Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 209
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 625(7996): 760-767, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38092039

RESUMEN

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.


Asunto(s)
Factor 15 de Diferenciación de Crecimiento , Hiperemesis Gravídica , Náusea , Vómitos , Animales , Femenino , Humanos , Ratones , Embarazo , Talasemia beta/sangre , Talasemia beta/metabolismo , Feto/metabolismo , Factor 15 de Diferenciación de Crecimiento/sangre , Factor 15 de Diferenciación de Crecimiento/metabolismo , Hormonas/sangre , Hormonas/metabolismo , Hiperemesis Gravídica/complicaciones , Hiperemesis Gravídica/metabolismo , Hiperemesis Gravídica/prevención & control , Hiperemesis Gravídica/terapia , Náusea/sangre , Náusea/complicaciones , Náusea/metabolismo , Placenta/metabolismo , Vómitos/sangre , Vómitos/complicaciones , Vómitos/metabolismo
2.
Nature ; 599(7885): 436-441, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34732894

RESUMEN

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.


Asunto(s)
Desarrollo Infantil/fisiología , Estado Nutricional/fisiología , Pubertad/fisiología , Receptor de Melanocortina Tipo 3/metabolismo , Maduración Sexual/fisiología , Adolescente , Anciano de 80 o más Años , Animales , Niño , Ciclo Estral/genética , Ciclo Estral/fisiología , Femenino , Homocigoto , Humanos , Hipotálamo/citología , Hipotálamo/fisiología , Factor I del Crecimiento Similar a la Insulina/metabolismo , Masculino , Melanocortinas/metabolismo , Menarquia/genética , Menarquia/fisiología , Ratones , Fenotipo , Pubertad/genética , Receptor de Melanocortina Tipo 3/deficiencia , Receptor de Melanocortina Tipo 3/genética , Maduración Sexual/genética , Factores de Tiempo , Aumento de Peso
3.
Clin Exp Immunol ; 212(1): 52-60, 2023 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-36722378

RESUMEN

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.


Asunto(s)
Clusterina , Resistencia a la Insulina , Enfermedades Metabólicas , Properdina , Humanos , Masculino , Biomarcadores , Diabetes Mellitus Tipo 2 , Etnicidad , Hemoglobina Glucada , Población Blanca , Población Negra , Enfermedades Metabólicas/etnología , Complemento C4 , Complemento C3
4.
J Transl Med ; 17(1): 409, 2019 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-31801546

RESUMEN

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.


Asunto(s)
Síndrome de Fatiga Crónica/sangre , Factor 15 de Diferenciación de Crecimiento/sangre , Adulto , Estudios de Casos y Controles , Femenino , Estudios de Seguimiento , Humanos , Modelos Lineales , Estudios Longitudinales , Masculino , Persona de Mediana Edad , Análisis Multivariante , Evaluación de Resultado en la Atención de Salud , Autoinforme , Factores de Tiempo
5.
Int J Obes (Lond) ; 42(8): 1524-1531, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29980761

RESUMEN

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.


Asunto(s)
Índice de Masa Corporal , Peso Corporal/genética , Predisposición Genética a la Enfermedad/genética , Estudio de Asociación del Genoma Completo , Obesidad/genética , Humanos
6.
Mol Psychiatry ; 18(12): 1287-93, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23147384

RESUMEN

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.


Asunto(s)
Bulimia/tratamiento farmacológico , Conducta Alimentaria/efectos de los fármacos , Indanos/farmacología , Receptores Opioides mu/antagonistas & inhibidores , Triazoles/farmacología , Adolescente , Adulto , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Femenino , Humanos , Indanos/administración & dosificación , Indanos/uso terapéutico , Masculino , Persona de Mediana Edad , Triazoles/administración & dosificación , Triazoles/uso terapéutico , Adulto Joven
7.
Int J Obes (Lond) ; 37(5): 744-7, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-22614055

RESUMEN

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.


Asunto(s)
Aminoácidos Esenciales/metabolismo , Núcleo Arqueado del Hipotálamo/metabolismo , Glucosa/metabolismo , Oxigenasas de Función Mixta/metabolismo , Obesidad/metabolismo , Oxo-Ácido-Liasas/metabolismo , Proteínas/metabolismo , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato , Animales , Western Blotting , Línea Celular , Dieta Alta en Grasa , Regulación hacia Abajo , Regulación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Células HEK293 , Humanos , Ratones , Obesidad/genética , Obesidad/fisiopatología , Polimorfismo de Nucleótido Simple , ARN Mensajero/metabolismo , Factores de Tiempo
8.
Nat Genet ; 16(3): 303-6, 1997 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-9207799

RESUMEN

Human obesity has an inherited component, but in contrast to rodent obesity, precise genetic defects have yet to be defined. A mutation of carboxypeptidase E (CPE), an enzyme active in the processing and sorting of prohormones, causes obesity in the fat/fat mouse. We have previously described a women with extreme childhood obesity (Fig. 1), abnormal glucose homeostasis, hypogonadotrophic hypogonadism, hypocortisolism and elevated plasma proinsulin and pro-opiomelanocortin (POMC) concentrations but a very low insulin level, suggestive of a defective prohormone processing by the endopeptidase, prohormone convertase 1 (PC1; ref. 4). We now report this proband to be a compound heterozygote for mutations in PC1. Gly-->Arg483 prevents processing of proPC1 and leads to its retention in the endoplasmic reticulum (ER). A-->C+4 of the intro-5 donor splice site causes skipping of exon 5 leading to loss of 26 residues, a frameshift and creation of a premature stop codon within the catalytic domain. PC1 acts proximally to CPE in the pathway of post-translational processing of prohormones and neuropeptides. In view of the similarity between the proband and the fat/fat mouse phenotype, we infer that molecular defects in prohormone conversion may represent a generic mechanism for obesity, common to humans and rodents.


Asunto(s)
Ácido Aspártico Endopeptidasas/genética , Mutación , Obesidad/genética , Proproteína Convertasa 1 , Secuencia de Aminoácidos , Animales , Ácido Aspártico Endopeptidasas/metabolismo , Células CHO , Carboxipeptidasa H , Carboxipeptidasas/metabolismo , Cricetinae , Retículo Endoplásmico/enzimología , Femenino , Técnica del Anticuerpo Fluorescente , Heterocigoto , Humanos , Ratones , Ratones Endogámicos , Microscopía Fluorescente , Datos de Secuencia Molecular , Obesidad/enzimología , Reacción en Cadena de la Polimerasa , Polimorfismo Conformacional Retorcido-Simple , Proproteína Convertasas , Precursores de Proteínas/metabolismo , Procesamiento Proteico-Postraduccional , Empalme del ARN , ARN Mensajero/genética , Transfección
9.
Nat Genet ; 24(2): 153-6, 2000 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10655060

RESUMEN

The lipodystrophies are a group of disorders characterized by the absence or reduction of subcutaneous adipose tissue. Partial lipodystrophy (PLD; MIM 151660) is an inherited condition in which a regional (trunk and limbs) loss of fat occurs during the peri-pubertal phase. Additionally, variable degrees of resistance to insulin action, together with a hyperlipidaemic state, may occur and simulate the metabolic features commonly associated with predisposition to atherosclerotic disease. The PLD locus has been mapped to chromosome 1q with no evidence of genetic heterogeneity. We, and others, have refined the location to a 5.3-cM interval between markers D1S305 and D1S1600 (refs 5, 6). Through a positional cloning approach we have identified five different missense mutations in LMNA among ten kindreds and three individuals with PLD. The protein product of LMNA is lamin A/C, which is a component of the nuclear envelope. Heterozygous mutations in LMNA have recently been identified in kindreds with the variant form of muscular dystrophy (MD) known as autosomal dominant Emery-Dreifuss MD (EDMD-AD; ref. 7) and dilated cardiomyopathy and conduction-system disease (CMD1A). As LMNA is ubiquitously expressed, the finding of site-specific amino acid substitutions in PLD, EDMD-AD and CMD1A reveals distinct functional domains of the lamin A/C protein required for the maintenance and integrity of different cell types.


Asunto(s)
Cromosomas Humanos Par 1 , Lipodistrofia/genética , Proteínas Nucleares/genética , Mutación Puntual , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Secuencia de Bases , Mapeo Cromosómico , Femenino , Marcadores Genéticos , Heterocigoto , Humanos , Lamina Tipo A , Laminas , Masculino , Ratones , Datos de Secuencia Molecular , Proteínas Nucleares/química , Linaje , Ratas , Alineación de Secuencia , Homología de Secuencia de Aminoácido
10.
Nat Genet ; 28(4): 365-70, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11479539

RESUMEN

Congenital generalized lipodystrophy, or Berardinelli-Seip syndrome (BSCL), is a rare autosomal recessive disease characterized by a near-absence of adipose tissue from birth or early infancy and severe insulin resistance. Other clinical and biological features include acanthosis nigricans, hyperandrogenism, muscular hypertrophy, hepatomegaly, altered glucose tolerance or diabetes mellitus, and hypertriglyceridemia. A locus (BSCL1) has been mapped to 9q34 with evidence of heterogeneity. Here, we report a genome screen of nine BSCL families from two geographical clusters (in Lebanon and Norway). We identified a new disease locus, designated BSCL2, within the 2.5-Mb interval flanked by markers D11S4076 and D11S480 on chromosome 11q13. Analysis of 20 additional families of various ethnic origins led to the identification of 11 families in which the disease cosegregates with the 11q13 locus; the remaining families provide confirmation of linkage to 9q34. Sequence analysis of genes located in the 11q13 interval disclosed mutations in a gene homologous to the murine guanine nucleotide-binding protein (G protein), gamma3-linked gene (Gng3lg) in all BSCL2-linked families. BSCL2 is most highly expressed in brain and testis and encodes a protein (which we have called seipin) of unknown function. Most of the variants are null mutations and probably result in a severe disruption of the protein. These findings are of general importance for understanding the molecular mechanisms underlying regulation of body fat distribution and insulin resistance.


Asunto(s)
Cromosomas Humanos Par 11/genética , Subunidades gamma de la Proteína de Unión al GTP , Lipodistrofia/congénito , Lipodistrofia/genética , Proteínas/genética , Acantosis Nigricans/complicaciones , Cromosomas Humanos Par 9/genética , Análisis por Conglomerados , Análisis Mutacional de ADN , Complicaciones de la Diabetes , Femenino , Genes Recesivos , Ligamiento Genético , Marcadores Genéticos , Pruebas Genéticas , Haplotipos , Hepatomegalia/complicaciones , Proteínas de Unión al GTP Heterotriméricas/genética , Humanos , Hiperandrogenismo/complicaciones , Hipertrigliceridemia/complicaciones , Resistencia a la Insulina/genética , Líbano/epidemiología , Lipodistrofia/complicaciones , Lipodistrofia/epidemiología , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Mutación , Noruega/epidemiología , Especificidad de Órganos , Linaje , Estructura Terciaria de Proteína , Proteínas/metabolismo , Homología de Secuencia de Aminoácido
11.
bioRxiv ; 2023 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-37398065

RESUMEN

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.

12.
Int J Obes (Lond) ; 36(9): 1248-51, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22124452

RESUMEN

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.


Asunto(s)
Factor de Transcripción Activador 6/metabolismo , Adipogénesis , Retículo Endoplásmico/metabolismo , Respuesta de Proteína Desplegada , Factor de Transcripción Activador 6/genética , Animales , Diferenciación Celular , Línea Celular , Retículo Endoplásmico/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C3H , Regiones Promotoras Genéticas , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Respuesta de Proteína Desplegada/genética , eIF-2 Quinasa/metabolismo
13.
Diabetologia ; 54(5): 1057-65, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21318406

RESUMEN

AIMS/HYPOTHESIS: Genetic insulin receptoropathies are a rare cause of severe insulin resistance. We identified the Ile119Met missense mutation in the insulin receptor INSR gene, previously reported in a Yemeni kindred, in four unrelated patients with Somali ancestry. We aimed to investigate a possible genetic founder effect, and to study the mechanism of loss of function of the mutant receptor. METHODS: Biochemical profiling and DNA haplotype analysis of affected patients were performed. Insulin receptor expression in lymphoblastoid cells from a homozygous p.Ile119Met INSR patient, and in cells heterologously expressing the mutant receptor, was examined. Insulin binding, insulin-stimulated receptor autophosphorylation, and cooperativity and pH dependency of insulin dissociation were also assessed. RESULTS: All patients had biochemical profiles pathognomonic of insulin receptoropathy, while haplotype analysis revealed the putative shared region around the INSR mutant to be no larger than 28 kb. An increased insulin proreceptor to ß subunit ratio was seen in patient-derived cells. Steady state insulin binding and insulin-stimulated autophosphorylation of the mutant receptor was normal; however it exhibited decreased insulin dissociation rates with preserved cooperativity, a difference accentuated at low pH. CONCLUSIONS/INTERPRETATION: The p.Ile119Met INSR appears to have arisen around the Horn of Africa, and should be sought first in severely insulin resistant patients with ancestry from this region. Despite collectively compelling genetic, clinical and biochemical evidence for its pathogenicity, loss of function in conventional in vitro assays is subtle, suggesting mildly impaired receptor recycling only.


Asunto(s)
Resistencia a la Insulina/fisiología , Receptor de Insulina/genética , Receptor de Insulina/metabolismo , Adulto , África , Células Cultivadas , Niño , Femenino , Haplotipos , Humanos , Lactante , Resistencia a la Insulina/genética , Masculino , Mutagénesis Sitio-Dirigida , Mutación , Reacción en Cadena de la Polimerasa , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Adulto Joven
14.
Diabet Med ; 28(12): 1476-86, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21992440

RESUMEN

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.


Asunto(s)
Tejido Adiposo/metabolismo , Diabetes Mellitus Tipo 2/genética , Resistencia a la Insulina/genética , Obesidad/genética , Receptor de Insulina/genética , Tejido Adiposo/patología , Diabetes Mellitus Tipo 2/etiología , Humanos , Lipodistrofia/genética , Lipodistrofia/metabolismo , Obesidad/complicaciones , Obesidad/patología , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Receptor de Insulina/metabolismo
15.
Mol Metab ; 43: 101127, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33242659

RESUMEN

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.


Asunto(s)
Apetito/genética , Apetito/fisiología , Conducta Alimentaria/fisiología , Animales , Índice de Masa Corporal , Encéfalo , Drosophila melanogaster/genética , Metabolismo Energético , Estudio de Asociación del Genoma Completo , Genotipo , Homeostasis , Hipotálamo/metabolismo , Neuronas/metabolismo , Estado Nutricional , Obesidad/metabolismo , Fenotipo
16.
Int J Obes (Lond) ; 34(1): 190-4, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19844213

RESUMEN

CONTEXT: The PNPLA3 I148M variant (rs738409) is robustly associated with hepatic steatosis. Intriguingly, initial findings in cohorts with a mean body mass index (BMI) of 30 kg m(-2) also suggested that it is associated with elevated liver enzymes but not with insulin resistance and dyslipidaemia. OBJECTIVE: To determine whether the PNPLA3 variant alters the susceptibility of morbidly obese subjects to develop liver injury and metabolic sequelae. PARTICIPANTS AND METHODS: The study was carried out in 678 obese Italians (mean BMI = 41 kg m(-2)) who were genotyped for the I148M variant. All participants provided fasting blood samples and then underwent oral glucose tolerance tests. MAIN OUTCOME MEASURES: Indices of liver injury (alanine transaminase (ALT), aspartate transaminase (AST)), glucose tolerance and insulin resistance were measured. RESULTS: Markers of hepatic injury such as ALT and AST were significantly higher in carriers of the 148M allele (P = 2.2 x 10(-5) and 0.001, respectively). In all, 50% of 148M risk allele homozygotes had pathological levels of ALT (>40 U l(-1)) compared with 25% of 148I allele homozygotes (P = 0.005). Glucose tolerance and insulin sensitivity were similar in all three genotypes. CONCLUSION: Obese Southern Europeans carrying the 148M allele have increased indices of liver damage uncoupled from proxy measures of insulin resistance.


Asunto(s)
Hígado Graso/enzimología , Variación Genética/genética , Resistencia a la Insulina/genética , Lipasa/genética , Proteínas de la Membrana/genética , Obesidad Mórbida/genética , Adulto , Alanina Transaminasa/sangre , Aspartato Aminotransferasas/sangre , Biomarcadores/sangre , Índice de Masa Corporal , Hígado Graso/sangre , Femenino , Predisposición Genética a la Enfermedad/genética , Genotipo , Prueba de Tolerancia a la Glucosa , Humanos , Italia , Lipasa/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Obesidad Mórbida/sangre , Obesidad Mórbida/etnología
17.
J Inherit Metab Dis ; 33 Suppl 3: S227-32, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20521171

RESUMEN

In Npc1 null mice, a model for Niemann Pick Disease Type C1, it has been reported that hepatocyte insulin receptor function is significantly impaired, consistent with growing evidence that membrane fluidity and microdomain structure have an important role in insulin signal transduction. However, whether insulin receptor function is also compromised in human Niemann Pick disease Type C1 is unclear. We now report a girl who developed progressive dementia, ataxia and opthalmoplegia from 9 years old, followed by severe acanthosis nigricans, hirsutism and acne at 11 years old. She was diagnosed with Niemann Pick Disease type C1 (OMIM#257220) based on positive filipin staining and reduced cholesterol-esterifying activity in dermal fibroblasts, and homozygosity for the p.Ile1061Thr NPC1 mutation. Further analysis revealed her also to be heterozygous for a novel trinucleotide deletion (c.3659 + 1_3659 + 3delGTG) at the end of exon 20 of INSR, encoding the insulin receptor, leading to deletion of Trp1193 in the intracellular tyrosine kinase domain. INSR mRNA and protein levels were normal in dermal fibroblasts, consistent with a primary signal transduction defect in the mutant receptor. Although the proband was significantly more insulin resistant than her father, who carried the INSR mutation but was only heterozygous for the NPC1 variant, their respective degrees of IR were very similar to those previously reported in a father-daughter pair with the closely related p.Trp1193Leu INSR mutation. This suggests that loss of NPC1 function, with attendant changes in membrane cholesterol composition, does not significantly modify the IR phenotype, even in the context of severely impaired INSR function.


Asunto(s)
Antígenos CD/genética , Proteínas Portadoras/genética , Resistencia a la Insulina/genética , Glicoproteínas de Membrana/genética , Mutación , Enfermedad de Niemann-Pick Tipo C/genética , Receptor de Insulina/genética , Adulto , Anciano , Secuencia de Aminoácidos , Antígenos CD/metabolismo , Biomarcadores/sangre , Células Cultivadas , Niño , Análisis Mutacional de ADN , Femenino , Fibroblastos/metabolismo , Predisposición Genética a la Enfermedad , Herencia , Heterocigoto , Homocigoto , Humanos , Péptidos y Proteínas de Señalización Intracelular , Masculino , Datos de Secuencia Molecular , Proteína Niemann-Pick C1 , Enfermedad de Niemann-Pick Tipo C/sangre , Enfermedad de Niemann-Pick Tipo C/diagnóstico , Linaje , Fenotipo , ARN Mensajero/metabolismo , Receptor de Insulina/metabolismo , Índice de Severidad de la Enfermedad
18.
Diabetologia ; 52(11): 2345-2349, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19756488

RESUMEN

AIMS/HYPOTHESIS: Obesity is the dominant cause of insulin resistance. In adult humans it is characterised by a combination of adipocyte hypertrophy and, to a lesser extent, adipocyte hyperplasia. As hypertrophic adipocytes secrete more leptin and less adiponectin, the plasma leptin:adiponectin ratio (LAR) has been proposed as a potentially useful measure of insulin resistance and vascular risk. We sought to assess the usefulness of the LAR as a measure of insulin resistance in non-diabetic white adults. METHODS: Leptin and adiponectin levels were measured in 2,097 non-diabetic individuals from the Ely and European Group for the Study of Insulin Resistance (EGIR) Relationship between Insulin Sensitivity and Cardiovascular Risk (RISC) study cohorts. LAR was compared with fasting insulin and HOMA-derived insulin sensitivity (HOMA-S) in all individuals and with the insulin sensitivity index (M/I) from hyperinsulinaemic-euglycaemic clamp studies in 1,226 EGIR RISC participants. RESULTS: The LAR was highly correlated with HOMA-S in men (r = -0.58, p = 4.5 x 10(-33) and r = -0.65, p = 1.1 x 10(-66) within the Ely and EGIR RISC study cohorts, respectively) and in women (r = -0.51, p = 2.8 x 10(-36) and r = -0.61, p = 2.5 x 10(-73)). The LAR was also strongly correlated with the clamp M/I value (r = -0.52, p = 4.5 x 10(-38) and r = -0.47, p = 6.6 x 10(-40) in men and women, respectively), similar to correlations between HOMA-S and the M/I value. CONCLUSIONS/INTERPRETATION: The leptin:adiponectin ratio is a useful measure of insulin resistance in non-diabetic white adults. These data highlight the central role of adipocyte dysfunction in the pathogenesis of insulin resistance. Given that variations between fasting and postprandial leptin and adiponectin levels tend to be small, the leptin to adiponectin ratio might also have potential value in assessing insulin sensitivity in the non-fasted state.


Asunto(s)
Adiponectina/sangre , Resistencia a la Insulina/fisiología , Leptina/sangre , Adipocitos/fisiología , Adulto , Glucemia/metabolismo , Estudios de Cohortes , Femenino , Técnica de Clampeo de la Glucosa , Humanos , Masculino , Persona de Mediana Edad , Factores de Riesgo , Caracteres Sexuales , Enfermedades Vasculares/epidemiología , Población Blanca
19.
Diabet Med ; 26(1): 79-82, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19125765

RESUMEN

Anti-insulin antibodies have been described in two contexts: in insulin-naive individuals (so-called 'insulin autoimmune syndrome') and in patients with insulin-treated diabetes, in whom antibodies are rarely of clinical significance. We report the case of an 68-year-old woman who exhibited a local allergic reaction to subcutaneous insulin followed by severe insulin resistance, evidenced by poor glycaemic control despite treatment with > 3.5 U/kg of insulin per day. She was found to have circulating polyclonal anti-insulin antibodies of the IgG subtype and responded clinically to a course of plasma exchange and immunosuppression with mycophenolate mofetil and, subsequently, intravenous immunoglobulin. Falling titres of antibodies on this regimen correlated with improved glycaemic control. This case suggests that clinicians should be alert to the possibility of insulin resistance due to anti-insulin antibodies and that immunosuppression in this situation may be a valuable therapeutic option.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Inmunosupresores/uso terapéutico , Anticuerpos Insulínicos/inmunología , Resistencia a la Insulina/inmunología , Insulina/inmunología , Ácido Micofenólico/análogos & derivados , Anciano , Reacciones Antígeno-Anticuerpo/inmunología , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/inmunología , Femenino , Humanos , Inyecciones Subcutáneas/métodos , Insulina/sangre , Anticuerpos Insulínicos/sangre , Ácido Micofenólico/uso terapéutico , Intercambio Plasmático/métodos
20.
Int J Obes (Lond) ; 32 Suppl 7: S55-61, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19136992

RESUMEN

In the spirit of celebration associated with the 20th anniversary of the Pennington Biomedical Research Center, we have seized the opportunity of taking a highly personal and not at all comprehensive 'whistle-stop tour' of a large body of evidence that, we feel, supports the following conclusions: (1) that body fat stores are regulated by biological control processes in humans as they are in lower animals; (2) that there are major inherited influences on the efficiency whereby such control processes operate in humans; (3) that the precise nature of those genetic and biological influences and how they interact with environmental factors are beginning to be understood; (4) that most of the genes discovered thus far have their principal impact on hunger, satiety and food intake; (5) that while there is understandable resistance to the notion that genes can influence a human behavior such as the habitual ingestion of food, the implications of these discoveries are essentially benign. Indeed, we hope that they may eventually lead to improved treatment for patients and, in addition, help to inculcate a more enlightened attitude to the obese with a reduction in their experience of social and economic discrimination.


Asunto(s)
Metabolismo Energético , Obesidad/genética , Adiposidad/genética , Adiposidad/fisiología , Índice de Masa Corporal , Conducta Alimentaria , Predisposición Genética a la Enfermedad , Humanos , Hambre/fisiología , Leptina/genética , Leptina/fisiología , Obesidad/fisiopatología , Respuesta de Saciedad/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA