Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 209
Filter
1.
Nature ; 625(7996): 760-767, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38092039

ABSTRACT

GDF15, a hormone acting on the brainstem, has been implicated in the nausea and vomiting of pregnancy, including its most severe form, hyperemesis gravidarum (HG), but a full mechanistic understanding is lacking1-4. Here we report that fetal production of GDF15 and maternal sensitivity to it both contribute substantially to the risk of HG. We confirmed that higher GDF15 levels in maternal blood are associated with vomiting in pregnancy and HG. Using mass spectrometry to detect a naturally labelled GDF15 variant, we demonstrate that the vast majority of GDF15 in the maternal plasma is derived from the feto-placental unit. By studying carriers of rare and common genetic variants, we found that low levels of GDF15 in the non-pregnant state increase the risk of developing HG. Conversely, women with ß-thalassaemia, a condition in which GDF15 levels are chronically high5, report very low levels of nausea and vomiting of pregnancy. In mice, the acute food intake response to a bolus of GDF15 is influenced bi-directionally by prior levels of circulating GDF15 in a manner suggesting that this system is susceptible to desensitization. Our findings support a putative causal role for fetally derived GDF15 in the nausea and vomiting of human pregnancy, with maternal sensitivity, at least partly determined by prepregnancy exposure to the hormone, being a major influence on its severity. They also suggest mechanism-based approaches to the treatment and prevention of HG.


Subject(s)
Growth Differentiation Factor 15 , Hyperemesis Gravidarum , Nausea , Vomiting , Animals , Female , Humans , Mice , Pregnancy , beta-Thalassemia/blood , beta-Thalassemia/metabolism , Fetus/metabolism , Growth Differentiation Factor 15/blood , Growth Differentiation Factor 15/metabolism , Hormones/blood , Hormones/metabolism , Hyperemesis Gravidarum/complications , Hyperemesis Gravidarum/metabolism , Hyperemesis Gravidarum/prevention & control , Hyperemesis Gravidarum/therapy , Nausea/blood , Nausea/complications , Nausea/metabolism , Placenta/metabolism , Vomiting/blood , Vomiting/complications , Vomiting/metabolism
2.
bioRxiv ; 2023 Jun 04.
Article in English | MEDLINE | ID: mdl-37398065

ABSTRACT

Human pregnancy is frequently accompanied by nausea and vomiting that may become severe and life-threatening, as in hyperemesis gravidarum (HG), the cause of which is unknown. Growth Differentiation Factor-15 (GDF15), a hormone known to act on the hindbrain to cause emesis, is highly expressed in the placenta and its levels in maternal blood rise rapidly in pregnancy. Variants in the maternal GDF15 gene are associated with HG. Here we report that fetal production of GDF15, and maternal sensitivity to it, both contribute substantially to the risk of HG. We found that the great majority of GDF15 in maternal circulation is derived from the feto-placental unit and that higher GDF15 levels in maternal blood are associated with vomiting and are further elevated in patients with HG. Conversely, we found that lower levels of GDF15 in the non-pregnant state predispose women to HG. A rare C211G variant in GDF15 which strongly predisposes mothers to HG, particularly when the fetus is wild-type, was found to markedly impair cellular secretion of GDF15 and associate with low circulating levels of GDF15 in the non-pregnant state. Consistent with this, two common GDF15 haplotypes which predispose to HG were associated with lower circulating levels outside pregnancy. The administration of a long-acting form of GDF15 to wild-type mice markedly reduced subsequent responses to an acute dose, establishing that desensitisation is a feature of this system. GDF15 levels are known to be highly and chronically elevated in patients with beta thalassemia. In women with this disorder, reports of symptoms of nausea or vomiting in pregnancy were strikingly diminished. Our findings support a causal role for fetal derived GDF15 in the nausea and vomiting of human pregnancy, with maternal sensitivity, at least partly determined by pre-pregnancy exposure to GDF15, being a major influence on its severity. They also suggest mechanism-based approaches to the treatment and prevention of HG.

3.
Clin Exp Immunol ; 212(1): 52-60, 2023 04 07.
Article in English | MEDLINE | ID: mdl-36722378

ABSTRACT

Inflammation plays a fundamental role in the development of several metabolic diseases, including obesity and type 2 diabetes (T2D); the complement system has been implicated in their development. People of Black African (BA) ethnicity are disproportionately affected by T2D and other metabolic diseases but the impact of ethnicity on the complement system has not been explored. We investigated ethnic differences in complement biomarkers and activation status between men of BA and White European (WE) ethnicity and explored their association with parameters of metabolic health. We measured a panel of 15 complement components, regulators, and activation products in fasting plasma from 89 BA and 96 WE men. Ethnic differences were statistically validated. Association of complement biomarkers with metabolic health indices (BMI, waist circumference, insulin resistance, and HbA1c) were assessed in the groups. Plasma levels of the key complement components C3 and C4, the regulators clusterin and properdin and the activation marker iC3b were significantly higher in BA compared to WE men after age adjustment, while FD levels were significantly lower. C3 and C4 levels positively correlated with some or all markers of metabolic dysfunction in both ethnic groups while FD was inversely associated with HbA1c in both groups, and clusterin and properdin were inversely associated with some markers of metabolic dysfunction only in the WE group. Our findings of increased levels of complement components and activation products in BA compared to WE men suggest differences in complement regulation that may impact susceptibility to poor metabolic health.


Subject(s)
Clusterin , Insulin Resistance , Metabolic Diseases , Properdin , Humans , Male , Biomarkers , Diabetes Mellitus, Type 2 , Ethnicity , Glycated Hemoglobin , White People , Black People , Metabolic Diseases/ethnology , Complement C4 , Complement C3
4.
Nature ; 599(7885): 436-441, 2021 11.
Article in English | MEDLINE | ID: mdl-34732894

ABSTRACT

The state of somatic energy stores in metazoans is communicated to the brain, which regulates key aspects of behaviour, growth, nutrient partitioning and development1. The central melanocortin system acts through melanocortin 4 receptor (MC4R) to control appetite, food intake and energy expenditure2. Here we present evidence that MC3R regulates the timing of sexual maturation, the rate of linear growth and the accrual of lean mass, which are all energy-sensitive processes. We found that humans who carry loss-of-function mutations in MC3R, including a rare homozygote individual, have a later onset of puberty. Consistent with previous findings in mice, they also had reduced linear growth, lean mass and circulating levels of IGF1. Mice lacking Mc3r had delayed sexual maturation and an insensitivity of reproductive cycle length to nutritional perturbation. The expression of Mc3r is enriched in hypothalamic neurons that control reproduction and growth, and expression increases during postnatal development in a manner that is consistent with a role in the regulation of sexual maturation. These findings suggest a bifurcating model of nutrient sensing by the central melanocortin pathway with signalling through MC4R controlling the acquisition and retention of calories, whereas signalling through MC3R primarily regulates the disposition of calories into growth, lean mass and the timing of sexual maturation.


Subject(s)
Child Development/physiology , Nutritional Status/physiology , Puberty/physiology , Receptor, Melanocortin, Type 3/metabolism , Sexual Maturation/physiology , Adolescent , Aged, 80 and over , Animals , Child , Estrous Cycle/genetics , Estrous Cycle/physiology , Female , Homozygote , Humans , Hypothalamus/cytology , Hypothalamus/physiology , Insulin-Like Growth Factor I/metabolism , Male , Melanocortins/metabolism , Menarche/genetics , Menarche/physiology , Mice , Phenotype , Puberty/genetics , Receptor, Melanocortin, Type 3/deficiency , Receptor, Melanocortin, Type 3/genetics , Sexual Maturation/genetics , Time Factors , Weight Gain
6.
Mol Metab ; 43: 101127, 2021 01.
Article in English | MEDLINE | ID: mdl-33242659

ABSTRACT

OBJECTIVE: More than 300 genetic variants have been robustly associated with measures of human adiposity. Highly penetrant mutations causing human obesity do so largely by disrupting satiety pathways in the brain and increasing food intake. Most of the common obesity-predisposing variants are in, or near, genes expressed highly in the brain, but little is known of their function. Exploring the biology of these genes at scale in mammalian systems is challenging. We sought to establish and validate the use of a multicomponent screen for feeding behaviour phenotypes, taking advantage of the tractable model organism Drosophila melanogaster. METHODS: We validated a screen for feeding behaviour in Drosophila by comparing results after disrupting the expression of centrally expressed genes that influence energy balance in flies to those of 10 control genes. We then used this screen to explore the effects of disrupted expression of genes either a) implicated in energy homeostasis through human genome-wide association studies (GWAS) or b) expressed and nutritionally responsive in specific populations of hypothalamic neurons with a known role in feeding/fasting. RESULTS: Using data from the validation study to classify responses, we studied 53 Drosophila orthologues of genes implicated by human GWAS in body mass index and found that 15 significantly influenced feeding behaviour or energy homeostasis in the Drosophila screen. We then studied 50 Drosophila homologues of 47 murine genes reciprocally nutritionally regulated in POMC and agouti-related peptide neurons. Seven of these 50 genes were found by our screen to influence feeding behaviour in flies. CONCLUSION: We demonstrated the utility of Drosophila as a tractable model organism in a high-throughput genetic screen for food intake phenotypes. This simple, cost-efficient strategy is ideal for high-throughput interrogation of genes implicated in feeding behaviour and obesity in mammals and will facilitate the process of reaching a functional understanding of obesity pathogenesis.


Subject(s)
Appetite/genetics , Appetite/physiology , Feeding Behavior/physiology , Animals , Body Mass Index , Brain , Drosophila melanogaster/genetics , Energy Metabolism , Genome-Wide Association Study , Genotype , Homeostasis , Hypothalamus/metabolism , Neurons/metabolism , Nutritional Status , Obesity/metabolism , Phenotype
7.
QJM ; 114(3): 159-162, 2021 05 19.
Article in English | MEDLINE | ID: mdl-33245119

Subject(s)
Videoconferencing , Humans
8.
J Transl Med ; 17(1): 409, 2019 12 04.
Article in English | MEDLINE | ID: mdl-31801546

ABSTRACT

BACKGROUND: Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is a debilitating condition characterised by fatigue and post-exertional malaise. Its pathogenesis is poorly understood. GDF15 is a circulating protein secreted by cells in response to a variety of stressors. The receptor for GDF15 is expressed in the brain, where its activation results in a range of responses. Among the conditions in which circulating GDF15 levels are highly elevated are mitochondrial disorders, where early skeletal muscle fatigue is a key symptom. We hypothesised that GDF15 may represent a marker of cellular stress in ME/CFS. METHODS: GDF15 was measured in serum from patients with ME/CFS (n = 150; 100 with mild/moderate and 50 with severe symptoms), "healthy volunteers" (n = 150) and a cohort of patients with multiple sclerosis (n = 50). RESULTS: Circulating GDF15 remained stable in a subset of ME/CFS patients when sampled on two occasions ~ 7 months (IQR 6.7-8.8) apart, 720 pg/ml (95% CI 625-816) vs 670 pg/ml (95% CI 598-796), P = 0.5. GDF15 levels were 491 pg/ml in controls (95% CI 429-553), 546 pg/ml (95% CI 478-614) in MS patients, 560 pg/ml (95% CI 502-617) in mild/moderate ME/CFS patients and 602 pg/ml (95% CI 531-674) in severely affected ME/CFS patients. Accounting for potential confounders, severely affected ME/CFS patients had GDF15 concentrations that were significantly increased compared to healthy controls (P = 0.01). GDF15 levels were positively correlated (P = 0.026) with fatigue scores in ME/CFS. CONCLUSIONS: Severe ME/CFS is associated with increased levels of GDF15, a circulating biomarker of cellular stress that appears which stable over several months.


Subject(s)
Fatigue Syndrome, Chronic/blood , Growth Differentiation Factor 15/blood , Adult , Case-Control Studies , Female , Follow-Up Studies , Humans , Linear Models , Longitudinal Studies , Male , Middle Aged , Multivariate Analysis , Outcome Assessment, Health Care , Self Report , Time Factors
9.
Int J Obes (Lond) ; 42(8): 1524-1531, 2018 08.
Article in English | MEDLINE | ID: mdl-29980761

ABSTRACT

Muller et al. [1] have provided a strong critique of the Genome-Wide Association Studies (GWAS) of body-mass index (BMI), arguing that the GWAS approach for the study of BMI is flawed, and has provided us with few biological insights. They suggest that what is needed instead is a new start, involving GWAS for more complex energy balance related traits. In this invited counter-point, we highlight the substantial advances that have occurred in the obesity field, directly stimulated by the GWAS of BMI. We agree that GWAS for BMI is not perfect, but consider that the best route forward for additional discoveries will likely be to expand the search for common and rare variants linked to BMI and other easily obtained measures of obesity, rather than attempting to perform new, much smaller GWAS for energy balance traits that are complex and expensive to measure. For GWAS in general, we emphasise that the power from increasing the sample size of a crude but easily measured phenotype outweighs the benefits of better phenotyping.


Subject(s)
Body Mass Index , Body Weight/genetics , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study , Obesity/genetics , Humans
10.
Curr Opin Genet Dev ; 50: 60-67, 2018 06.
Article in English | MEDLINE | ID: mdl-29477938

ABSTRACT

Insulin resistance underpins the link between obesity and most of its associated metabolic disorders including type 2 diabetes, fatty liver disease, dyslipidaemia and cardiovascular disease. Despite its importance and extensive scientific endeavour, its precise molecular pathogenesis remains unclear. Monogenic syndromes of extreme insulin resistance, whilst rare in themselves, can provide unique insights into the pathogenesis of human insulin resistance. Severe insulin resistance syndromes are broadly classified into three categories: lipodystrophies, primary insulin signalling defects or complex syndromes including severe insulin resistance. Genetically confirmed classification has facilitated the identification of robust diagnostic biochemical features accelerating accurate clinical diagnosis. Interestingly the biochemical features of lipodystrophies are far more closely aligned to what is seen in prevalent forms of insulin resistance than those of primary insulin signalling defects, suggesting that lipodystrophy could be a relevant model for common disease. This assertion is supported by genome-wide association data indicating that SNPs associated with fasting hyperinsulinemia and metabolic dyslipidaemia, are strongly associated with a subtle reduction in hip fat, suggesting that subtle forms of lipodystrophy are likely to be a significant contributor to prevalent insulin resistance.


Subject(s)
Cardiovascular Diseases/genetics , Dyslipidemias/genetics , Fatty Liver/genetics , Insulin Resistance/genetics , Adipose Tissue/pathology , Cardiovascular Diseases/pathology , Dyslipidemias/pathology , Fatty Liver/pathology , Genome-Wide Association Study , Humans , Hyperinsulinism/genetics , Hyperinsulinism/pathology , Severity of Illness Index
11.
J Endocrinol ; 230(1): 13-26, 2016 07.
Article in English | MEDLINE | ID: mdl-27106110

ABSTRACT

Melanocortin receptor accessory protein 2 (MRAP2) is a transmembrane accessory protein predominantly expressed in the brain. Both global and brain-specific deletion of Mrap2 in mice results in severe obesity. Loss-of-function MRAP2 mutations have also been associated with obesity in humans. Although MRAP2 has been shown to interact with MC4R, a G protein-coupled receptor with an established role in energy homeostasis, appetite regulation and lipid metabolism, the mechanisms through which loss of MRAP2 causes obesity remains uncertain. In this study, we used two independently derived lines of Mrap2 deficient mice (Mrap2(tm1a/tm1a)) to further study the role of Mrap2 in the regulation of energy balance and peripheral lipid metabolism. Mrap2(tm1a/tm1a) mice have a significant increase in body weight, with increased fat and lean mass, but without detectable changes in food intake or energy expenditure. Transcriptomic analysis showed significantly decreased expression of Sim1, Trh, Oxt and Crh within the hypothalamic paraventricular nucleus of Mrap2(tm1a/tm1a) mice. Circulating levels of both high-density lipoprotein and low-density lipoprotein were significantly increased in Mrap2 deficient mice. Taken together, these data corroborate the role of MRAP2 in metabolic regulation and indicate that, at least in part, this may be due to defective central melanocortin signalling.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cholesterol/blood , Energy Metabolism/genetics , Receptor Activity-Modifying Proteins/metabolism , Repressor Proteins/metabolism , Adaptor Proteins, Signal Transducing , Animals , Anxiety/genetics , Anxiety/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Behavior, Animal/physiology , Body Weight/genetics , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/metabolism , Eating/genetics , Lipid Metabolism/genetics , Mice , Mice, Knockout , Motor Activity/genetics , Neurons/metabolism , Oxytocin/genetics , Oxytocin/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Receptor Activity-Modifying Proteins/genetics , Repressor Proteins/genetics , Thyrotropin-Releasing Hormone/genetics , Thyrotropin-Releasing Hormone/metabolism
12.
Diabetes ; 64(1): 299-310, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25114292

ABSTRACT

Perilipin 1 is a lipid droplet coat protein predominantly expressed in adipocytes, where it inhibits basal and facilitates stimulated lipolysis. Loss-of-function mutations in the PLIN1 gene were recently reported in patients with a novel subtype of familial partial lipodystrophy, designated as FPLD4. We now report the identification and characterization of a novel heterozygous frameshift mutation affecting the carboxy-terminus (439fs) of perilipin 1 in two unrelated families. The mutation cosegregated with a similar phenotype including partial lipodystrophy, severe insulin resistance and type 2 diabetes, extreme hypertriglyceridemia, and nonalcoholic fatty liver disease in both families. Poor metabolic control despite maximal medical therapy prompted two patients to undergo bariatric surgery, with remarkably beneficial consequences. Functional studies indicated that expression levels of the mutant protein were lower than wild-type protein, and in stably transfected preadipocytes the mutant protein was associated with smaller lipid droplets. Interestingly, unlike the previously reported 398 and 404 frameshift mutants, this variant binds and stabilizes ABHD5 expression but still fails to inhibit basal lipolysis as effectively as wild-type perilipin 1. Collectively, these findings highlight the physiological need for exquisite regulation of neutral lipid storage within adipocyte lipid droplets, as well as the possible metabolic benefits of bariatric surgery in this serious disease.


Subject(s)
Carrier Proteins/genetics , Diabetes Mellitus, Type 2/genetics , Frameshift Mutation , Hyperlipoproteinemia Type IV/genetics , Lipodystrophy, Familial Partial/genetics , Phosphoproteins/genetics , 1-Acylglycerol-3-Phosphate O-Acyltransferase/metabolism , 3T3-L1 Cells , Adipocytes, White/physiology , Adolescent , Adult , Amino Acid Sequence , Animals , Base Sequence , Carrier Proteins/metabolism , Family Health , Female , Humans , Insulin Resistance/genetics , Male , Mice , Middle Aged , Molecular Sequence Data , Mutagenesis, Site-Directed , Pedigree , Perilipin-1 , Phosphoproteins/metabolism
13.
JIMD Rep ; 9: 73-80, 2013.
Article in English | MEDLINE | ID: mdl-23430550

ABSTRACT

Loss-of-function mutations in AGPAT2, encoding 1-acylglycerol-3-phosphate-O-acyltransferase 2 (AGPAT2), produce congenital generalised lipodystrophy (CGL). We screened the AGPAT2 gene in two siblings who presented with pseudoacromegaly, diabetes and severe dyslipidaemia and identified a novel mutation in AGPAT2 causing a single amino acid substitution, p.Cys48Arg. We subsequently investigated the molecular pathogenic mechanism linking both this mutation and the previously reported p.Leu228Pro mutation to clinical disease. Wild-type and mutant AGPAT2 were expressed in control and AGPAT2-deficient preadipocyte cell lines. mRNA and protein expression was determined, and the ability of each AGPAT2 species to rescue adipocyte differentiation in AGPAT2-deficient cells was assessed. Protein levels of both p.Cys48Arg and p.Leu228Pro AGPAT2 were significantly reduced compared with that of wild-type AGPAT2 despite equivalent mRNA levels. Stable expression of wild-type AGPAT2 partially rescued adipogenesis in AGPAT2 deficient preadipocytes, whereas stable expression of p.Cys48Arg or p.Leu228Pro AGPAT2 did not. In conclusion, unusually severe dyslipidaemia and pseudoacromegaloid overgrowth in patients with diabetes should alert physicians to the possibility of lipodystrophy. Both the previously unreported pathogenic p.Cys48Arg mutation in AGPAT2, and the known p.Leu228Pro mutation result in decreased AGPAT2 protein expression in developing adipocytes. It is most likely that the CGL seen in homozygous carriers of these mutations is largely accounted for by loss of protein expression.

14.
Eur J Endocrinol ; 168(1): 101-6, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23047304

ABSTRACT

OBJECTIVE: It has been suggested that elevated levels of C-reactive protein (CRP) might interfere with leptin signalling and contribute to leptin resistance. Our aim was to assess whether plasma levels of CRP influence leptin resistance in humans, and our hypothesis was that CRP levels would modify the cross-sectional relationships between leptin and measures of adiposity. DESIGN AND METHODS: W assessed four measures of adiposity: BMI, waist circumference, fat mass and body fat (%) in 2113 British Regional Heart Study (BRHS) men (mean (s.d.) age 69 (5) years), with replication in 760 (age 69 (6) years) European Male Ageing Study (EMAS) subjects. RESULTS: IN BRHS subjects, leptin correlated with CRP (SPEARMAN'S R=0.22, P0.0001). Leptin and crp correlated with all four measures of adiposity (R VALUE RANGE: 0.22-0.57, all P<0.0001). Age-adjusted mean levels for adiposity measures increased in relation to leptin levels, but CRP level did not consistently influence the ß-coefficients of the regression lines in a CRP-stratified analysis. In BRHS subjects, the BMI vs leptin relationship demonstrated a weak statistical interaction with CRP (P=0.04). We observed no similar interaction in EMAS subjects and no significant interactions with other measures of adiposity in BRHS or EMAS cohorts. CONCLUSION: We have shown that plasma CRP has little influence on the relationship between measures of adiposity and serum leptin levels in these middle-aged and elderly male European cohorts. This study provides epidemiological evidence against CRP having a significant role in causing leptin resistance.


Subject(s)
C-Reactive Protein/metabolism , Leptin/blood , Adipose Tissue/anatomy & histology , Adiposity , Adult , Aged , Body Mass Index , Cohort Studies , Drug Resistance , Humans , Male , Middle Aged , Waist Circumference
15.
Mol Psychiatry ; 18(12): 1287-93, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23147384

ABSTRACT

The opioid system is implicated in the hedonic and motivational processing of food, and in binge eating, a behaviour strongly linked to obesity. The aim of this study was to evaluate the effects of 4 weeks of treatment with the mu-opioid receptor antagonist GSK1521498 on eating behaviour in binge-eating obese subjects. Adults with body mass index ≥ 30 kg m(-2) and binge eating scale scores ≥ 19 received 1-week single-blind placebo run-in, and were then randomized to 28 days with either 2 mg day(-1) GSK1521498, 5 mg day(-1) GSK1521498 or placebo (N=21 per arm) in a double-blind parallel group design. The outcome measures were body weight, fat mass, hedonic and consummatory eating behaviour during inpatient food challenges, safety and pharmacokinetics. The primary analysis was the comparison of change scores in the higher-dose treatment group versus placebo using analysis of covariance at each relevant time point. GSK1521498 (2 mg and 5 mg) was not different from placebo in its effects on weight, fat mass and binge eating scores. However, compared with placebo, GSK1521498 5 mg day(-1) caused a significant reduction in hedonic responses to sweetened dairy products and reduced calorific intake, particularly of high-fat foods during ad libitum buffet meals, with some of these effects correlating with systemic exposure of GSK1521498. There were no significant effects of GSK1521498 2 mg day(-1) on eating behaviour, indicating dose dependency of pharmacodynamics. GSK1521498 was generally well tolerated and no previously unidentified safety signals were detected. The potential for these findings to translate into clinically significant effects in the context of binge eating and weight regain prevention requires further investigation.


Subject(s)
Bulimia/drug therapy , Feeding Behavior/drug effects , Indans/pharmacology , Receptors, Opioid, mu/antagonists & inhibitors , Triazoles/pharmacology , Adolescent , Adult , Dose-Response Relationship, Drug , Double-Blind Method , Female , Humans , Indans/administration & dosage , Indans/therapeutic use , Male , Middle Aged , Triazoles/administration & dosage , Triazoles/therapeutic use , Young Adult
16.
Int J Obes (Lond) ; 37(5): 744-7, 2013 May.
Article in English | MEDLINE | ID: mdl-22614055

ABSTRACT

Genome-wide association studies have revealed that single nucleotide polymorphisms in fat mass and obesity-associated transcript (FTO) are robustly associated with body mass index and obesity. Expression of Fto in the hypothalamic arcuate nucleus is bidirectionally regulated as a function of nutritional status; decreasing following a 48-h fast and increasing after 10-week exposure to a high-fat diet. Here, we utilize an in vitro approach to determine which nutrients could regulate FTO levels at a cellular level. Using mouse and human cell lines, we find that FTO levels are not influenced by serum starvation. We demonstrate, however, that both glucose and total amino-acid deprivation regulates FTO expression. In particular, we have found that FTO mRNA and protein levels are dramatically downregulated by total amino-acid deprivation in mouse hypothalamic N46 cells, mouse embryonic fibroblasts and in human HEK293 cells. The drop rate of Fto mRNA is faster than its rate of natural degradation, pointing to regulation at the transcriptional level, which is reversible upon amino-acid replacement. Strikingly, this downregulation was seen only with essential amino-acid deficiency and not nonessential amino acids. These data suggest that FTO might have a role in the sensing of essential amino-acid availability.


Subject(s)
Amino Acids, Essential/metabolism , Arcuate Nucleus of Hypothalamus/metabolism , Glucose/metabolism , Mixed Function Oxygenases/metabolism , Obesity/metabolism , Oxo-Acid-Lyases/metabolism , Proteins/metabolism , Alpha-Ketoglutarate-Dependent Dioxygenase FTO , Animals , Blotting, Western , Cell Line , Diet, High-Fat , Down-Regulation , Gene Expression Regulation , Genome-Wide Association Study , HEK293 Cells , Humans , Mice , Obesity/genetics , Obesity/physiopathology , Polymorphism, Single Nucleotide , RNA, Messenger/metabolism , Time Factors
18.
Int J Obes (Lond) ; 36(9): 1248-51, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22124452

ABSTRACT

The unfolded protein response (UPR) is activated by endoplasmic reticulum stress resulting from an accumulation of unfolded or mis-folded proteins. The UPR is divided into three arms, involving the activation of ATF-6, PERK and IRE-1, that together act to restrict new protein synthesis and increase the production of chaperones. Recent studies have implicated the PERK and IRE-1 components of the UPR in adipocyte differentiation. In this study, we investigate the importance of ATF6α during adipogenesis using stable knockdown of this protein in the model adipogenic cell line, C3H10T1/2. Reduction of ATF6α expression by >70% resulted in impaired expression of key adipogenic genes and reduced lipid accumulation following the induction of adipogenesis. In contrast, loss of ATF6α did not impair the ability of cells to undergo osteogenic differentiation. Overall, our data indicate that all three arms of the UPR, including ATF6α, must be intact to permit adipogenesis to occur.


Subject(s)
Activating Transcription Factor 6/metabolism , Adipogenesis , Endoplasmic Reticulum/metabolism , Unfolded Protein Response , Activating Transcription Factor 6/genetics , Animals , Cell Differentiation , Cell Line , Endoplasmic Reticulum/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C3H , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Unfolded Protein Response/genetics , eIF-2 Kinase/metabolism
19.
Science ; 334(6055): 474, 2011 Oct 28.
Article in English | MEDLINE | ID: mdl-21979934

ABSTRACT

Pathological fasting hypoglycemia in humans is usually explained by excessive circulating insulin or insulin-like molecules or by inborn errors of metabolism impairing liver glucose production. We studied three unrelated children with unexplained, recurrent, and severe fasting hypoglycemia and asymmetrical growth. All were found to carry the same de novo mutation, p.Glu17Lys, in the serine/threonine kinase AKT2, in two cases as heterozygotes and in one case in mosaic form. In heterologous cells, the mutant AKT2 was constitutively recruited to the plasma membrane, leading to insulin-independent activation of downstream signaling. Thus, systemic metabolic disease can result from constitutive, cell-autonomous activation of signaling pathways normally controlled by insulin.


Subject(s)
Hypoglycemia/genetics , Hypoglycemia/metabolism , Mutation , Proto-Oncogene Proteins c-akt/genetics , Amino Acid Substitution , Cell Membrane/metabolism , Cell Nucleus/metabolism , Child , Female , Growth , HeLa Cells , Heterozygote , Humans , Insulin/blood , Insulin/metabolism , Male , Mosaicism , Pedigree , Protein Interaction Domains and Motifs , Proto-Oncogene Proteins c-akt/chemistry , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
20.
Diabet Med ; 28(12): 1476-86, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21992440

ABSTRACT

Sir Harold Himsworth first observed and articulated the phenomenon of insulin resistance in the late 1930s. Although a long delay followed before his observations were acknowledged and enshrined in formal diagnostic classifications of diabetes mellitus, insulin resistance-related pathology in the early 21st century poses one of the major global healthcare challenges for contemporary physicians. Whilst insulin resistance is closely related to obesity and decreased physical fitness, despite intensive investigation it has proved extremely challenging to discriminate key events in its causation from epiphenomena, many related to compensation for the primary defect. Thus, a complete account of the molecular pathogenesis of insulin resistance-related diseases remains elusive. One approach circumventing such problems is the study of patients with single gene defects causing severe insulin resistance. In such patients the primary defect is known, and thus lessons may be learned about human physiology from detailed physiological study allied to knowledge of the function of the mutated protein. This review discusses developments in understanding of monogenic severe insulin resistance since discovery of the first insulin receptor mutations in 1988 and reviews the physiological lessons learnt, including the critical role of adipose tissue in human metabolic health and the meaning and importance of 'partial' insulin resistance for major human disease.


Subject(s)
Adipose Tissue/metabolism , Diabetes Mellitus, Type 2/genetics , Insulin Resistance/genetics , Obesity/genetics , Receptor, Insulin/genetics , Adipose Tissue/pathology , Diabetes Mellitus, Type 2/etiology , Humans , Lipodystrophy/genetics , Lipodystrophy/metabolism , Obesity/complications , Obesity/pathology , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Receptor, Insulin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...