57 results on '"Elana Henning"'
Search Results
2. Neural correlates of fat preference in frontotemporal dementia: translating insights from the obesity literature
- Author
-
Rebekah M. Ahmed, Nga Yan Tse, Yu Chen, Elana Henning, John R. Hodges, Matthew C. Kiernan, Muireann Irish, I. Sadaf Farooqi, and Olivier Piguet
- Subjects
Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Objective Alterations in eating behaviour are one of the diagnostic features of behavioural variant frontotemporal dementia (bvFTD). It is hypothesised that underlying brain network disturbances and atrophy to key structures may affect macronutrient preference in bvFTD. We aimed to establish whether a preference for dietary fat exists in bvFTD, its association with cognitive symptoms and the underlying neural mechanisms driving these changes. Methods Using a test meal paradigm, adapted from the obesity literature, with variable fat content (low 20%, medium 40% and high 60%), preference for fat in 20 bvFTD was compared to 16 Alzheimer’s disease (AD) and 13 control participants. MRI brain scans were analysed to determine the neural correlates of fat preference. Results Behavioural variant FTD patients preferred the high‐fat meal compared to both AD (U = 61.5; p = 0.001) and controls (U = 41.5; p = 0.001), with 85% of bvFTD participants consistently rating the high‐fat content meal as their preferred option. This increased preference for the high‐fat meal was associated with total behavioural change (Cambridge Behavioural Inventory: rs = 0.462; p = 0.001), as well as overall functional decline (Frontotemporal Dementia Rating Scale: rs = −0.420; p = 0.03). A preference for high‐fat content in bvFTD was associated with atrophy in an extended brain network including frontopolar, anterior cingulate, insular cortices, putamen and amygdala extending into lateral temporal, posteromedial parietal and occipital cortices. Conclusions Increased preference for fat content is associated with many of the canonical features of bvFTD. These findings offer new insights into markers of disease progression and pathogenesis, providing potential treatment targets.
- Published
- 2021
- Full Text
- View/download PDF
3. Visualization of sympathetic neural innervation in human white adipose tissue
- Author
-
Aliki Perdikari, Tessa Cacciottolo, Elana Henning, Edson Mendes de Oliveira, Julia M. Keogh, and I. Sadaf Farooqi
- Subjects
obesity ,human adipose tissue ,sympathetic innervation ,whole tissue immunolabelling ,three-dimensional microscopy ,Biology (General) ,QH301-705.5 - Abstract
Obesity, defined as an excess of adipose tissue that adversely affects health, is a major cause of morbidity and mortality. However, to date, understanding the structure and function of human adipose tissue has been limited by the inability to visualize cellular components due to the innate structure of adipocytes, which are characterized by large lipid droplets. Combining the iDISCO and the CUBIC protocols for whole tissue staining and optical clearing, we developed a protocol to enable immunostaining and clearing of human subcutaneous white adipose tissue (WAT) obtained from individuals with severe obesity. We were able to perform immunolabelling of sympathetic nerve terminals in whole WAT and subsequent optical clearing by eliminating lipids to render the opaque tissue completely transparent. We then used light sheet confocal microscopy to visualize sympathetic innervation of human WAT from obese individuals in a three-dimensional manner. We demonstrate the visualization of sympathetic nerve terminals in human WAT. This protocol can be modified to visualize other structures such as blood vessels involved in the development, maintenance and function of human adipose tissue in health and disease.
- Published
- 2022
- Full Text
- View/download PDF
4. Neural networks associated with body composition in frontotemporal dementia
- Author
-
Rebekah M. Ahmed, Ramon Landin‐Romero, Cheng T. Liang, Julia M. Keogh, Elana Henning, Cherie Strikwerda‐Brown, Emma M. Devenney, John R. Hodges, Matthew C. Kiernan, I. Sadaf Farooqi, and Olivier Piguet
- Subjects
Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Background Frontotemporal dementia (FTD) is associated with complex changes in eating behavior and metabolism, which potentially affect disease pathogenesis and survival. It is currently not known if body composition changes and changes in fat deposition also exist in FTD, the relationship of these changes in eating behavior and appetite, and whether these changes are centrally mediated. Methods Body composition was measured in 28 people with behavioral‐variant frontotemporal dementia (bvFTD), 16 with Alzheimer’s disease (AD), and 19 healthy controls, using dual energy x‐ray absorptiometry. Changes in body composition were correlated to brain grey matter atrophy using voxel‐based morphometry on high‐resolution magnetic resonance imaging. Results Behavioral‐variant FTD was characterized by changes in body composition, with increased total fat mass, visceral adipose tissue area (VAT area), and android: gynoid ratio compared to control and AD participants (all P values
- Published
- 2019
- Full Text
- View/download PDF
5. Steroid receptor coactivator-1 modulates the function of Pomc neurons and energy homeostasis
- Author
-
Yongjie Yang, Agatha A. van der Klaauw, Liangru Zhu, Tessa M. Cacciottolo, Yanlin He, Lukas K. J. Stadler, Chunmei Wang, Pingwen Xu, Kenji Saito, Antentor Hinton, Xiaofeng Yan, Julia M. Keogh, Elana Henning, Matthew C. Banton, Audrey E. Hendricks, Elena G. Bochukova, Vanisha Mistry, Katherine L. Lawler, Lan Liao, Jianming Xu, Stephen O’Rahilly, Qingchun Tong, UK10K Consortium, Inês Barroso, Bert W. O’Malley, I. Sadaf Farooqi, and Yong Xu
- Subjects
Science - Abstract
Neurons expressing pro-opiomelanocortin (Pomc) regulate food intake and body weight. Here the authors show that Steroid Receptor Coactivator-1 (SRC-1) regulates the function of Pomc expressing neurons, and that rare heterozygous variants found in obese individuals lead to loss of SRC-1 function.
