Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Obesity (Silver Spring) ; 26(11): 1721-1726, 2018 11.
Article in English | MEDLINE | ID: mdl-30358156

ABSTRACT

OBJECTIVE: The satiating effect of protein compared with other nutrients has been well described and is thought to be mediated, in part, by gut hormone release. Previously, it has been shown that oral L-arginine acts as a GLP-1 secretagogue both in vitro and in vivo in rodents. Here, the effect of L-arginine on gut hormone release in humans was investigated. METHODS: The hypothesis was tested in two separate studies. The first study assessed the tolerability of oral L-arginine in healthy human subjects. The second study assessed the effect of oral L-arginine on gut hormone release following an ad libitum meal. Subjects were given L-arginine, glycine (control amino acid), or vehicle control in a randomized double-blind fashion. RESULTS: At a dose of 17.1 mmol, L-arginine was well tolerated and stimulated the release of plasma GLP-1 (P < 0.05) and PYY (P < 0.001) following an ad libitum meal. Food diaries showed a trend toward lower energy intake and particularly fat intake following L-arginine treatment. CONCLUSIONS: L-arginine can significantly elevate GLP-1 and PYY in healthy human volunteers in combination with a meal. Further work is required to investigate whether L-arginine may have utility in the suppression of appetite and food intake.


Subject(s)
Appetite Depressants/therapeutic use , Arginine/therapeutic use , Eating/drug effects , Glucagon-Like Peptide 1/drug effects , Peptide YY/drug effects , Postprandial Period/drug effects , Adult , Appetite Depressants/pharmacology , Arginine/pharmacology , Double-Blind Method , Female , Glucagon-Like Peptide 1/blood , Humans , Male , Peptide YY/blood
2.
Biochem J ; 464(2): 179-92, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25220164

ABSTRACT

Extensive actin cytoskeleton remodelling occurs during adipocyte development. We have previously shown that disruption of stress fibres by the actin-severing protein cofilin is a requisite step in adipogenesis. However, it remains unclear whether actin nucleation and assembly into the cortical structure are essential for adipocyte development. In the present study we investigated the role of cortical actin assembly and of actin nucleation by the actin-related protein 2/3 (Arp2/3) complex in adipogenesis. Cortical actin structure formation started with accumulation of filamentous actin (F-actin) patches near the plasma membrane during adipogenesis. Depletion of Arp2/3 by knockdown of its subunits Arp3 or ARPC3 strongly impaired adipocyte differentiation, although adipogenesis-initiating factors were unaffected. Moreover, the assembly of F-actin-rich structures at the plasma membrane was suppressed and the cortical actin structure poorly developed after adipogenic induction in Arp2/3-deficient cells. Finally, we provide evidence that the cortical actin cytoskeleton is essential for efficient glucose transporter 4 (GLUT4) vesicle exocytosis and insulin signal transduction. These results show that the Arp2/3 complex is an essential regulator of adipocyte development through control of the formation of cortical actin structures, which may facilitate nutrient uptake and signalling events.


Subject(s)
Actin Cytoskeleton/metabolism , Actin-Related Protein 2-3 Complex/metabolism , Actin-Related Protein 2/metabolism , Actin-Related Protein 3/metabolism , Adipogenesis , Actin Cytoskeleton/genetics , Actin-Related Protein 2-3 Complex/chemistry , Adipocytes/metabolism , Animals , Cell Differentiation/genetics , Glucose Transporter Type 4/metabolism , Insulin/metabolism , Mice , Signal Transduction
3.
Hum Mol Genet ; 23(2): 502-13, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24026679

ABSTRACT

Seipin regulates lipid homeostasis by preventing lipid droplet (LD) formation in non-adipocytes but promoting it in developing adipocytes. Here, we report that seipin interacts with 14-3-3ß through its N- and C-termini. Expression of 14-3-3ß is upregulated during adipogenesis, and its deletion results in defective adipogenesis without affecting key adipogenic transcription factors. We further identified the actin-severing protein cofilin-1 as an interacting partner to 14-3-3ß. Cofilin-1 was spatiotemporally recruited by 14-3-3ß in the cytoplasm during adipocyte differentiation. Extensive actin cytoskeleton remodelling, from stress fibres to cortical structures, was apparent during adipogenesis, but not under lipogenic conditions, indicating that actin cytoskeleton remodelling is only required for adipocyte development. Similar to seipin and 14-3-3ß, cofilin-1 knockdown led to impaired adipocyte development. At the cellular level, differentiated cells with knockdown of cofilin-1, 14-3-3ß or seipin continued to maintain relatively intact stress fibres, in contrast to cortical actin structure in control cells. Finally, 3T3-L1 cells expressing a severing-resistant actin mutant exhibited impaired adipogenesis. We propose that seipin regulates adipogenesis by recruiting cofilin-1 to remodel actin cytoskeleton through the 14-3-3ß protein.


Subject(s)
14-3-3 Proteins/metabolism , Actin Cytoskeleton/metabolism , Adipocytes/physiology , Adipogenesis , Cofilin 1/metabolism , Heterotrimeric GTP-Binding Proteins/metabolism , 3T3-L1 Cells , Animals , GTP-Binding Protein gamma Subunits/genetics , GTP-Binding Protein gamma Subunits/metabolism , Gene Knockdown Techniques , HEK293 Cells , Heterotrimeric GTP-Binding Proteins/genetics , Humans , Mass Spectrometry , Mice , Mice, Inbred C57BL
4.
Biochem J ; 452(1): 37-44, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23458123

ABSTRACT

Homozygous mutations in BSCL2 (Berardinelli-Seip congenital lipodystrophy)/seipin cause CGL2 (congenital generalized lipodystrophy type 2). Recent data suggest that seipin regulates LD (lipid droplet) dynamics and adipocyte differentiation, but whether these roles are mechanistically linked remains unclear. To understand how seipin regulates these processes, we investigated the evolutionary changes of seipin orthologues, and studied individual domains in regulating lipid accumulation in non-adipocytes and adipocytes. Mammalian seipins comprise at least two distinct functional domains, a conserved core sequence and an evolutionarily acquired C-terminus. Despite its requirement for adipocyte formation, seipin overexpression inhibited oleate-induced LD formation and accumulation in nonadipocytes, which was mediated by the core sequence. In contrast, seipin overexpression did not inhibit LD accumulation during adipocyte differentiation or the adipogenic process in 3T3-L1 cells. However, adipogenesis and LD accumulation were impaired in 3T3-L1 cells expressing a seipin mutant lacking the C-terminus. Furthermore, expression of the same mutant without the C-terminus failed to rescue the adipogenic defects in seipin-knockdown cells, demonstrating the importance of the C-terminus for seipin's function in adipocyte development. We propose that seipin is involved in lipid homoeostasis by restricting lipogenesis and LD accumulation in non-adipocytes, while promoting adipogenesis to accommodate excess energy storage.


Subject(s)
Adipogenesis/physiology , Conserved Sequence/physiology , Evolution, Molecular , Heterotrimeric GTP-Binding Proteins/physiology , Lipid Metabolism/physiology , Lipogenesis/physiology , 3T3-L1 Cells , Adipocytes/chemistry , Adipocytes/metabolism , Adipocytes/physiology , Animals , GTP-Binding Protein gamma Subunits , HEK293 Cells , Humans , Lipids/antagonists & inhibitors , Lipids/biosynthesis , Mice , Protein Structure, Tertiary/physiology
5.
Biochem J ; 449(3): 605-12, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23126280

ABSTRACT

Cytoskeleton remodelling is a prerequisite step for the morphological transition from preadipocytes to mature adipocytes. Although microtubules play a pivotal role in organizing cellular structure, regulation of microtubule dynamics during adipogenesis remains unclear. In the present paper we show that acetylation of α-tubulin is up-regulated during adipogenesis, and adipocyte development is dependent on α-tubulin acetylation, as expression of an acetylation-resistant α-tubulin mutant significantly inhibits adipogenesis. Moreover, acetylation of α-tubulin is under the control of the acetyltransferase MEC-17 and deacetylases SIRT2 (Sirtuin 2) and HDAC6 (histone deacetylase 6). Adipocyte development is inhibited in MEC-17-knockdown cells, but enhanced in MEC-17-overexpressing cells. Finally, we show that katanin, a microtubule-severing protein with enhanced activity on acetylated α-tubulin, is actively involved in adipogenesis. We propose that co-ordinated up-regulation of α-tubulin acetylation initiates cytoskeleton remodelling by promoting α-tubulin severing by katanin which, in turn, allows expansion of lipid droplets and accelerates the morphological transition toward mature adipocytes.


Subject(s)
Acetyltransferases/metabolism , Adipogenesis/physiology , Tubulin/chemistry , Tubulin/metabolism , 3T3-L1 Cells , Acetylation , Acetyltransferases/antagonists & inhibitors , Acetyltransferases/genetics , Adenosine Triphosphatases/metabolism , Adipocytes/cytology , Adipocytes/metabolism , Adipogenesis/genetics , Animals , Cell Differentiation , Cytoskeleton/metabolism , Gene Knockdown Techniques , Histone Deacetylase 6 , Histone Deacetylases/metabolism , Katanin , Lipid Metabolism , Male , Mice , Mice, Inbred C57BL , Microtubules/metabolism , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Sirtuin 2/metabolism , Tubulin/genetics , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...