Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 247
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 187(15): 3877-3879, 2024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-39059363

RESUMEN

In this interview with Cell, Dr. Giles Yeo shares his journey into metabolism research, his strategies for engaging audiences, and the challenges of simplifying complex concepts. He also discusses the impact of social media, the advantages of podcasting, and the question he's most afraid of.


Asunto(s)
Medios de Comunicación Sociales , Humanos , Comunicación , Metabolismo , Historia del Siglo XXI
2.
Cell ; 164(3): 353-64, 2016 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-26824653

RESUMEN

More than one-half billion people are obese, and despite progress in genetic research, much of the heritability of obesity remains enigmatic. Here, we identify a Trim28-dependent network capable of triggering obesity in a non-Mendelian, "on/off" manner. Trim28(+/D9) mutant mice exhibit a bi-modal body-weight distribution, with isogenic animals randomly emerging as either normal or obese and few intermediates. We find that the obese-"on" state is characterized by reduced expression of an imprinted gene network including Nnat, Peg3, Cdkn1c, and Plagl1 and that independent targeting of these alleles recapitulates the stochastic bi-stable disease phenotype. Adipose tissue transcriptome analyses in children indicate that humans too cluster into distinct sub-populations, stratifying according to Trim28 expression, transcriptome organization, and obesity-associated imprinted gene dysregulation. These data provide evidence of discrete polyphenism in mouse and man and thus carry important implications for complex trait genetics, evolution, and medicine.


Asunto(s)
Epigénesis Genética , Haploinsuficiencia , Proteínas Nucleares/genética , Obesidad/genética , Proteínas Represoras/genética , Delgadez/genética , Adolescente , Animales , Índice de Masa Corporal , Niño , Preescolar , Humanos , Ratones , Encuestas Nutricionales , Polimorfismo Genético , Proteína 28 que Contiene Motivos Tripartito
3.
Nat Rev Genet ; 23(2): 120-133, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34556834

RESUMEN

The prevalence of obesity has tripled over the past four decades, imposing an enormous burden on people's health. Polygenic (or common) obesity and rare, severe, early-onset monogenic obesity are often polarized as distinct diseases. However, gene discovery studies for both forms of obesity show that they have shared genetic and biological underpinnings, pointing to a key role for the brain in the control of body weight. Genome-wide association studies (GWAS) with increasing sample sizes and advances in sequencing technology are the main drivers behind a recent flurry of new discoveries. However, it is the post-GWAS, cross-disciplinary collaborations, which combine new omics technologies and analytical approaches, that have started to facilitate translation of genetic loci into meaningful biology and new avenues for treatment.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Variación Genética , Genoma Humano/genética , Estudio de Asociación del Genoma Completo/métodos , Obesidad/genética , Secuenciación Completa del Genoma/métodos , Animales , Ingestión de Alimentos/genética , Interacción Gen-Ambiente , Humanos , Herencia Multifactorial/genética , Sobrepeso/genética
4.
Nature ; 578(7795): 444-448, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31875646

RESUMEN

Metformin, the world's most prescribed anti-diabetic drug, is also effective in preventing type 2 diabetes in people at high risk1,2. More than 60% of this effect is attributable to the ability of metformin to lower body weight in a sustained manner3. The molecular mechanisms by which metformin lowers body weight are unknown. Here we show-in two independent randomized controlled clinical trials-that metformin increases circulating levels of the peptide hormone growth/differentiation factor 15 (GDF15), which has been shown to reduce food intake and lower body weight through a brain-stem-restricted receptor. In wild-type mice, oral metformin increased circulating GDF15, with GDF15 expression increasing predominantly in the distal intestine and the kidney. Metformin prevented weight gain in response to a high-fat diet in wild-type mice but not in mice lacking GDF15 or its receptor GDNF family receptor α-like (GFRAL). In obese mice on a high-fat diet, the effects of metformin to reduce body weight were reversed by a GFRAL-antagonist antibody. Metformin had effects on both energy intake and energy expenditure that were dependent on GDF15, but retained its ability to lower circulating glucose levels in the absence of GDF15 activity. In summary, metformin elevates circulating levels of GDF15, which is necessary to obtain its beneficial effects on energy balance and body weight, major contributors to its action as a chemopreventive agent.


Asunto(s)
Peso Corporal/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Factor 15 de Diferenciación de Crecimiento/metabolismo , Metformina/farmacología , Administración Oral , Adulto , Anciano , Animales , Glucemia/análisis , Glucemia/metabolismo , Dieta Alta en Grasa , Método Doble Ciego , Ingestión de Energía/efectos de los fármacos , Enterocitos/citología , Enterocitos/efectos de los fármacos , Femenino , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/antagonistas & inhibidores , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/deficiencia , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Factor 15 de Diferenciación de Crecimiento/sangre , Factor 15 de Diferenciación de Crecimiento/deficiencia , Factor 15 de Diferenciación de Crecimiento/genética , Homeostasis/efectos de los fármacos , Humanos , Intestinos/citología , Intestinos/efectos de los fármacos , Masculino , Metformina/administración & dosificación , Ratones , Ratones Obesos , Persona de Mediana Edad , Pérdida de Peso/efectos de los fármacos
6.
PLoS Med ; 21(7): e1004437, 2024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-39052689

RESUMEN

In the Editorial, Giles Yeo discusses if it is possible that the UPF concept could be doing more harm than good.

7.
Diabetes Metab Res Rev ; 40(5): e3829, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38850100

RESUMEN

AIMS: Pancreatic polypeptide (PP) is elevated in people with vascular risk factors such as type 2 diabetes or increased visceral fat. We investigated potential relationships between PP and microvascular and macrovascular complications of diabetes. MATERIALS AND METHODS: Animal study: Subcutaneous PP infusion for 4 weeks in high fat diet mouse model. Retinal mRNA submitted for Ingenuity Pathway Analysis. Human study: fasting PP measured in 1478 participants and vascular complications recorded over median 5.5 (IQR 4.9-5.8) years follow-up. RESULTS: Animal study: The retinal transcriptional response to PP was indicative of cellular stress and damage, and this footprint matched responses described in previously published studies of retinal disease. Of mechanistic importance the transcriptional landscape was consistent with upregulation of folliculin, a recently identified susceptibility gene for diabetic retinopathy. Human study: Adjusting for established risk factors, PP was associated with prevalent and incident clinically significant retinopathy (odds ratio (OR) 1.289 (1.107-1.501) p = 0.001; hazard ratio (HR) 1.259 (1.035-1.531) p = 0.0213), albuminuria (OR 1.277 (1.124-1.454), p = 0.0002; HR 1.608 (1.208-2.141) p = 0.0011), and macrovascular disease (OR 1.021 (1.006-1.037) p = 0.0068; HR 1.324 (1.089-1.61), p = 0.0049), in individuals with type 2 diabetes, and progression to diabetes in non-diabetic individuals (HR 1.402 (1.081-1.818), p = 0.0109). CONCLUSIONS: Elevated fasting PP is independently associated with vascular complications of diabetes and affects retinal pathways potentially influencing retinal neuronal survival. Our results suggest possible new roles for PP-fold peptides in the pathophysiology of diabetes complications and vascular risk stratification.


Asunto(s)
Diabetes Mellitus Tipo 2 , Angiopatías Diabéticas , Retinopatía Diabética , Ayuno , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/patología , Humanos , Masculino , Femenino , Persona de Mediana Edad , Angiopatías Diabéticas/etiología , Angiopatías Diabéticas/epidemiología , Animales , Ratones , Estudios de Seguimiento , Retinopatía Diabética/etiología , Retinopatía Diabética/epidemiología , Retinopatía Diabética/patología , Pronóstico , Incidencia , Biomarcadores/análisis , Factores de Riesgo , Anciano
8.
Proc Natl Acad Sci U S A ; 118(27)2021 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-34187898

RESUMEN

An acute increase in the circulating concentration of glucocorticoid hormones is essential for the survival of severe somatic stresses. Circulating concentrations of GDF15, a hormone that acts in the brain to reduce food intake, are frequently elevated in stressful states. We now report that GDF15 potently activates the hypothalamic-pituitary-adrenal (HPA) axis in mice and rats. A blocking antibody to the GDNF-family receptor α-like receptor completely prevented the corticosterone response to GDF15 administration. In wild-type mice exposed to a range of stressful stimuli, circulating levels of both corticosterone and GDF15 rose acutely. In the case of Escherichia coli or lipopolysaccharide injections, the vigorous proinflammatory cytokine response elicited was sufficient to produce a near-maximal HPA response, regardless of the presence or absence of GDF15. In contrast, the activation of the HPA axis seen in wild-type mice in response to the administration of genotoxic or endoplasmic reticulum toxins, which do not provoke a marked rise in cytokines, was absent in Gdf15-/- mice. In conclusion, consistent with its proposed role as a sentinel hormone, endogenous GDF15 is required for the activation of the protective HPA response to toxins that do not induce a substantial cytokine response. In the context of efforts to develop GDF15 as an antiobesity therapeutic, these findings identify a biomarker of target engagement and a previously unrecognized pharmacodynamic effect, which will require monitoring in human studies.


Asunto(s)
Factor 15 de Diferenciación de Crecimiento/metabolismo , Sistema Hipotálamo-Hipofisario/metabolismo , Sistema Hipófiso-Suprarrenal/metabolismo , Animales , Cisplatino/administración & dosificación , Cisplatino/farmacología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Glucocorticoides/metabolismo , Factor 15 de Diferenciación de Crecimiento/administración & dosificación , Humanos , Lipopolisacáridos , Ratones , Ratas , Tunicamicina/farmacología
9.
Hum Mutat ; 43(11): 1545-1556, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36047340

RESUMEN

De novo mutations (DNMs) play an important role in severe genetic disorders that reduce fitness. To better understand their role in disease, it is important to determine the parent-of-origin and timing of mutational events that give rise to these mutations, especially in sex-specific developmental disorders such as male infertility. However, currently available short-read sequencing approaches are not ideally suited for phasing, as this requires long continuous DNA strands that span both the DNM and one or more informative single-nucleotide polymorphisms. To overcome these challenges, we optimized and implemented a multiplexed long-read sequencing approach using Oxford Nanopore technologies MinION platform. We focused on improving target amplification, integrating long-read sequenced data with high-quality short-read sequence data, and developing an anchored phasing computational method. This approach handled the inherent phasing challenges of long-range target amplification and the normal accumulation of sequencing error associated with long-read sequencing. In total, 77 of 109 DNMs (71%) were successfully phased and parent-of-origin identified. The majority of phased DNMs were prezygotic (90%), the accuracy of which is highlighted by an average mutant allele frequency of 49.6% and standard error of 0.84%. This study demonstrates the benefits of employing an integrated short-read and long-read sequencing approach for large-scale DNM phasing.


Asunto(s)
Secuenciación de Nucleótidos de Alto Rendimiento , Femenino , Humanos , Masculino , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Mutación , Análisis de Secuencia de ADN/métodos , Polimorfismo de Nucleótido Simple
10.
Circ Res ; 125(11): 1019-1034, 2019 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-31610723

RESUMEN

RATIONALE: Atherosclerosis is a chronic inflammatory disease. Recent studies have shown that dysfunctional autophagy in endothelial cells, smooth muscle cells, and macrophages, plays a detrimental role during atherogenesis, leading to the suggestion that autophagy-stimulating approaches may provide benefit. OBJECTIVE: Dendritic cells (DCs) are at the crossroad of innate and adaptive immune responses and profoundly modulate the development of atherosclerosis. Intriguingly, the role of autophagy in DC function during atherosclerosis and how the autophagy process would impact disease development has not been addressed. METHODS AND RESULTS: Here, we show that the autophagic flux in atherosclerosis-susceptible Ldlr-/- (low-density lipoprotein receptor-deficient) mice is substantially higher in splenic and aortic DCs compared with macrophages and is further activated under hypercholesterolemic conditions. RNA sequencing and functional studies on selective cell populations reveal that disruption of autophagy through deletion of Atg16l1 differentially affects the biology and functions of DC subsets in Ldlr-/- mice under high-fat diet. Atg16l1 deficient CD11b+ DCs develop a TGF (transforming growth factor)-ß-dependent tolerogenic phenotype and promote the expansion of regulatory T cells, whereas no such effects are seen with Atg16l1 deficient CD8α+ DCs. Atg16l1 deletion in DCs (all CD11c-expressing cells) expands aortic regulatory T cells in vivo, limits the accumulation of T helper cells type 1, and reduces the development of atherosclerosis in Ldlr-/- mice. In contrast, no such effects are seen when Atg16l1 is deleted selectively in conventional CD8α+ DCs and CD103+ DCs. Total T-cell or selective regulatory T-cell depletion abrogates the atheroprotective effect of Atg16l1 deficient DCs. CONCLUSIONS: In contrast to its proatherogenic role in macrophages, autophagy disruption in DCs induces a counter-regulatory response that maintains immune homeostasis in Ldlr-/- mice under high-fat diet and limits atherogenesis. Selective modulation of autophagy in DCs could constitute an interesting therapeutic target in atherosclerosis.


Asunto(s)
Aorta/inmunología , Enfermedades de la Aorta/prevención & control , Aterosclerosis/prevención & control , Autofagia , Antígeno CD11b/inmunología , Comunicación Celular , Proliferación Celular , Células Dendríticas/inmunología , Activación de Linfocitos , Linfocitos T Reguladores/inmunología , Animales , Aorta/metabolismo , Aorta/patología , Enfermedades de la Aorta/inmunología , Enfermedades de la Aorta/metabolismo , Enfermedades de la Aorta/patología , Aterosclerosis/inmunología , Aterosclerosis/metabolismo , Aterosclerosis/patología , Proteína 5 Relacionada con la Autofagia/metabolismo , Proteínas Relacionadas con la Autofagia/genética , Proteínas Relacionadas con la Autofagia/metabolismo , Trasplante de Médula Ósea , Antígenos CD11/genética , Antígenos CD11/metabolismo , Antígeno CD11b/metabolismo , Células Cultivadas , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Femenino , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Placa Aterosclerótica , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Receptores de LDL/genética , Receptores de LDL/metabolismo , Transducción de Señal , Linfocitos T Reguladores/metabolismo
11.
Prenat Diagn ; 41(2): 258-270, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33251640

RESUMEN

BACKGROUND: Posterior fossa abnormalities (PFAs) are commonly identified within routine screening and are a frequent indication for fetal magnetic resonance imaging (MRI). Although biometric measurements of the posterior fossa (PF) are established on fetal ultrasound and MRI, qualitative visual assessments are predominantly used to differentiate PFAs. OBJECTIVES: This systematic review aimed to assess 2-dimensional (2D) biometric measurements currently in use for assessing the PF on fetal MRI to delineate different PFAs. METHODS: The protocol was registered (PROSPERO ID CRD42019142162). Eligible studies included T2-weighted MRI PF measurements in fetuses with and without PFAs, including measurements of the PF, or other brain areas relevant to PFAs. RESULTS: 59 studies were included - 6859 fetuses had 62 2D PF and related measurements. These included linear, area and angular measurements, representing measures of PF size, cerebellum/vermis, brainstem, and supratentorial measurements. 11 measurements were used in 10 or more studies and at least 1200 fetuses. These dimensions were used to characterise normal for gestational age, diagnose a range of pathologies, and predict outcome. CONCLUSION: A selection of validated 2D biometric measurements of the PF on fetal MRI may be useful for identification of PFA in different clinical settings. Consistent use of these measures, both clinically and for research, is recommended.


Asunto(s)
Tronco Encefálico/diagnóstico por imagen , Cerebelo/diagnóstico por imagen , Fosa Craneal Posterior/diagnóstico por imagen , Feto/diagnóstico por imagen , Imagen por Resonancia Magnética , Biometría , Tronco Encefálico/anomalías , Cerebelo/anomalías , Fosa Craneal Posterior/anomalías , Femenino , Humanos , Tamaño de los Órganos , Embarazo , Ultrasonografía Prenatal
12.
Int J Obes (Lond) ; 44(9): 1946-1957, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32719434

RESUMEN

BACKGROUND: Though it is well established that neonatal nutrition plays a major role in lifelong offspring health, the mechanisms underpinning this have not been well defined. Early postnatal accelerated growth resulting from maternal nutritional status is associated with increased appetite and body weight. Likewise, slow growth correlates with decreased appetite and body weight. Food consumption and food-seeking behaviour are directly modulated by central serotonergic (5-hydroxytryptamine, 5-HT) pathways. This study examined the effect of a rat maternal postnatal low protein (PLP) diet on 5-HT receptor mediated food intake in offspring. METHODS: Microarray analyses, in situ hybridization or laser capture microdissection of the ARC followed by RT-PCR were used to identify genes up- or down-regulated in the arcuate nucleus of the hypothalamus (ARC) of 3-month-old male PLP rats. Third ventricle cannulation was used to identify altered sensitivity to serotonin receptor agonists and antagonists with respect to food intake. RESULTS: Male PLP offspring consumed less food and had lower growth rates up to 3 months of age compared with Control offspring from dams fed a normal diet. In total, 97 genes were upregulated including the 5-HT5A receptor (5-HT5AR) and 149 downregulated genes in PLP rats compared with Controls. The former obesity medication fenfluramine and the 5-HT receptor agonist 5-Carboxamidotryptamine (5-CT) significantly suppressed food intake in both groups, but the PLP offspring were more sensitive to d-fenfluramine and 5-CT compared with Controls. The effect of 5-CT was antagonized by the 5-HT5AR antagonist SB699551. 5-CT also reduced NPY-induced hyperphagia in both Control and PLP rats but was more effective in PLP offspring. CONCLUSIONS: Postnatal low protein programming of growth in rats enhances the central effects of serotonin on appetite by increasing hypothalamic 5-HT5AR expression and sensitivity. These findings provide insight into the possible mechanisms through which a maternal low protein diet during lactation programs reduced growth and appetite in offspring.


Asunto(s)
Apetito/fisiología , Peso Corporal/fisiología , Regulación del Desarrollo de la Expresión Génica/genética , Hipotálamo/metabolismo , Receptores de Serotonina , Animales , Dieta , Femenino , Masculino , Obesidad/metabolismo , Ratas , Receptores de Serotonina/genética , Receptores de Serotonina/metabolismo , Transcriptoma/genética
13.
Mol Cell ; 48(1): 39-51, 2012 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-22981861

RESUMEN

Endoplasmic reticulum (ER) thiol oxidases initiate a disulfide relay to oxidatively fold secreted proteins. We found that combined loss-of-function mutations in genes encoding the ER thiol oxidases ERO1α, ERO1ß, and PRDX4 compromised the extracellular matrix in mice and interfered with the intracellular maturation of procollagen. These severe abnormalities were associated with an unexpectedly modest delay in disulfide bond formation in secreted proteins but a profound, 5-fold lower procollagen 4-hydroxyproline content and enhanced cysteinyl sulfenic acid modification of ER proteins. Tissue ascorbic acid content was lower in mutant mice, and ascorbic acid supplementation improved procollagen maturation and lowered sulfenic acid content in vivo. In vitro, the presence of a sulfenic acid donor accelerated the oxidative inactivation of ascorbate by an H(2)O(2)-generating system. Compromised ER disulfide relay thus exposes protein thiols to competing oxidation to sulfenic acid, resulting in depletion of ascorbic acid, impaired procollagen proline 4-hydroxylation, and a noncanonical form of scurvy.


Asunto(s)
Ácido Ascórbico/metabolismo , Retículo Endoplásmico/metabolismo , Glicoproteínas/metabolismo , Oxidorreductasas/metabolismo , Escorbuto/etiología , Escorbuto/metabolismo , Animales , Ácido Ascórbico/farmacología , Células Cultivadas , Tejido Conectivo/metabolismo , Tejido Conectivo/patología , Modelos Animales de Enfermedad , Disulfuros/metabolismo , Femenino , Glicoproteínas/deficiencia , Glicoproteínas/genética , Masculino , Ratones , Ratones Mutantes , Mutación , Oxidación-Reducción , Oxidorreductasas/deficiencia , Oxidorreductasas/genética , Peroxirredoxinas/deficiencia , Peroxirredoxinas/genética , Peroxirredoxinas/metabolismo , Procolágeno/metabolismo , Pliegue de Proteína , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Escorbuto/genética , Escorbuto/patología , Ácidos Sulfénicos/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
14.
Proc Natl Acad Sci U S A ; 114(35): 9421-9426, 2017 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-28811369

RESUMEN

An intergenic region of human chromosome 2 (2p25.3) harbors genetic variants which are among those most strongly and reproducibly associated with obesity. The gene closest to these variants is TMEM18, although the molecular mechanisms mediating these effects remain entirely unknown. Tmem18 expression in the murine hypothalamic paraventricular nucleus (PVN) was altered by changes in nutritional state. Germline loss of Tmem18 in mice resulted in increased body weight, which was exacerbated by high fat diet and driven by increased food intake. Selective overexpression of Tmem18 in the PVN of wild-type mice reduced food intake and also increased energy expenditure. We provide evidence that TMEM18 has four, not three, transmembrane domains and that it physically interacts with key components of the nuclear pore complex. Our data support the hypothesis that TMEM18 itself, acting within the central nervous system, is a plausible mediator of the impact of adjacent genetic variation on human adiposity.


Asunto(s)
Apetito/genética , Peso Corporal/genética , Proteínas de la Membrana/metabolismo , Obesidad/genética , Animales , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Núcleo Hipotalámico Paraventricular/metabolismo , Proteínas de Transporte Vesicular
16.
Hematol Oncol ; 36(1): 320-323, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28401573

RESUMEN

Castleman disease is a rare lymphoproliferative disorder with 2 distinctly defined clinical forms. While multicentric Castleman disease (UCD) poses a potential therapeutic challenge, unicentric variant has historically been considered curable with surgical resection. Hence, little is known to guide management of patients with UCD, refractory to surgical resection and combination chemotherapy. We present a case of a patient, negative for HIV and HHV-8, who had an unsuccessful surgical intervention and no response to radiotherapy and chemotherapy. He had severe paraneoplastic pemphigus and was treated with tocilizumab, an anti-interleukin-6 receptor monoclonal antibody that has demonstrated good response rates in multicentric Castleman disease but demonstrated no clinical response despite 2 months of treatment. Our report is the first to describe a lack of response to tocilizumab in the rare setting of refractory UCD and discuss potential for distinct disease biology.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Enfermedad de Castleman/tratamiento farmacológico , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales Humanizados/farmacología , Enfermedad de Castleman/patología , Humanos , Masculino , Persona de Mediana Edad
19.
Gut ; 66(11): 1926-1935, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-27558924

RESUMEN

OBJECTIVE: Myelosuppression is a life-threatening complication of thiopurine therapy, and the incidence of thiopurine-induced myelosuppression is higher in East Asians than in Europeans. We investigated genetic factors associated with thiopurine-induced leukopenia in patients with IBD. DESIGN: A genome-wide association study (GWAS) was conducted in thiopurine-treated patients with IBD, followed by high-throughput sequencing of genes identified as significant in the GWAS or those involved in thiopurine metabolism (n=331). Significant loci associated with thiopurine-induced leukopenia were validated in two additional replication cohorts (n=437 and n=330). Functional consequences of FTO (fat mass and obesity-associated) variant were examined both in vitro and in vivo. RESULTS: The GWAS identified two loci associated with thiopurine-induced leukopenia (rs16957920, FTO intron; rs2834826, RUNX1 intergenic). High-throughput targeted sequencing indicated that an FTO coding variant (rs79206939, p.A134T) linked to rs16957920 is associated with thiopurine-induced leukopenia. This result was further validated in two replication cohorts (combined p=1.3×10-8, OR=4.3). The frequency of FTO p.A134T is 5.1% in Koreans but less than 0.1% in Western populations. The p.A134T variation reduced FTO activity by 65% in the nucleotide demethylase assay. In vivo experiments revealed that Fto-/- and Fto+/- mice were more susceptible to thiopurine-induced myelosuppression than wild-type mice. CONCLUSIONS: The results suggest that the hypomorphic FTO p.A134T variant is associated with thiopurine-induced leukopenia. These results shed light on the novel physiological role of FTO and provide a potential pharmacogenetic biomarker for thiopurine therapy.


Asunto(s)
Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/genética , Azatioprina/efectos adversos , Inmunosupresores/efectos adversos , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Leucopenia/inducido químicamente , Mercaptopurina/efectos adversos , Polimorfismo de Nucleótido Simple , Adolescente , Adulto , Anciano , Animales , Azatioprina/uso terapéutico , Estudios de Casos y Controles , Estudios de Cohortes , Femenino , Marcadores Genéticos , Estudio de Asociación del Genoma Completo , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Inmunosupresores/uso terapéutico , Enfermedades Inflamatorias del Intestino/genética , Leucopenia/genética , Masculino , Mercaptopurina/uso terapéutico , Ratones , Ratones Noqueados , Persona de Mediana Edad , República de Corea , Análisis de Secuencia de ADN , Adulto Joven
20.
Diabetologia ; 60(5): 778-783, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28013339

RESUMEN

At one level, obesity is clearly a problem of simple physics, a result of eating too much and not expending enough energy. The more complex question, however, is why do some people eat more than others? Studies of human and mouse genetics over the past two decades have uncovered a number of pathways within the brain that play a key role in the control of food intake. A prime example is the leptin-melanocortin pathway, which we now know greatly contributes to mammalian appetitive behaviour. However, genetic disruption of this pathway remains rare and does not represent the major burden of the disease that is carried by those of us with 'common obesity'. In recent years, genome-wide association studies have revealed more than 100 different candidate genes linked to BMI, with most (including many components of the melanocortin pathway) acting in the central nervous system and influencing food intake. So while severe disruption of the melanocortin pathway results in severe obesity, subtle variations in these genes influence where you might sit in the normal distribution of BMI. As we now enter this 'post-genomics' world, can this new information influence our treatment and management of obese patients?


Asunto(s)
Obesidad/etiología , Animales , Índice de Masa Corporal , Perros , Ingestión de Alimentos , Estudio de Asociación del Genoma Completo , Humanos , Hipotálamo/metabolismo , Melanocortinas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA