Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Obes Rev ; 22(1): e13114, 2021 01.
Article in English | MEDLINE | ID: mdl-33030294

ABSTRACT

Most known types of nonsyndromic monogenic obesity are caused by rare mutations in genes of the leptin-melanocortin pathway controlling appetite and adiposity. In contrast, congenital generalized lipodystrophy represents the most extreme form of leanness in humans caused by recessive mutations in four genes involved in phospholipid/triglyceride synthesis and lipid droplet/caveolae structure. In this disease, the inability to store triglyceride in adipocytes results in hypoleptinemia and ectopic hepatic and muscle fat accumulation leading to fatty liver, hypertriglyceridemia and severe insulin resistance. As a result of hypoleptinemia, patients with lipodystrophy show alterations in eating behaviour characterized by constant increased energy intake. As it occurs in obesity caused by genetic leptin deficiency, exogenous leptin rapidly reduces hunger scores in patients with congenital generalized lipodystrophy, with additional beneficial effects on glucose homeostasis and metabolic profile normalization. The melanocortin-4 receptor agonist setmelanotide has been used in the treatment of monogenic obesities. There is only one report on the effect of setmelanotide in a patient with partial lipodystrophy resulting in mild reductions in hunger scores, with no improvements in metabolic status. The assessment of contrasting phenotypes of obesity/leanness represents an adequate strategy to understand the pathophysiology and altered eating behaviour associated with adipose tissue excessive accumulation/paucity.


Subject(s)
Adiposity , Feeding Behavior , Lipodystrophy, Congenital Generalized , Obesity , Humans , Leptin , Lipodystrophy, Congenital Generalized/genetics , Obesity/genetics , Phenotype
2.
Obes Rev ; 21(4): e12983, 2020 04.
Article in English | MEDLINE | ID: mdl-31814283

ABSTRACT

Obesity and cholesterol gallstone disease (GSD) are frequently coexisting diseases; therefore and considering the current worldwide obesity epidemics, a precise understanding of the pathophysiological relationships between GSD and insulin resistance (IR) is important. Classically, obesity has been understood as a risk factor for GSD and the gallbladder (GB) viewed as a simple bile reservoir, with no metabolic roles whatsoever. However, consistent evidence has showed that both GSD and cholecystectomy associates with fatty liver and IR, raising the possibility that the GB is indeed an organ with metabolic regulatory roles. Herein, we review the pathophysiological mechanisms by which GSD, IR, and obesity are interconnected, with emphasis in the actions of the GB as a regulator of bile acids kinetics and a hormone secreting organ, with metabolic actions at the systemic level. We also examine the relationships between increased hepatic lipogenic in IR states and GSD pathogenesis. We propose a model in which GSD and hepatic IR mutually interact to determine a state of dysregulated lipid and energy metabolism that potentiate the metabolic dysregulation of obesity.


Subject(s)
Cholelithiasis/complications , Cholelithiasis/physiopathology , Insulin Resistance/physiology , Obesity/complications , Obesity/physiopathology , Adipose Tissue/physiopathology , Animals , Bile Acids and Salts/metabolism , Cholecystectomy/statistics & numerical data , Energy Metabolism/physiology , Fatty Liver/complications , Fatty Liver/physiopathology , Female , Gallbladder/physiopathology , Humans , Intestines/physiopathology , Lipid Metabolism/physiology , Liver/physiopathology , Risk Factors
3.
PLoS One ; 13(3): e0194644, 2018.
Article in English | MEDLINE | ID: mdl-29579094

ABSTRACT

BACKGROUND/OBJECTIVES: Abdominal obesity (AO) is associated with elevated risk for cardiovascular diseases; however, this association is less clear for non-obese people. We estimated the association of AO and cardiovascular risk factors (CVRF) and disease in non-obese adult individuals from Chile. SUBJECTS/METHODS: 5248 adults (15 years of age or older) of both sexes from the Chilean National Health Survey (October 2009 -September 2010, response rate 85%.) were included. Information on myocardial infarction and stroke was self-reported. BMI, waist circumference (WC), arterial pressure, plasma glucose, and cholesterol levels were measured. Predictive accuracy of WC was evaluated by area under curve of receiver operating characteristic analysis and cut off points were established by Youden Index. Relationship between AO and CVRF was analyzed by Chi-squared tests. RESULTS: Normal weight/overweight/obesity were present in 34.4%/45.2%/18.1% of men and 33.4%/33.6%/27.5% of women. Predictive accuracy of WC to identify at least one CVRF was 0.70/0.67 and optimal cutoff points for WC in non-obese subjects were 91/83 cm in men/women, respectively. AO was present in 98.2%/99.1% of obese, 70.5%/77.4% of overweight and 12.4%/16.4% of normal weight men/women. AO was associated with increased frequency of CVRF in overweight men (6/8 and stroke) and women (4/8) and higher frequency in normal weight men (8/8 and myocardial infarction/stroke) and women (6/8 and myocardial infarction). CONCLUSIONS: WC cutoff points calculated for non-obese chilean population discriminate more differences in CVRF in normal weight woman. AO significantly increases the frequency of CVRF and diseases in overweight and especially normal weight individuals. WC can be used as a low cost, feasible and reproducible predictor for CVRF in non-obese individuals in most clinical settings.


Subject(s)
Cardiovascular Diseases/etiology , Obesity, Abdominal/complications , Overweight/complications , Adult , Area Under Curve , Blood Glucose/analysis , Body Mass Index , Chile , Cholesterol/blood , Cross-Sectional Studies , Female , Health Surveys , Humans , Male , Obesity, Abdominal/diagnosis , Overweight/diagnosis , ROC Curve , Risk Factors , Waist Circumference
4.
Biol Rev Camb Philos Soc ; 93(2): 1145-1164, 2018 05.
Article in English | MEDLINE | ID: mdl-29230933

ABSTRACT

The discovery of metabolically active brown adipose tissue (BAT) in adult humans has fuelled the research of diverse aspects of this previously neglected tissue. BAT is solely present in mammals and its clearest physiological role is non-shivering thermogenesis, owing to the capacity of brown adipocytes to dissipate metabolic energy as heat. Recently, a number of other possible functions have been proposed, including direct regulation of glucose and lipid homeostasis and the secretion of a number of factors with diverse regulatory actions. Herein, we review recent advances in general biological knowledge of BAT and discuss the possible implications of this tissue in human metabolic health. In particular, we confront the claimed thermogenic potential of BAT for human energy balance and body mass regulation, mostly based on animal studies, with the most recent quantifications of human BAT.


Subject(s)
Adipose Tissue, Brown/physiology , Obesity/pathology , Animals , Humans , Obesity/metabolism , Obesity/prevention & control
5.
Elife ; 62017 02 28.
Article in English | MEDLINE | ID: mdl-28244871

ABSTRACT

The synthesis of cholesterol and fatty acids (FA) in the liver is independently regulated by SREBP-2 and SREBP-1c, respectively. Here, we genetically deleted Srebf-2 from hepatocytes and confirmed that SREBP-2 regulates all genes involved in cholesterol biosynthesis, the LDL receptor, and PCSK9; a secreted protein that degrades LDL receptors in the liver. Surprisingly, we found that elimination of Srebf-2 in hepatocytes of mice also markedly reduced SREBP-1c and the expression of all genes involved in FA and triglyceride synthesis that are normally regulated by SREBP-1c. The nuclear receptor LXR is necessary for Srebf-1c transcription. The deletion of Srebf-2 and subsequent lower sterol synthesis in hepatocytes eliminated the production of an endogenous sterol ligand required for LXR activity and SREBP-1c expression. These studies demonstrate that cholesterol and FA synthesis in hepatocytes are coupled and that flux through the cholesterol biosynthetic pathway is required for the maximal SREBP-1c expression and high rates of FA synthesis.


Subject(s)
Gene Expression Regulation , Liver X Receptors/metabolism , Liver/physiology , Sterol Regulatory Element Binding Protein 1/biosynthesis , Sterol Regulatory Element Binding Protein 2/metabolism , Animals , Cholesterol/metabolism , Fatty Acids/metabolism , Gene Knockout Techniques , Mice , Mice, Knockout , Sterol Regulatory Element Binding Protein 2/genetics , Transcription, Genetic
6.
PLoS One ; 12(1): e0170213, 2017.
Article in English | MEDLINE | ID: mdl-28129327

ABSTRACT

AIMS: Pancreatic ß-cells synthesize and release serotonin (5 hydroxytryptamine, 5HT); however, the role of 5HT receptors on glucose stimulated insulin secretion (GSIS) and the mechanisms mediating this function is not fully understood. The aims of this study were to determine the expression profile of 5HT receptors in murine MIN6 ß-cells and to examine the effects of pharmacological activation of 5HT receptor Htr2b on GSIS and mitochondrial function. MATERIALS AND METHODS: mRNA levels of 5HT receptors in MIN6 cells were quantified by RT qPCR. GSIS was assessed in MIN6 cells in response to global serotonergic activation with 5HT and pharmacological Htr2b activation or inhibition with BW723C86 or SB204741, respectively. In response to Htr2b activation also was evaluated the mRNA and protein levels of PGC1α and PPARy by RT-qPCR and western blotting and mitochondrial function by oxygen consumption rate (OCR) and ATP cellular content. RESULTS: We found that mRNA levels of most 5HT receptors were either very low or undetectable in MIN6 cells. By contrast, Htr2b mRNA was present at moderate levels in these cells. Preincubation (6 h) of MIN6 cells with 5HT or BW723C86 reduced GSIS and the effect of 5HT was prevented by SB204741. Preincubation with BW723C86 increased PGC1α and PPARy mRNA and protein levels and decreased mitochondrial respiration and ATP content in MIN6 cells. CONCLUSIONS: Our results indicate that prolonged Htr2b activation in murine ß-cells decreases glucose-stimulated insulin secretion and mitochondrial activity by mechanisms likely dependent on enhanced PGC1α/PPARy expression.


Subject(s)
Insulin/metabolism , PPAR gamma/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Receptors, Serotonin/genetics , Serotonin/metabolism , Adenosine Triphosphate/metabolism , Animals , Cell Line , Gene Expression Regulation/drug effects , Glucose/metabolism , Humans , Indoles/pharmacology , Insulin/genetics , Insulin-Secreting Cells/metabolism , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Mice , Mitochondria/genetics , Mitochondria/metabolism , Oxygen Consumption/genetics , PPAR gamma/biosynthesis , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/biosynthesis , Receptors, Serotonin/biosynthesis , Serotonin/genetics , Serotonin/pharmacology , Thiophenes/pharmacology , Urea/analogs & derivatives , Urea/pharmacology
7.
Mol Metab ; 5(7): 491-505, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27408775

ABSTRACT

OBJECTIVE: Characterize the cellular and molecular events responsible for lipodystrophy in AGPAT2 deficient mice. METHODS: Adipose tissue and differentiated MEF were assessed using light and electron microscopy, followed by protein (immunoblots) and mRNA analysis (qPCR). Phospholipid profiling was determined by electrospray ionization tandem mass spectrometry (ESI-MS/MS). RESULTS: In contrast to adult Agpat2 (-/-) mice, fetuses and newborn Agpat2 (-/-) mice have normal mass of white and brown adipose tissue. Loss of both the adipose tissue depots occurs during the first week of postnatal life as a consequence of adipocyte death and inflammatory infiltration of the adipose tissue. At the ultrastructural level, adipose tissue of newborn Agpat2 (-/-) mice is virtually devoid of caveolae and has abnormal mitochondria and lipid droplets. Autophagic structures are also abundant. Consistent with these findings, differentiated Agpat2 (-/-) mouse embryonic fibroblasts (MEFs) also have impaired adipogenesis, characterized by a lower number of lipid-laden cells and ultrastructural abnormalities in lipid droplets, mitochondria and plasma membrane. Overexpression of PPARγ, the master regulator of adipogenesis, increased the number of Agpat2 (-/-) MEFs that differentiated into adipocyte-like cells but did not prevent morphological abnormalities and cell death. Furthermore, differentiated Agpat2 (-/-) MEFs have abnormal phospholipid compositions with 3-fold increased levels of phosphatidic acid. CONCLUSION: We conclude that lipodystrophy in Agpat2 (-/-) mice results from postnatal cell death of adipose tissue in association with acute local inflammation. It is possible that AGPAT2 deficient adipocytes have an altered lipid filling or a reduced capacity to adapt the massive lipid availability associated with postnatal feeding.

8.
Biochem Biophys Res Commun ; 467(1): 39-45, 2015 Nov 06.
Article in English | MEDLINE | ID: mdl-26417690

ABSTRACT

AIMS: Mutations in 1-acylglycerol-3-phosphate O-acyltransferase 2 (AGPAT2) result in lipodystrophy, insulin resistance and diabetes. Autophagy is required for normal adipogenesis and adipose tissue development. The aim of this study was to determine whether impaired autophagy or excessive cell death underlie the adipogenic inability of Agpat2(-/-) mice preadipocytes. METHODS: Preadipocytes were isolated from interscapular brown adipose tissue (BAT) of Agpat2(-/-) and Agpat2(+/+) newborn mice and cultured/differentiated in vitro. Intracellular lipids were quantified by oil red O staining. Cell death was assessed by lactate dehydrogenase (LDH) activity. Apoptosis and autophagy regulatory factors were determined at the mRNA and protein level with Real-time PCR, immunoblot and immunofluorescence. RESULTS: Adipogenically induced Agpat2(-/-) preadipocytes had fewer lipid-loaded cells and lower levels of adipocyte markers than wild type preadipocytes. Before adipogenic differentiation, autophagy-related proteins (ATGs) ATG3, ATG5-ATG12 complex, ATG7 and LC3II were increased but autophagic flux was reduced, as suggested by increased p62 levels, in Agpat2(-/-) preadipocytes. Adipogenic induction increased LDH levels in the culture media in Agpat2(-/-) preadipocytes but no differences were observed in the activation of Caspase 3 or in markers of autophagic flux. CONCLUSIONS: AGPAT2 is required for in vitro adipogenesis of mouse preadipocytes. Autophagy defects or apoptosis are not involved in the adipogenic failure of Agpat2(-/-) preadipocytes.


Subject(s)
Acyltransferases/deficiency , Adipocytes, Brown/cytology , Adipocytes, Brown/enzymology , Adipogenesis/physiology , Acyltransferases/genetics , Adipogenesis/genetics , Animals , Apoptosis , Autophagy , Cell Differentiation , Cells, Cultured , Lipid Metabolism , Mice , Mice, Knockout , Stem Cells/cytology , Stem Cells/metabolism
9.
J. physiol. biochem ; 71(3): 471-478, sept. 2015. tab
Article in English | IBECS | ID: ibc-142443

ABSTRACT

Lipodystrophy encompass a group of heterogeneous disorders consisting in marked reduction, absence, and/or the redistribution of adipose tissue. Lipodystrophy is frequently complicated with severe insulin resistance, diabetes, hyperlipidemia, and fatty liver. Anatomically, lipodystrophies can be partial or generalized. Etiologically, they can be congenital or acquired. Lipodystrophy diagnosis can be challenging, and it has been suggested that partial forms can be easily misdiagnosed as common central obesity with associated metabolic syndrome. Conventional insulin-sensitizing approaches usually fail to fully ameliorate insulin resistance in lipodystrophic patients. Leptin replacement is an approved therapy for the metabolic complications of congenital generalized lipodystrophy. Novel nutritional interventions are promising complementary approaches for treating lipodystrophy metabolic complications


Subject(s)
Humans , Lipodystrophy/physiopathology , Metabolic Diseases/physiopathology , Adipose Tissue/metabolism , Insulin Resistance , Hypertriglyceridemia/physiopathology , Hypertriglyceridemic Waist/physiopathology , Leptin
10.
J Physiol Biochem ; 71(3): 471-8, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25833179

ABSTRACT

Lipodystrophy encompass a group of heterogeneous disorders consisting in marked reduction, absence, and/or the redistribution of adipose tissue. Lipodystrophy is frequently complicated with severe insulin resistance, diabetes, hyperlipidemia, and fatty liver. Anatomically, lipodystrophies can be partial or generalized. Etiologically, they can be congenital or acquired. Lipodystrophy diagnosis can be challenging, and it has been suggested that partial forms can be easily misdiagnosed as common central obesity with associated metabolic syndrome. Conventional insulin-sensitizing approaches usually fail to fully ameliorate insulin resistance in lipodystrophic patients. Leptin replacement is an approved therapy for the metabolic complications of congenital generalized lipodystrophy. Novel nutritional interventions are promising complementary approaches for treating lipodystrophy metabolic complications.


Subject(s)
Adipose Tissue/metabolism , Lipodystrophy/metabolism , Adipose Tissue/pathology , Animals , Humans , Insulin Resistance , Lipodystrophy/drug therapy , Lipodystrophy/etiology
11.
Rev Med Chil ; 142(6): 738-47, 2014 Jun.
Article in Spanish | MEDLINE | ID: mdl-25327319

ABSTRACT

The adipose tissue is an endocrine organ that produces a variety of protein hormones. One of them is leptin, which regulates several critical functions at the central nervous system such as caloric intake, basal energy expenditure, reproduction, glucose and lipid metabolism and osteogenesis. Acting at a local level, leptin modulates the immune system and promotes liver fibrogenesis. The most promising therapeutic implications of leptin will possibly be in type 1 diabetes mellitus (DM1). Its supplementation in animal models of DM1 prevents hyperglycemia and ketoacidosis. These actions depend on the activation of leptin receptors in the central nervous system and the suppression of glucagon signaling in the liver.


Subject(s)
Leptin/physiology , Adipose Tissue/physiology , Animals , Diabetes Mellitus, Type 1/therapy , Diabetes Mellitus, Type 2/therapy , Energy Metabolism/physiology , Humans , Leptin/therapeutic use , Mice , Rats , Receptors, Leptin/physiology
12.
Rev. méd. Chile ; 142(6): 738-747, jun. 2014. ilus
Article in Spanish | LILACS | ID: lil-722924

ABSTRACT

The adipose tissue is an endocrine organ that produces a variety of protein hormones. One of them is leptin, which regulates several critical functions at the central nervous system such as caloric intake, basal energy expenditure, reproduction, glucose and lipid metabolism and osteogenesis. Acting at a local level, leptin modulates the immune system and promotes liver fibrogenesis. The most promising therapeutic implications of leptin will possibly be in type 1 diabetes mellitus (DM1). Its supplementation in animal models of DM1 prevents hyperglycemia and ketoacidosis. These actions depend on the activation of leptin receptors in the central nervous system and the suppression of glucagon signaling in the liver.


Subject(s)
Animals , Humans , Mice , Rats , Leptin/physiology , Adipose Tissue/physiology , Diabetes Mellitus, Type 1/therapy , /therapy , Energy Metabolism/physiology , Leptin/therapeutic use , Receptors, Leptin/physiology
13.
PLoS One ; 9(1): e87173, 2014.
Article in English | MEDLINE | ID: mdl-24498038

ABSTRACT

Congenital generalized lipodystrophy (CGL) is a rare autosomal recessive disorder characterized by extreme reduction of white adipose tissue (WAT) mass. CGL type 1 is the most frequent form and is caused by mutations in AGPAT2. Genetic and clinical studies were performed in two affected sisters of a Chilean family. These patients have notoriously dissimilar metabolic abnormalities that correlate with differential levels of circulating leptin and soluble leptin receptor fraction. Sequencing of AGPAT2 exons and exon-intron boundaries revealed two homozygous mutations in both sisters. Missense mutation c.299G>A changes a conserved serine in the acyltransferase NHX4D motif of AGPAT2 (p.Ser100Asn). Intronic c.493-1G>C mutation destroy a conserved splicing site that likely leads to exon 4 skipping and deletion of whole AGPAT2 substrate binding domain. In silico protein modeling provided insights of the mechanisms of lack of catalytic activity owing to both mutations.


Subject(s)
Acyltransferases/genetics , Genetic Predisposition to Disease/genetics , Lipodystrophy, Congenital Generalized/genetics , Mutation , Siblings , Acyltransferases/chemistry , Acyltransferases/metabolism , Adult , Base Sequence , DNA Mutational Analysis , Female , Homozygote , Humans , Leptin/blood , Leptin/metabolism , Lipodystrophy, Congenital Generalized/blood , Lipodystrophy, Congenital Generalized/metabolism , Models, Molecular , Phenotype , Protein Structure, Tertiary , Receptors, Leptin/metabolism
14.
Front Biosci (Landmark Ed) ; 19(3): 416-28, 2014 01 01.
Article in English | MEDLINE | ID: mdl-24389193

ABSTRACT

Cholesterol has evolved to fulfill sophisticated biophysical, cell signaling and endocrine requirements of animal systems. At a cellular level, cholesterol is found in membranes, where it increases both bilayer stiffness and impermeability to water and ions. Furthermore, cholesterol is integrated into specialized lipid-protein membrane microdomains with critical topographical and signaling functions. At an organismal level, cholesterol is the precursor for all steroid hormones, including gluco- and mineralo-corticoids, sex hormones and vitamin D, all of which regulate carbohydrate, sodium, reproductive and bone homeostasis, respectively. This sterol is also the precursor for bile acids, which are important for intestinal absorption of dietary lipids as well as energy and glucose metabolic regulation. Importantly, complex mechanisms maintain cholesterol within physiological ranges and the disregulation of these mechanisms results in embryonic or adult diseases, caused by either excessive or reduced tissue cholesterol levels. The causative role of cholesterol in these diseases has been demonstrated by diverse genetic and pharmacologic animal models that are commented in this review.


Subject(s)
Cholesterol/physiology , Atherosclerosis/physiopathology , Biological Transport , Cholesterol/biosynthesis , Cholesterol/metabolism , Fetal Development/physiology , Humans
15.
J Lipid Res ; 55(2): 276-88, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24293639

ABSTRACT

Leptin is essential for energy homeostasis and regulation of food intake. Patients with congenital generalized lipodystrophy (CGL) due to mutations in 1-acylglycerol-3-phosphate-O-acyltransferase 2 (AGPAT2) and the CGL murine model (Agpat2(-/-) mice) both have severe insulin resistance, diabetes mellitus, hepatic steatosis, and low plasma leptin levels. In this study, we show that continuous leptin treatment of Agpat2(-/-) mice for 28 days reduced plasma insulin and glucose levels and normalized hepatic steatosis and hypertriglyceridemia. Leptin also partially, but significantly, reversed the low plasma thyroxine and high corticosterone levels found in Agpat2(-/-) mice. Levels of carbohydrate response element binding protein (ChREBP) were reduced, whereas lipogenic gene expression were increased in the livers of Agpat2(-/-) mice, suggesting that deregulated ChREBP contributed to the development of fatty livers in these mice and that this transcription factor is a target of leptin's beneficial metabolic action. Leptin administration did not change hepatic fatty acid oxidation enzymes mRNA levels in Agpat2(-/-) mice. The selective deletion of leptin receptors only in hepatocytes did not prevent the positive metabolic actions of leptin in Agpat2(-/-) mice, supporting the notion that the majority of metabolic actions of leptin are dependent on its action in nonhepatocyte cells and/or the central nervous system.


Subject(s)
1-Acylglycerol-3-Phosphate O-Acyltransferase/deficiency , Fatty Liver/complications , Fatty Liver/metabolism , Hepatocytes/metabolism , Insulin Resistance , Leptin/pharmacology , Lipodystrophy/complications , 1-Acylglycerol-3-Phosphate O-Acyltransferase/genetics , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Corticosterone/blood , Fatty Acids/biosynthesis , Fatty Acids/metabolism , Fatty Liver/blood , Gene Deletion , Gene Expression Regulation/drug effects , Glucose/metabolism , Glycogen/metabolism , Hepatocytes/drug effects , Lipogenesis/drug effects , Liver/metabolism , Liver/pathology , Male , Mice , Nuclear Proteins/metabolism , Oxidation-Reduction/drug effects , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Leptin/deficiency , Receptors, Leptin/genetics , Receptors, Leptin/metabolism , Thyroxine/blood , Transcription Factors/metabolism , Transcription, Genetic/drug effects , Triglycerides/metabolism
16.
Biol Rev Camb Philos Soc ; 88(4): 825-43, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23445165

ABSTRACT

Cholesterol has evolved to fulfill sophisticated biophysical, cell signalling, and endocrine functions in animal systems. At the cellular level, cholesterol is found in membranes where it increases both bilayer stiffness and impermeability to water and ions. Furthermore, cholesterol is integrated into specialized lipid-protein membrane microdomains with critical topographical and signalling functions. At the organismal level, cholesterol is the precursor of all steroid hormones, including gluco- and mineralo-corticoids, sex hormones, and vitamin D, which regulate carbohydrate, sodium, reproductive, and bone homeostasis, respectively. This sterol is also the immediate precursor of bile acids, which are important for intestinal absorption of dietary lipids as well as energy homeostasis and glucose regulation. Complex mechanisms maintain cholesterol within physiological ranges and the dysregulation of these mechanisms results in embryonic or adult diseases, caused by either excessive or reduced tissue cholesterol levels. The causative role of cholesterol in these conditions has been demonstrated by genetic and pharmacological manipulations in animal models of human disease that are discussed herein. Importantly, the understanding of basic aspects of cholesterol biology has led to the development of high-impact pharmaceutical therapies during the past century. The continuing effort to offer successful treatments for prevalent cholesterol-related diseases, such as atherosclerosis and neurodegenerative disorders, warrants further interdisciplinary research in the coming decades.

17.
J Biol Chem ; 286(43): 37676-91, 2011 Oct 28.
Article in English | MEDLINE | ID: mdl-21873652

ABSTRACT

Loss-of-function mutations in 1-acylglycerol-3-phosphate O-acyltransferase (AGPAT) 2 in humans and mice result in loss of both the white and brown adipose tissues from birth. AGPAT2 generates precursors for the synthesis of glycerophospholipids and triacylglycerols. Loss of adipose tissue, or lipodystrophy, results in hyperinsulinemia, diabetes mellitus, and severe hepatic steatosis. Here, we analyzed biochemical properties of human AGPAT2 and its close homolog, AGPAT1, and we studied their role in liver by transducing their expression via recombinant adenoviruses in Agpat2(-/-) mice. The in vitro substrate specificities of AGPAT1 and AGPAT2 are quite similar for lysophosphatidic acid and acyl-CoA. Protein homology modeling of both the AGPATs with glycerol-3-phosphate acyltransferase 1 (GPAT1) revealed that they have similar tertiary protein structure, which is consistent with their similar substrate specificities. When co-expressed, both isoforms co-localize to the endoplasmic reticulum. Despite such similarities, restoring AGPAT activity in liver by overexpression of either AGPAT1 or AGPAT2 in Agpat2(-/-) mice failed to ameliorate the hepatic steatosis. From these studies, we suggest that the role of AGPAT1 or AGPAT2 in liver lipogenesis is minimal and that accumulation of liver fat is primarily a consequence of insulin resistance and loss of adipose tissue in Agpat2(-/-) mice.


Subject(s)
1-Acylglycerol-3-Phosphate O-Acyltransferase/metabolism , Adipose Tissue/enzymology , Endoplasmic Reticulum/enzymology , Fatty Liver/enzymology , Lipodystrophy/enzymology , Liver/enzymology , 1-Acylglycerol-3-Phosphate O-Acyltransferase/genetics , Adipose Tissue/pathology , Animals , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/pathology , Fatty Liver/genetics , Fatty Liver/pathology , Glycerophospholipids/biosynthesis , Glycerophospholipids/genetics , HEK293 Cells , Humans , Insulin Resistance/genetics , Isoenzymes/genetics , Isoenzymes/metabolism , Lipodystrophy/genetics , Lipodystrophy/pathology , Liver/pathology , Mice , Mice, Knockout , Transduction, Genetic , Triglycerides/biosynthesis , Triglycerides/genetics
18.
Biochem J ; 432(2): 249-54, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-20854262

ABSTRACT

GSD-1 (glycogen storage disease type 1) is caused by an inherited defect in glucose-6-phosphatase activity, resulting in a massive accumulation of hepatic glycogen content and an induction of de novo lipogenesis. The chlorogenic acid derivative S4048 is a pharmacological inhibitor of the glucose 6-phosphate transporter, which is part of glucose-6-phosphatase, and allows for mechanistic studies concerning metabolic defects in GSD-1. Treatment of mice with S4048 resulted in an ~60% reduction in blood glucose, increased hepatic glycogen and triacylglycerol (triglyceride) content, and a markedly enhanced hepatic lipogenic gene expression. In mammals, hepatic expression of lipogenic genes is regulated by the co-ordinated action of the transcription factors SREBP (sterol-regulatory-element-binding protein)-1c, LXRα (liver X receptor α) and ChREBP (carbohydrate-response-element-binding protein). Treatment of Lxra-/- mice and Chrebp-/- mice with S4048 demonstrated that ChREBP, but not LXRα, mediates the induction of hepatic lipogenic gene expression in this murine model of GSD-1. Thus ChREBP is an attractive target to alleviate derangements in lipid metabolism observed in patients with GSD-1.


Subject(s)
Gene Expression Regulation , Glycogen Storage Disease/genetics , Nuclear Proteins/deficiency , Transcription Factors/deficiency , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Cholesterol/metabolism , Disease Models, Animal , Glucose-6-Phosphatase/adverse effects , Glucose-6-Phosphatase/genetics , Glucose-6-Phosphatase/metabolism , Glycogen Storage Disease/enzymology , Glycogen Storage Disease/metabolism , Humans , Imidazoles/administration & dosage , Imidazoles/pharmacology , Liver/enzymology , Liver/metabolism , Liver Glycogen/metabolism , Liver X Receptors , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Orphan Nuclear Receptors/deficiency , Orphan Nuclear Receptors/genetics , Orphan Nuclear Receptors/metabolism , Pyridines/administration & dosage , Pyridines/pharmacology , RNA/genetics , RNA/isolation & purification , Transcription Factors/genetics , Transcription Factors/metabolism , Triglycerides/metabolism
19.
Cell Metab ; 9(2): 165-76, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19187773

ABSTRACT

Mutations in 1-acylglycerol-3-phosphate-O-acyltransferase 2 (AGPAT2) cause congenital generalized lipodystrophy. To understand the molecular mechanisms underlying the metabolic complications associated with AGPAT2 deficiency, Agpat2 null mice were generated. Agpat2(-/-) mice develop severe lipodystrophy affecting both white and brown adipose tissue, extreme insulin resistance, diabetes, and hepatic steatosis. The expression of lipogenic genes and rates of de novo fatty acid biosynthesis were increased approximately 4-fold in Agpat2(-/-) mouse livers. The mRNA and protein levels of monoacylglycerol acyltransferase isoform 1 were markedly increased in the livers of Agpat2(-/-) mice, suggesting that the alternative monoacylglycerol pathway for triglyceride biosynthesis is activated in the absence of AGPAT2. Feeding a fat-free diet reduced liver triglycerides by approximately 50% in Agpat2(-/-) mice. These observations suggest that both dietary fat and hepatic triglyceride biosynthesis via a monoacylglycerol pathway may contribute to hepatic steatosis in Agpat2(-/-) mice.


Subject(s)
1-Acylglycerol-3-Phosphate O-Acyltransferase/metabolism , Fatty Liver/metabolism , Insulin Resistance/genetics , Lipodystrophy, Congenital Generalized/metabolism , 1-Acylglycerol-3-Phosphate O-Acyltransferase/genetics , Adipose Tissue/metabolism , Animals , Energy Metabolism , Lipodystrophy, Congenital Generalized/genetics , Mice , Mice, Knockout , Models, Animal , Triglycerides/biosynthesis
20.
J Opt Soc Am A Opt Image Sci Vis ; 19(10): 2043-52, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12365624

ABSTRACT

We present experimental measurements of the angular distribution of light scattered from large-scale composite randomly rough surfaces (oceanlike surfaces) with different statistical parameters illuminated at small and large angles of incidence. The surfaces are composed of a small-scale roughness superimposed on a slowly (large-scale) varying surface. The large-scale surfaces are diamond-machined periodic surfaces made on aluminum substrates and have either a sinusoidal or a Stokes wave profile. The small-scale roughness is added with microlithographic techniques, and the surfaces are then gold coated. For a linearly polarized incident beam, it is found that the diffusely scattered light is strongly depolarized and that its pattern is rather different for each large-scale surface profile. Enhanced backscattering is also observed.

SELECTION OF CITATIONS
SEARCH DETAIL
...