCG1 protein levels in human THP1 macrophages, indicating opposite roles for the Ras/MAPK pathway in the regulation of ABC transporter activity in macrophages compared to steroidogenic and hepatic cell types. In summary, this study suggests that Ras/MAPK signaling modulates PPARα- and LXR-dependent protein degradation pathways in a cell-specific manner to regulate the expression levels of ABCA1 and ABCG1 transporters.


Asunto(s)
Transportador 1 de Casete de Unión a ATP/metabolismo , Transportadoras de Casetes de Unión a ATP/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Proteolisis/efectos de los fármacos , Transportador 1 de Casete de Unión a ATP/química , Transportador 1 de Casete de Unión a ATP/genética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1 , Transportadoras de Casetes de Unión a ATP/química , Transportadoras de Casetes de Unión a ATP/genética , Animales , Transporte Biológico/efectos de los fármacos , Células CHO , Colesterol/metabolismo , Cricetinae , Cricetulus , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Estabilidad Proteica/efectos de los fármacos , Proteínas ras/metabolismo
17.
Curr Biol ; 23(15): 1489-96, 2013 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-23871243

RESUMEN

Lipid droplets (LDs) are dynamic organelles that collect, store, and supply lipids [1]. LDs have a central role in the exchange of lipids occurring between the cell and the environment and provide cells with substrates for energy metabolism, membrane synthesis, and production of lipid-derived molecules such as lipoproteins or hormones. However, lipid-derived metabolites also cause progressive lipotoxicity [2], accumulation of reactive oxygen species (ROS), endoplasmic reticulum stress, mitochondrial malfunctioning, and cell death [2]. Intracellular accumulation of LDs is a hallmark of prevalent human diseases, including obesity, steatosis, diabetes, myopathies, and arteriosclerosis [3]. Indeed, nonalcoholic fatty liver disease is the most common cause of abnormal hepatic function among adults [4, 5]. Lipotoxicity gradually promotes cellular ballooning and disarray, megamitochondria, accumulation of Mallory's hyaline in hepatocytes, and inflammation, fibrosis, and cirrhosis in the liver. Here, using confocal microscopy, serial-block-face scanning electron microscopy, and flow cytometry, we show that LD accumulation is heterogeneous within a cell population and follows a positive skewed distribution. Lipid availability and fluctuations in biochemical networks controlling lipolysis, fatty acid oxidation, and protein synthesis contribute to cell-to-cell heterogeneity. Critically, this reversible variability generates a subpopulation of cells that effectively collect and store lipids. This high-lipid subpopulation accumulates more LDs and more ROS and reduces the risk of lipotoxicity to the population without impairing overall lipid homeostasis, since high-lipid cells can supply stored lipids to the other cells. In conclusion, we demonstrate fat storage compartmentalization within a cell population and propose that this is a protective social organization to reduce lipotoxicity.


Asunto(s)
Hepatocitos/citología , Metabolismo de los Lípidos , Lípidos/química , Animales , Compuestos de Boro/metabolismo , Ácidos Grasos/metabolismo , Citometría de Flujo , Hepatocitos/metabolismo , Lípidos/fisiología , Ratones Endogámicos C57BL , Especies Reactivas de Oxígeno/metabolismo
18.
Mol Biol Cell ; 22(21): 4108-23, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22039070

RESUMEN

Cholesterol regulates plasma membrane (PM) association and functioning of syntaxin-4 and soluble N-ethylmaleimide-sensitive fusion protein 23 (SNAP23) in the secretory pathway. However, the molecular mechanism and cellular cholesterol pools that determine the localization and assembly of these target membrane SNAP receptors (t-SNAREs) are largely unknown. We recently demonstrated that high levels of annexin A6 (AnxA6) induce accumulation of cholesterol in late endosomes, thereby reducing cholesterol in the Golgi and PM. This leads to an impaired supply of cholesterol needed for cytosolic phospholipase A(2) (cPLA(2)) to drive Golgi vesiculation and caveolin transport to the cell surface. Using AnxA6-overexpressing cells as a model for cellular cholesterol imbalance, we identify impaired cholesterol egress from late endosomes and diminution of Golgi cholesterol as correlating with the sequestration of SNAP23/syntaxin-4 in Golgi membranes. Pharmacological accumulation of late endosomal cholesterol and cPLA(2) inhibition induces a similar phenotype in control cells with low AnxA6 levels. Ectopic expression of Niemann-Pick C1 (NPC1) or exogenous cholesterol restores the location of SNAP23 and syntaxin-4 within the PM. Importantly, AnxA6-mediated mislocalization of these t-SNAREs correlates with reduced secretion of cargo via the SNAP23/syntaxin-4-dependent constitutive exocytic pathway. We thus conclude that inhibition of late endosomal export and Golgi cholesterol depletion modulate t-SNARE localization and functioning along the exocytic pathway.


Asunto(s)
Colesterol/metabolismo , Endosomas/metabolismo , Aparato de Golgi/metabolismo , Multimerización de Proteína , Transporte de Proteínas , Proteínas SNARE/metabolismo , Animales , Anexina A6/metabolismo , Células CHO , Caveolina 1/metabolismo , Línea Celular Tumoral , Cricetinae , Fibronectinas/metabolismo , Humanos , Glicoproteínas de Membrana/metabolismo , Microdominios de Membrana/metabolismo , Microscopía Fluorescente , Fosfolipasas A2 Citosólicas/metabolismo , Proteínas Qa-SNARE/metabolismo , Proteínas Qb-SNARE/metabolismo , Proteínas Qc-SNARE/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA