Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Diabetologia ; 60(2): 324-335, 2017 02.
Article in English | MEDLINE | ID: mdl-27866222

ABSTRACT

AIMS/HYPOTHESIS: Septins are newly identified members of the cytoskeleton that have been proposed as biomarkers of a number of diseases. However, septins have not been characterised in adipose tissue and their relationship with obesity and insulin resistance remains unknown. Herein, we characterised a member of this family, septin 11 (SEPT11), in human adipose tissue and analysed its potential involvement in the regulation of adipocyte metabolism. METHODS: Gene and protein expression levels of SEPT11 were analysed in human adipose tissue. SEPT11 distribution was evaluated by immunocytochemistry, electron microscopy and subcellular fractionation techniques. Glutathione S-transferase (GST) pull-down, immunoprecipitation and yeast two-hybrid screening were used to identify the SEPT11 interactome. Gene silencing was used to assess the role of SEPT11 in the regulation of insulin signalling and lipid metabolism in adipocytes. RESULTS: We demonstrate the expression of SEPT11 in human adipocytes and its upregulation in obese individuals, with SEPT11 mRNA content positively correlating with variables of insulin resistance in subcutaneous adipose tissue. SEPT11 content was regulated by lipogenic, lipolytic and proinflammatory stimuli in human adipocytes. SEPT11 associated with caveolae in mature adipocytes and interacted with both caveolin-1 and the intracellular fatty acid chaperone, fatty acid binding protein 5 (FABP5). Lipid loading of adipocytes caused the association of the three proteins with the surface of lipid droplets. SEPT11 silencing impaired insulin signalling and insulin-induced lipid accumulation in adipocytes. CONCLUSIONS/INTERPRETATION: Our findings support a role for SEPT11 in lipid traffic and metabolism in adipocytes and open new avenues for research on the control of lipid storage in obesity and insulin resistance.


Subject(s)
Adipocytes/metabolism , Obesity/metabolism , Septins/metabolism , Adult , Caveolae/metabolism , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Female , Gene Silencing/physiology , Humans , Immunoblotting , Immunohistochemistry , Insulin Resistance/genetics , Insulin Resistance/physiology , Lipid Metabolism/genetics , Lipid Metabolism/physiology , Male , Middle Aged , Obesity/genetics , Real-Time Polymerase Chain Reaction , Septins/genetics
2.
J Biol Chem ; 288(5): 3112-25, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23255609

ABSTRACT

Adiponectin binds to two widely expressed receptors (AdipoR1 and AdipoR2) that contain seven transmembrane domains but, unlike G-protein coupled receptors, present an extracellular C terminus and a cytosolic N terminus. Recently, AdipoR1 was found to associate in high order complexes. However, it is still unknown whether AdipoR2 may also form homomers or heteromers with AdipoR1 or if such interactions may be functionally relevant. Herein, we have analyzed the oligomerization pattern of AdipoRs by FRET and immunoprecipitation and evaluated both the internalization of AdipoRs in response to various adiponectin isoforms and the effect of adiponectin binding to different AdipoR combinations on AMP-activated protein kinase phosphorylation and peroxisome proliferator-activated receptor α activation. Transfection of HEK293AD cells with AdipoR1 and AdipoR2 showed that both receptors colocalize at both the plasma membrane and the endoplasmic reticulum. Co-transfection with the different AdipoR pairs yielded high FRET efficiencies in non-stimulated cells, which indicates that AdipoR1 and AdipoR2 form homo- and heteromeric complexes under resting conditions. Live FRET imaging suggested that both homo- and heteromeric AdipoR complexes dissociate in response to adiponectin, but heteromers separate faster than homomers. Finally, phosphorylation of AMP-activated protein kinase in response to adiponectin was delayed in cells wherein heteromer formation was favored. In sum, our findings indicate that AdipoR1 and AdipoR2 form homo- and heteromers that present unique interaction behaviors and signaling properties. This raises the possibility that the pleiotropic, tissue-dependent functions of adiponectin depend on the expression levels of AdipoR1 and AdipoR2 and, therefore, on the steady-state proportion of homo- and heteromeric complexes.


Subject(s)
Intracellular Space/metabolism , Protein Multimerization , Receptors, Adiponectin/metabolism , Signal Transduction , AMP-Activated Protein Kinases/metabolism , Adiponectin/pharmacology , Endocytosis/drug effects , Fluorescence Resonance Energy Transfer , HEK293 Cells , Hep G2 Cells , Humans , Intracellular Space/drug effects , Ligands , Luminescent Proteins/metabolism , PPAR alpha/metabolism , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Multimerization/drug effects , Protein Transport/drug effects , Recombinant Proteins/metabolism , Signal Transduction/drug effects , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Time Factors
3.
Mol Cell Proteomics ; 10(11): M111.008094, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21828285

ABSTRACT

Lipodystrophy is a major disease involving severe alterations of adipose tissue distribution and metabolism. Mutations in genes encoding the nuclear envelope protein lamin A or its processing enzyme, the metalloproteinase Zmpste24, cause diverse human progeroid syndromes that are commonly characterized by a selective loss of adipose tissue. Similarly to humans, mice deficient in Zmpste24 accumulate prelamin A and display phenotypic features of accelerated aging, including lipodystrophy. Herein, we report the proteome and phosphoproteome of adipose tissue as well as serum metabolome in lipodystrophy by using Zmpste24(-/-) mice as experimental model. We show that Zmpste24 deficiency enhanced lipolysis, fatty acid biogenesis and ß-oxidation as well as decreased fatty acid re-esterification, thus pointing to an increased partitioning of fatty acid toward ß-oxidation and away from storage that likely underlies the observed size reduction of Zmpste24-null adipocytes. Besides the mitochondrial proteins related to lipid metabolism, other protein networks related to mitochondrial function, including those involved in tricarboxylic acid cycle and oxidative phosphorylation, were up-regulated in Zmpste24(-/-) mice. These results, together with the observation of an increased mitochondrial response to oxidative stress, support the relationship between defective prelamin A processing and mitochondrial dysfunction and highlight the relevance of oxidative damage in lipoatrophy and aging. We also show that absence of Zmpste24 profoundly alters the processing of the cytoskeletal protein vimentin and identify a novel protein dysregulated in lipodystrophy, High-Mobility Group Box-1 Protein. Finally, we found several lipid derivates with important roles in energy balance, such as Lysophosphatidylcholine or 2-arachidonoylglycerol, to be dysregulated in Zmpste24(-/-) serum. Together, our findings in Zmpste24(-/-) mice may be useful to unveil the mechanisms underlying adipose tissue dysfunction and its overall contribution to body homeostasis in progeria and other lipodystrophy syndromes as well as to develop novel strategies to prevent or ameliorate these diseases.


Subject(s)
Aging, Premature/metabolism , Lipodystrophy/metabolism , Membrane Proteins/genetics , Metalloendopeptidases/genetics , Mitochondria/metabolism , Proteome/metabolism , Vimentin/metabolism , Aging, Premature/genetics , Animals , Biomarkers/blood , Blood Glucose/metabolism , Gene Expression Regulation , HMGB1 Protein/genetics , HMGB1 Protein/metabolism , Intra-Abdominal Fat/metabolism , Intra-Abdominal Fat/pathology , Lamin Type A , Lipid Metabolism , Lipids/blood , Lipodystrophy/genetics , Male , Metabolome , Mice , Mice, Knockout , Mitochondria/enzymology , Nuclear Proteins/metabolism , Oxidation-Reduction , Phosphoproteins/metabolism , Protein Interaction Maps , Protein Precursors/metabolism , Proteome/genetics
4.
Biochem J ; 443(2): 387-96, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22250954

ABSTRACT

Golgi-associated long coiled-coil proteins, often referred to as golgins, are involved in the maintenance of the structural organization of the Golgi apparatus and the regulation of membrane traffic events occurring in this organelle. Little information is available on the contribution of golgins to Golgi function in cells specialized in secretion such as endocrine cells or neurons. In the present study, we characterize the intracellular distribution as well as the biochemical and functional properties of a novel long coiled-coil protein present in neuroendocrine tissues, NECC1 (neuroendocrine long coiled-coil protein 1). The present study shows that NECC1 is a peripheral membrane protein displaying high stability to detergent extraction, which distributes across the Golgi apparatus in neuroendocrine cells. In addition, NECC1 partially localizes to post-Golgi carriers containing secretory cargo in PC12 cells. Overexpression of NECC1 resulted in the formation of juxtanuclear aggregates together with a slight fragmentation of the Golgi and a decrease in K+-stimulated hormone release. In contrast, NECC1 silencing did not alter Golgi architecture, but enhanced K+-stimulated hormone secretion in PC12 cells. In all, the results of the present study identify NECC1 as a novel component of the Golgi matrix and support a role for this protein as a negative modulator of the regulated trafficking of secretory cargo in neuroendocrine cells.


Subject(s)
Golgi Apparatus/metabolism , Homeodomain Proteins/metabolism , Membrane Proteins/metabolism , Animals , Biological Transport , Gene Silencing , Homeodomain Proteins/genetics , Membrane Proteins/genetics , Neuroendocrine Cells/metabolism , PC12 Cells , Rats
5.
Am J Physiol Endocrinol Metab ; 303(11): E1325-34, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-23032684

ABSTRACT

Somatostatin (SST) and its related peptide cortistatin (CORT) exert their multiple actions through binding to the SST receptor (sst) family, generally considered to comprise five G protein-coupled receptors with seven transmembrane domains (TMD), named sst1-sst5, plus a splice sst2B variant. However, we recently discovered that human and rodent sst5 gene expression also generates, through noncanonical alternative splicing, novel truncated albeit functional sst5 variants with less than seven TMD. Here, we cloned and characterized for the first time the porcine wild-type sst5 (psst5, full-length) and identified two novel truncated psst5 variants with six and three TMD, thus termed psst5TMD6 and psst5TMD3, respectively. In line with that observed in human and rodent truncated sst5 variants, psst5TMD6 and psst5TMD3 are functional (e.g., activate calcium signaling), selectively respond to SST and CORT, respectively, and exhibit specific tissue expression profiles that differ from full-length psst5 and often overlaps with psst2 expression. Moreover, fluorescence resonance energy transfer analysis shows that psst5 truncated variants physically interact with psst2, thereby altering their localization at the plasma membrane and specifically disrupting the cellular response to SST and/or CORT. These results represent the first characterization of a key porcine SST receptor, psst5, and, together with our previous results, provide strong evidence that alternative splicing-derived, truncated sst5 variants with distinct functional capacities exist in the mammalian lineage, where they can act as dominant-negative receptors, by interacting directly with long, seven TMD variants, potentially contributing to modulate normal and pathological SST and CORT signaling.


Subject(s)
Calcium Signaling/physiology , Protein Structure, Tertiary/physiology , Receptors, Somatostatin/physiology , Signal Transduction/physiology , Alternative Splicing , Animals , CHO Cells , Cloning, Molecular , Cricetinae , Humans , Peptide Fragments , Protein Engineering , Protein Isoforms/physiology , Swine , Tissue Distribution
6.
Histochem Cell Biol ; 138(3): 489-501, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22614950

ABSTRACT

In the present study, we analyze the effects of ethanol on the Golgi structure and membrane transport in differentiated PC12 cells, which are used as a model of neurons. Chronic exposure to moderate doses of ethanol induces Golgi fragmentation, a common characteristic of many neurodegenerative diseases. Alcohol impaired the lateral linking of stacks without causing microtubule damage. Extensive immunocytochemical and western blot analyses of representative Golgi proteins showed that few, but important, proteins are significantly affected. Thus, alcohol exposure induced a significant ER-to-Golgi transport delay, the retention of the GTPase Rab1 in the Golgi membranes and the accumulation of tethering factor p115 in the cytosol. These modifications would explain the observed fragmentation. The amount of p115 and the stacking protein GRASP65 increased in alcohol-treated cells, which might be a mechanism to reverse Golgi damage. Importantly, the overexpression of GTP-tagged Rab1 but not of a dominant-negative Rab1 mutant, restored the Golgi morphology, suggesting that this protein is the main target of alcohol. Taken together, our results support the view that alcohol and neurodegenerative diseases such as Parkinson have similar effects on intracellular trafficking and provide new clues on the neuropathology of alcoholism.


Subject(s)
Cell Differentiation , Endoplasmic Reticulum/metabolism , Ethanol/pharmacology , Golgi Apparatus/metabolism , rab1 GTP-Binding Proteins/genetics , Animals , Golgi Matrix Proteins , Membrane Proteins/metabolism , PC12 Cells , Protein Transport , Rats , Vesicular Transport Proteins/metabolism , rab1 GTP-Binding Proteins/metabolism
7.
Gen Comp Endocrinol ; 175(1): 1-9, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-21907200

ABSTRACT

The regulated secretory pathway is a hallmark of endocrine and neuroendocrine cells. This process comprises different sequential steps, including ER-associated protein synthesis, ER-to-Golgi protein transport, Golgi-associated posttranslational modification, sorting and packing of secretory proteins into carrier granules, cytoskeleton-based granule transport towards the plasma membrane and tethering, docking and fusion of granules with specialized releasing zones in the plasma membrane. Each one of these steps is tightly regulated by a large number of factors that function in a spatially and temporarily coordinated fashion. During the past three decades, much effort has been devoted to characterize the precise role of the yet-known proteins participating in the different steps of this process and to identify new regulatory factors in order to obtain a unifying picture of the secretory pathway. In spite of this and given the enormous complexity of the process, certain steps are not fully understood yet and many players remain to be identified. In this review, we offer a summary of the current knowledge on the main molecular mechanisms that govern and ensure the correct release of secretory proteins. In addition, we have integrated the advance on the field made possible by studies carried out in non-mammalian vertebrates, which, although not very numerous, have substantially contributed to acquire a mechanistic understanding of the regulated secretory pathway.


Subject(s)
Endocrine System/physiology , Neuroendocrine Cells/physiology , Secretory Pathway/physiology , Animals , Anura , Endoplasmic Reticulum/physiology , Golgi Apparatus/physiology , Humans , Secretory Vesicles/physiology
8.
Proteomics ; 10(18): 3356-66, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20706982

ABSTRACT

Adipose tissue represents a complex tissue both in terms of its cellular composition, as it includes mature adipocytes and the various cell types comprising the stromal-vascular fraction (SVF), and in relation to the distinct biochemical, morphological and functional characteristics according to its anatomical location. Herein, we have characterized the proteomic profile of both mature adipocyte and SVF from human visceral adipose tissue (VAT) and subcutaneous adipose tissue (SAT) fat depots in order to unveil differences in the expression of proteins which may underlie the distinct association of VAT and SAT to several pathologies. Specifically, 24 proteins were observed to be differentially expressed between SAT SVF versus VAT SVF from lean individuals. Immunoblotting and RT-PCR analysis confirmed the differential regulation of the nuclear envelope proteins lamin A/C, the membrane-cytoskeletal linker ezrin and the enzyme involved in retinoic acid production, aldehyde dehydrogenase 1A2, in the two fat depots. In sum, the observation that proteins with important cell functions are differentially distributed between VAT and SAT and their characterization as components of SVF or mature adipocytes pave the way for future research on the molecular basis underlying diverse adipose tissue-related pathologies such as metabolic syndrome or lipodystrophy.


Subject(s)
Intra-Abdominal Fat/chemistry , Subcutaneous Fat, Abdominal/chemistry , Aged , Gene Expression Regulation , Humans , Intra-Abdominal Fat/metabolism , Middle Aged , RNA, Messenger/genetics , Subcutaneous Fat, Abdominal/metabolism
9.
J Clin Endocrinol Metab ; 93(6): 2269-76, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18349058

ABSTRACT

CONTEXT: Rab proteins regulate the sequential steps of intracellular membrane transport. Alterations of these GTPases and their associated proteins are emerging as the underlying cause for several human diseases involving dysregulated secretory activities. OBJECTIVE: Herein we investigated the role of Rab18, which negatively regulates hormone secretion by interacting with secretory granules, in relation to the altered functioning of tumoral pituitary somatotropes causing acromegaly. PATIENTS: A total of 18 patients diagnosed with pituitary tumors causing acromegaly (nine patients) or nonfunctioning adenomas (nine patients) underwent endoscopic transsphenoidal surgery. Adenomas were subsequently processed to evaluate Rab18 production in relation to GH secretion. RESULTS: We found that somatotropinoma cells are characterized by a high secretory activity concomitantly with a remarkably reduced Rab18 expression (15%) and protein content levels (30%), as compared with cells from nonfunctioning pituitary adenomas derived from patients with normal or reduced GH plasma levels (100%). Furthermore, immunoelectron microscopy revealed that Rab18 association with the surface of GH-containing secretory granules was significantly lower in somatotropes from acromegalies than nonfunctioning pituitary adenomas. Finally, we provide evidence that modulation of Rab18 gene expression can revert substantially the hypersecretory activity of cells because Rab18 overexpression reduced by 40% the capacity of cells from acromegalies to respond to GHRH stimulation. CONCLUSION: These results suggest that molecular alterations affecting individual components of the secretory granule traffic machinery can contribute to maintain a high level of GH in plasma. Accordingly, Rab18 constitutes a valuable target as a diagnostic, prognostic, and/or therapeutic tool for human acromegaly.


Subject(s)
Acromegaly/genetics , Adenoma/genetics , Growth Hormone-Secreting Pituitary Adenoma/genetics , Human Growth Hormone/metabolism , rab GTP-Binding Proteins/genetics , Acromegaly/etiology , Adenoma/metabolism , Cell Membrane/metabolism , Gene Expression Regulation, Neoplastic/physiology , Growth Hormone-Secreting Pituitary Adenoma/metabolism , Humans , RNA, Messenger/metabolism , Secretory Vesicles/metabolism , Somatotrophs/metabolism , Tissue Distribution , Transfection , Tumor Cells, Cultured , rab GTP-Binding Proteins/metabolism , rab GTP-Binding Proteins/physiology
10.
Peptides ; 29(5): 711-20, 2008 May.
Article in English | MEDLINE | ID: mdl-18289730

ABSTRACT

The UII and urotensin II-related peptide (URP) genes belong to the same superfamily as the somatostatin gene. It has been previously shown that somatostatin activates the UII-receptor (UTR). In contrast, the possible interaction between UII and URP and somatostatin receptors has remained scarcely analyzed. Herein, we have investigated the effects of UII and URP on cell proliferation and free cytosolic Ca2+ concentration ([Ca2+]i) in CHO-K1 cells stably expressing the porcine somatostatin receptor subtypes sst2 and sst5. Results show that both UII and URP induce stimulation of cell proliferation mediated by sst2 receptors and UII provokes inhibition of cell proliferation mediated by sst5 receptors. UII and URP also provoked an increase of [Ca2+]i in both sst2- and sst5-transfected cells. Together, our present data demonstrate that UII and URP directly activate sst2 and sst5 and thus mimic the effect of somatostatin on its cognate receptors.


Subject(s)
Cell Proliferation , Peptide Hormones/metabolism , Receptors, Somatostatin/metabolism , Urotensins/metabolism , Animals , CHO Cells , Calcium/metabolism , Cricetinae , Cricetulus , Humans , Quinolines/metabolism , Receptors, Somatostatin/agonists , Receptors, Somatostatin/genetics , Somatostatin/agonists , Somatostatin/metabolism , Swine , Urea/analogs & derivatives , Urea/metabolism
11.
Endocrinology ; 148(1): 411-21, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17053026

ABSTRACT

Somatostatin (SRIF) exerts its multiple actions, including inhibition of GH secretion and of tumoral growth, through a family of five receptor subtypes (sst1-sst5). We recently reported that an sst2-selective agonist markedly decreases GH release from pig somatotropes, suggesting important roles for this scarcely explored receptor, psst2. Here, functional expression of psst2 in Chinese hamster ovary-K1 and human embryonic kidney-293-AD cell lines was employed to determine its pharmacological features and functional ability to reduce cAMP, and to examine its homodimerization and internalization dynamics in real time in single living cells. Results show that psst2 is a high-affinity receptor (dissociation constant = 0.27 nM) displaying a typical sst2 profile (nM affinity for SRIF-14> or =SRIF-28>cortistatin>MK678>octreotide) and high selectivity (EC(50) = 1.1 nM) for the sst2 agonist l-779,976, but millimolar or undetectable affinity to other sst-specific agonists (sst3>sst1>sst5>>>sst4). Accordingly, SRIF dose-dependently inhibited forskolin-stimulated cAMP with high potency (EC(50) = 6.55 pm) and modest efficacy (maximum 29.1%) via psst2. Cotransfection of human embryonic kidney-293 and Chinese hamster ovary-K1 cells with two receptor constructs modified with distinct fluorescent tags (psst2-YFP/psst2-CFP) enabled fluorescence resonance energy transfer measurement of physical interaction between psst2 receptors and also receptor internalization in single living cells. This revealed that under basal conditions, psst2 forms constitutive homodimers/homomultimers, which dissociate immediately (11 sec) upon SRIF binding. Interestingly, contrary to human sst2, psst2 rapidly reassociates (110.5 sec) during a subsequent process that temporally overlaps with receptor internalization (half-maximal = 95.1 sec). Therefore, psst2 is a potent inhibitory receptor displaying a unique set of interrelated dynamic features of agonist-dependent dimerization, dissociation, internalization, and reassociation, a cascade of events that might be critical for receptor function.


Subject(s)
Pituitary Gland/cytology , Pituitary Gland/physiology , Receptors, Somatostatin/chemistry , Receptors, Somatostatin/metabolism , Animals , Antineoplastic Agents, Hormonal/pharmacology , CHO Cells , Calcium/metabolism , Cloning, Molecular , Cricetinae , Cricetulus , Cyclic AMP/metabolism , Dimerization , Endocytosis/physiology , Female , Fluorescence Resonance Energy Transfer , Hormone Antagonists/pharmacology , Neuropeptides/pharmacology , Octreotide/pharmacology , Peptides, Cyclic/pharmacology , Radioligand Assay , Receptors, Somatostatin/agonists , Receptors, Somatostatin/genetics , Somatostatin/analogs & derivatives , Somatostatin/metabolism , Somatostatin/pharmacology , Sus scrofa
12.
Cell Metab ; 26(1): 212-229.e12, 2017 Jul 05.
Article in English | MEDLINE | ID: mdl-28683288

ABSTRACT

Thyroid hormones (THs) act in the brain to modulate energy balance. We show that central triiodothyronine (T3) regulates de novo lipogenesis in liver and lipid oxidation in brown adipose tissue (BAT) through the parasympathetic (PSNS) and sympathetic nervous system (SNS), respectively. Central T3 promotes hepatic lipogenesis with parallel stimulation of the thermogenic program in BAT. The action of T3 depends on AMP-activated protein kinase (AMPK)-induced regulation of two signaling pathways in the ventromedial nucleus of the hypothalamus (VMH): decreased ceramide-induced endoplasmic reticulum (ER) stress, which promotes BAT thermogenesis, and increased c-Jun N-terminal kinase (JNK) activation, which controls hepatic lipid metabolism. Of note, ablation of AMPKα1 in steroidogenic factor 1 (SF1) neurons of the VMH fully recapitulated the effect of central T3, pointing to this population in mediating the effect of central THs on metabolism. Overall, these findings uncover the underlying pathways through which central T3 modulates peripheral metabolism.


Subject(s)
Energy Metabolism , Hypothalamus/metabolism , Mitogen-Activated Protein Kinase 8/metabolism , Signal Transduction , Thyroid Hormones/metabolism , Adipose Tissue, Brown/metabolism , Animals , Lipid Metabolism , Liver/metabolism , Male , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Thermogenesis , Triiodothyronine/metabolism
13.
J Clin Endocrinol Metab ; 91(6): 2225-31, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16551736

ABSTRACT

CONTEXT: In Cushing's disease, ACTH hypersecretion by pituitary corticotrope adenoma cells and resulting hypercortisolism is accompanied by a severely blunted GH secretory response. Interestingly, in Cushing's disease, ghrelin markedly increases plasma ACTH, whereas its stimulatory action on GH secretion is reduced. Although the reported expression of ghrelin receptors (GHS-R) in corticotrope tumors offers a potential mechanism for ghrelin-induced ACTH hypersecretion, studies on the direct effects of synthetic GH secretagogues on corticotropinoma cells offered contradictory results. OBJECTIVE AND DESIGN: To evaluate the direct action of ghrelin on corticotropinoma cells from two patients with Cushing's disease, we measured its effect on free cytosolic calcium concentration ([Ca(2+)](i)). Additionally, expression of GHS-R and its ligand ghrelin was examined in these cells and in five additional corticotropinomas. RESULTS: Ghrelin (10(-6) m) induced a marked [Ca(2+)](i) increase in 89.5% (case 1; n = 19 cells) and 85% (case 2; n = 13 cells) of corticotropinoma cells. Moreover, RT-PCR showed that expression of GHS-R isoforms is accompanied by that of ghrelin in all seven corticotrope adenomas examined. Importantly, double immunogold electron microscopy revealed that ghrelin is costored within ACTH secretory vesicles in densely granulated adenomatous corticotropes. CONCLUSIONS: These results constitute the first demonstration that ghrelin acts directly on corticotrope tumor cells derived from patients with Cushing's disease. The presence of ghrelin and GHS-R suggests that pituitary ghrelin may play an autocrine/paracrine role in regulating ACTH release in Cushing's disease. Our findings provide a plausible cellular basis for the exaggerated ACTH response to ghrelin in Cushing's disease and suggest novel research strategies to develop medical treatments for this disease.


Subject(s)
Adenoma/metabolism , Adrenocorticotropic Hormone/metabolism , Peptide Hormones/physiology , Pituitary Neoplasms/metabolism , Adult , Calcium/metabolism , Female , Fluorescent Antibody Technique , Ghrelin , Humans , Immunohistochemistry , Pro-Opiomelanocortin/genetics , RNA, Messenger/analysis , Receptors, G-Protein-Coupled/genetics , Receptors, Ghrelin
14.
BMC Cell Biol ; 7: 23, 2006 May 23.
Article in English | MEDLINE | ID: mdl-16719903

ABSTRACT

BACKGROUND: There exists abundant molecular and ultra-structural evidence to suggest that cytoplasmic actin can physically interact with the nuclear envelope (NE) membrane system. However, this interaction has yet to be characterised in living interphase cells. RESULTS: Using a fluorescent conjugate of the actin binding drug cytochalasin D (CD-BODIPY) we provide evidence that polymerising actin accumulates in vicinity to the NE. In addition, both transiently expressed fluorescent actin and cytoplasmic micro-injection of fluorescent actin resulted in accumulation of actin at the NE-membrane. Consistent with the idea that the cytoplasmic phase of NE-membranes can support this novel pool of perinuclear actin polymerisation we show that isolated, intact, differentiated primary hepatocyte nuclei support actin polymerisation in vitro. Further this phenomenon was inhibited by treatments hindering steric access to outer-nuclear-membrane proteins (e.g. wheat germ agglutinin, anti-nesprin and anti-nucleoporin antibodies). CONCLUSION: We conclude that actin polymerisation occurs around interphase nuclei of living cells at the cytoplasmic phase of NE-membranes.


Subject(s)
Actins/chemistry , Nuclear Envelope/chemistry , Animals , Binding Sites , Biopolymers , Boron Compounds/analysis , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Circular Dichroism , Cytochalasin D/pharmacology , Depsipeptides/pharmacology , Fluorescent Dyes/analysis , HeLa Cells , Humans , Liver/ultrastructure , Rabbits , Rats , Thiazoles/pharmacology , Thiazolidines
15.
Br J Pharmacol ; 173(11): 1820-34, 2016 06.
Article in English | MEDLINE | ID: mdl-26993859

ABSTRACT

BACKGROUND AND PURPOSE: Glucagon-like peptide-1 (GLP-1) analogues improve glycaemic control in type 2 diabetic (T2D) patients and cause weight loss in obese subjects by as yet unknown mechanisms. We recently demonstrated that the GLP-1 receptor, which is present in adipocytes and the stromal vascular fraction of human adipose tissue (AT), is up-regulated in AT of insulin-resistant morbidly obese subjects compared with healthy lean subjects. The aim of this study was to explore the effects of in vitro and in vivo administration of GLP-1 and its analogues on AT and adipocyte functions from T2D morbidly obese subjects. EXPERIMENTAL APPROACH: We analysed the effects of GLP-1 on human AT and isolated adipocytes in vitro and the effects of GLP-1 mimetics on AT of morbidly obese T2D subjects in vivo. KEY RESULTS: GLP-1 down-regulated the expression of lipogenic genes when administered during in vitro differentiation of human adipocytes from morbidly obese patients. GLP-1 also decreased the expression of adipogenic/lipogenic genes in AT explants and mature adipocytes, while increasing that of lipolytic markers and adiponectin. In 3T3-L1 adipocytes, GLP-1 decreased free cytosolic Ca2+ concentration ([Ca2+]i). GLP-1-induced responses were only partially blocked by GLP-1 receptor antagonist exendin (9­39). Moreover, administration of exenatide or liraglutide reduced adipogenic and inflammatory marker mRNA in AT of T2D obese subjects. CONCLUSIONS AND IMPLICATIONS: Our data suggest that the beneficial effects of GLP-1 are associated with changes in the adipogenic potential and ability of AT to expand, via activation of the canonical GLP-1 receptor and an additional, as yet unknown, receptor.


Subject(s)
Adipocytes/drug effects , Cell Differentiation/drug effects , Glucagon-Like Peptide 1/pharmacology , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/metabolism , Animals , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Exenatide , Gene Expression/drug effects , Genetic Markers , Humans , Mice , Obesity, Morbid/complications , Obesity, Morbid/metabolism , Obesity, Morbid/pathology , Peptides/therapeutic use , Pilot Projects , Prospective Studies , Venoms/therapeutic use
16.
Obes Surg ; 26(8): 1757-67, 2016 08.
Article in English | MEDLINE | ID: mdl-26678755

ABSTRACT

BACKGROUND: Adipose tissue (AT) dysfunction in obesity is commonly linked to insulin resistance and promotes the development of metabolic disease. Bariatric surgery (BS) represents an effective strategy to reduce weight and to improve metabolic health in morbidly obese subjects. However, the mechanisms and pathways that are modified in AT in response to BS are not fully understood, and few information is still available as to whether these may vary depending on the metabolic status of obese subjects. METHODS: Abdominal subcutaneous adipose tissue (SAT) samples were obtained from morbidly obese women (n = 18) before and 13.3 ± 0.37 months after BS. Obese women were stratified into two groups: normoglycemic (NG; Glu < 100 mg/dl, HbA1c <5.7 %) or insulin resistant (IR; Glu 100-126 mg/dl, HbA1c 5.7-6.4 %) (n = 9/group). A multi-comparative proteomic analysis was employed to identify differentially regulated SAT proteins by BS and/or the degree of insulin sensitivity. Serum levels of metabolic, inflammatory, and anti-oxidant markers were also analyzed. RESULTS: Before surgery, NG and IR subjects exhibited differences in AT proteins related to inflammation, metabolic processes, the cytoskeleton, and mitochondria. BS caused comparable weight reductions and improved glucose homeostasis in both groups. However, BS caused dissimilar changes in metabolic enzymes, inflammatory markers, cytoskeletal components, mitochondrial proteins, and angiogenesis regulators in NG and IR women. CONCLUSIONS: BS evokes significant molecular rearrangements indicative of improved AT function in morbidly obese women at either low or high metabolic risk, though selective adaptive changes in key cellular processes occur depending on the initial individual's metabolic status.


Subject(s)
Biomarkers/metabolism , Insulin Resistance , Metabolic Syndrome/metabolism , Obesity, Morbid/surgery , Subcutaneous Fat, Abdominal/metabolism , Weight Loss , Adult , Bariatric Surgery , Female , Humans , Obesity, Morbid/metabolism , Women's Health
17.
Mol Endocrinol ; 16(9): 2093-100, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12198245

ABSTRACT

Recent reports demonstrate that the rat GnRH promoter is activated in an episodic fashion in immortalized GnRH neurons, but little information is available on molecular processes that contribute to this phenomenon. In this study, we dissected the regions of the rat GnRH promoter that mediate these effects by testing a series of 5' deletion luciferase reporter constructs on the pattern of photonic emissions from single, living GT1-7 GnRH neuronal cells. Deletion analysis revealed that the region -2012/-1597 that contains the neuron-specific enhancer (NSE) was required for the elaboration of pulses of GnRH promoter activity. The importance of this region was supported by observations that episodic reporter activity could be transferred to a neutral nonpulsatile promoter (Rous sarcoma virus, RSV(180)). Immunoneutralization of Oct-1 as well as mutation of an octamer binding site located at -1787/-1783 (AT-a site) blocked the pulsatile GnRH promoter activity in GT1-7 neuronal cells. Taken together, our findings indicate that episodic GnRH gene expression is a promoter-dependent phenomenon, which is mediated by Oct-1 interaction with regulatory elements in the NSE region.


Subject(s)
DNA-Binding Proteins/metabolism , Gonadotropin-Releasing Hormone/genetics , Promoter Regions, Genetic/genetics , Transcription Factors/metabolism , Transcriptional Activation , Animals , Cells, Cultured , Host Cell Factor C1 , Mice , Octamer Transcription Factor-1 , Rats , Response Elements/genetics , Sequence Deletion/genetics
18.
Antioxid Redox Signal ; 23(7): 597-612, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-25714483

ABSTRACT

AIMS: Obesity is characterized by a low-grade systemic inflammatory state and adipose tissue (AT) dysfunction, which predispose individuals to the development of insulin resistance (IR) and metabolic disease. However, a subset of obese individuals, referred to as metabolically healthy obese (MHO) individuals, are protected from obesity-associated metabolic abnormalities. Here, we aim at identifying molecular factors and pathways in adipocytes that are responsible for the progression from the insulin-sensitive to the insulin-resistant, metabolically unhealthy obese (MUHO) phenotype. RESULTS: Proteomic analysis of paired samples of adipocytes from subcutaneous (SC) and omental (OM) human AT revealed that both types of cells are altered in the MUHO state. Specifically, the glutathione redox cycle and other antioxidant defense systems as well as the protein-folding machinery were dysregulated and endoplasmic reticulum stress was increased in adipocytes from IR subjects. Moreover, proteasome activity was also compromised in adipocytes of MUHO individuals, which was associated with enhanced accumulation of oxidized and ubiquitinated proteins in these cells. Proteasome activity was also impaired in adipocytes of diet-induced obese mice and in 3T3-L1 adipocytes exposed to palmitate. In line with these data, proteasome inhibition significantly impaired insulin signaling in 3T3-L1 adipocytes. INNOVATION: This study provides the first evidence of the occurrence of protein homeostasis deregulation in adipocytes in human obesity, which, together with oxidative damage, interferes with insulin signaling in these cells. CONCLUSION: Our results suggest that proteasomal dysfunction and impaired proteostasis in adipocytes, resulting from protein oxidation and/or misfolding, constitute major pathogenic mechanisms in the development of IR in obesity.


Subject(s)
Adipocytes/pathology , Insulin Resistance , Obesity, Metabolically Benign/physiopathology , Oxidative Stress , Proteasome Endopeptidase Complex/metabolism , 3T3-L1 Cells , Adipocytes/metabolism , Adult , Animals , Disease Models, Animal , Endoplasmic Reticulum Stress , Female , Gene Expression Regulation , Humans , Male , Mice , Obesity, Metabolically Benign/metabolism , Obesity, Metabolically Benign/pathology , Omentum/cytology , Omentum/metabolism , Omentum/pathology , Palmitic Acid/pharmacology , Proteomics/methods , Subcutaneous Fat/metabolism , Subcutaneous Fat/pathology , Unfolded Protein Response
19.
Endocrinology ; 143(3): 1126-33, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11861540

ABSTRACT

PRL gene expression in the anterior pituitary has been the focus of intensive investigation for many years, but very little information is available on the actual dynamics by which this process occurs in individual mammotrope cells. Here, we used single cell bioluminescent imaging microscopy and a recently refined reporter gene strategy to measure PRL promoter-driven gene expression (PRL-GE) in individual living primary mammotropes. Using this approach we report a new phenomenon involving repetitive on/off gene expression bursts that occurred in a distinctly noncircadian oscillatory pattern. Furthermore, we demonstrate a functional basis for these gene expression oscillations, inasmuch as PRL-GE pulses were sensitive to calcium-dependent modulation, which we show arose exclusively as changes in the shape of individual pulse episodes. Our results provide the first clear evidence that PRL-GE, in its homologous cell environment, displays oscillatory bursts of activity. Moreover, they strongly support the idea that these discrete on/off bursts of activity serve as an important determinant of the timing and level of PRL-GE under both basal and stimulated conditions.


Subject(s)
Periodicity , Pituitary Gland/cytology , Pituitary Gland/metabolism , Prolactin/biosynthesis , Prolactin/genetics , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Animals , Calcium Channels, L-Type/metabolism , Cells, Cultured , Female , Genes, Reporter/genetics , Genetic Vectors , Ion Channel Gating/physiology , Luciferases/biosynthesis , Luciferases/genetics , Luminescent Measurements , Microinjections , Plasmids/genetics , Rats
20.
Mol Nutr Food Res ; 58(11): 2177-88, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25044988

ABSTRACT

SCOPE: To determine whether the insulin resistance that exists in metabolic syndrome (MetS) patients is modulated by dietary fat composition. METHODS AND RESULTS: Seventy-five patients were randomly assigned to one of four diets for 12 wk: high-saturated fatty acids (HSFAs), high-MUFA (HMUFA), and two low-fat, high-complex carbohydrate (LFHCC) diets supplemented with long-chain n-3 (LFHCC n-3) PUFA or placebo. At the end of intervention, the LFHCC n-3 diet reduced plasma insulin, homeostasis model assessment of insulin resistance, and nonsterified fatty acid concentration (p < 0.05) as compared to baseline Spanish habitual (BSH) diet. Subcutaneous white adipose tissue (WAT) analysis revealed decreased EH-domain containing-2 mRNA levels and increased cbl-associated protein gene expression with the LFHCC n-3 compared to HSFA and HMUFA diets, respectively (p < 0.05). Moreover, the LFHCC n-3 decreased gene expression of glyceraldehyde-3-phosphate dehydrogenase with respect to HMUFA and BSH diets (p < 0.05). Finally, proteomic characterization of subcutaneous WAT identified three proteins of glucose metabolism downregulated by the LFHCC n-3 diet, including annexin A2. RT-PCR analysis confirmed the decrease of annexin A2 (p = 0.027) after this diet. CONCLUSION: Our data suggest that the LFHCC n-3 diet reduces systemic insulin resistance and improves insulin signaling in subcutaneous WAT of MetS patients compared to HSFA and BSH diets consumption.


Subject(s)
Adipose Tissue, White/metabolism , Diet , Dietary Fats/administration & dosage , Insulin Resistance , Metabolic Syndrome/metabolism , Subcutaneous Fat/metabolism , Annexin A2/genetics , Annexin A2/metabolism , Blood Pressure , Body Mass Index , Dietary Carbohydrates/administration & dosage , Fatty Acids, Monounsaturated , Fatty Acids, Unsaturated/administration & dosage , Glyceraldehyde-3-Phosphate Dehydrogenases/genetics , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Humans , Insulin/blood , Life Style , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
SELECTION OF CITATIONS
SEARCH DETAIL