- Published
- 2019
- Full Text
- View/download PDF
6. Human MC4R variants affect endocytosis, trafficking and dimerization revealing multiple cellular mechanisms involved in weight regulation
- Author
-
Bas Brouwers, Edson Mendes de Oliveira, Maria Marti-Solano, Fabiola B.F. Monteiro, Suli-Anne Laurin, Julia M. Keogh, Elana Henning, Rebecca Bounds, Carole A. Daly, Shane Houston, Vikram Ayinampudi, Natalia Wasiluk, David Clarke, Bianca Plouffe, Michel Bouvier, M. Madan Babu, I. Sadaf Farooqi, and Jacek Mokrosiński
- Subjects
obesity ,MC4R ,therapy ,weight loss ,GPCRs ,β-arrestin ,Biology (General) ,QH301-705.5 - Abstract
Summary: The Melanocortin-4 Receptor (MC4R) plays a pivotal role in energy homeostasis. We used human MC4R mutations associated with an increased or decreased risk of obesity to dissect mechanisms that regulate MC4R function. Most obesity-associated mutations impair trafficking to the plasma membrane (PM), whereas obesity-protecting mutations either accelerate recycling to the PM or decrease internalization, resulting in enhanced signaling. MC4R mutations that do not affect canonical Gαs protein-mediated signaling, previously considered to be non-pathogenic, nonetheless disrupt agonist-induced internalization, β-arrestin recruitment, and/or coupling to Gαs, establishing their causal role in severe obesity. Structural mapping reveals ligand-accessible sites by which MC4R couples to effectors and residues involved in the homodimerization of MC4R, which is disrupted by multiple obesity-associated mutations. Human genetic studies reveal that endocytosis, intracellular trafficking, and homodimerization regulate MC4R function to a level that is physiologically relevant, supporting the development of chaperones, agonists, and allosteric modulators of MC4R for weight loss therapy.
- Published
- 2021
- Full Text
- View/download PDF
7. Disruption of the homeodomain transcription factor orthopedia homeobox (Otp) is associated with obesity and anxiety
- Author
-
Lee Moir, Elena G. Bochukova, Rebecca Dumbell, Gareth Banks, Rasneer S. Bains, Patrick M. Nolan, Cheryl Scudamore, Michelle Simon, Kimberly A. Watson, Julia Keogh, Elana Henning, Audrey Hendricks, Stephen O'Rahilly, Inês Barroso, Adrienne E. Sullivan, David C. Bersten, Murray L. Whitelaw, Susan Kirsch, Elizabeth Bentley, I. Sadaf Farooqi, and Roger D. Cox
- Subjects
OTP ,Obesity ,Energy balance ,Mouse model ,Human mutation ,Oxytocin ,Vasopressin ,Internal medicine ,RC31-1245 - Abstract
Objective: Genetic studies in obese rodents and humans can provide novel insights into the mechanisms involved in energy homeostasis. Methods: In this study, we genetically mapped the chromosomal region underlying the development of severe obesity in a mouse line identified as part of a dominant N-ethyl-N-nitrosourea (ENU) mutagenesis screen. We characterized the metabolic and behavioral phenotype of obese mutant mice and examined changes in hypothalamic gene expression. In humans, we examined genetic data from people with severe early onset obesity. Results: We identified an obese mouse heterozygous for a missense mutation (pR108W) in orthopedia homeobox (Otp), a homeodomain containing transcription factor required for the development of neuroendocrine cell lineages in the hypothalamus, a region of the brain important in the regulation of energy homeostasis. OtpR108W/+ mice exhibit increased food intake, weight gain, and anxiety when in novel environments or singly housed, phenotypes that may be partially explained by reduced hypothalamic expression of oxytocin and arginine vasopressin. R108W affects the highly conserved homeodomain, impairs DNA binding, and alters transcriptional activity in cells. We sequenced OTP in 2548 people with severe early-onset obesity and found a rare heterozygous loss of function variant in the homeodomain (Q153R) in a patient who also had features of attention deficit disorder. Conclusions: OTP is involved in mammalian energy homeostasis and behavior and appears to be necessary for the development of hypothalamic neural circuits. Further studies will be needed to investigate the contribution of rare variants in OTP to human energy homeostasis.
- Published
- 2017
- Full Text
- View/download PDF
8. Evaluation of a melanocortin-4 receptor (MC4R) agonist (Setmelanotide) in MC4R deficiency
- Author
-
Tinh-Hai Collet, Béatrice Dubern, Jacek Mokrosinski, Hillori Connors, Julia M. Keogh, Edson Mendes de Oliveira, Elana Henning, Christine Poitou-Bernert, Jean-Michel Oppert, Patrick Tounian, Florence Marchelli, Rohia Alili, Johanne Le Beyec, Dominique Pépin, Jean-Marc Lacorte, Andrew Gottesdiener, Rebecca Bounds, Shubh Sharma, Cathy Folster, Bart Henderson, Stephen O'Rahilly, Elizabeth Stoner, Keith Gottesdiener, Brandon L. Panaro, Roger D. Cone, Karine Clément, I. Sadaf Farooqi, and Lex H.T. Van der Ploeg
- Subjects
Obesity ,Melanocortin 4 receptor ,Setmelanotide ,Stratification ,Internal medicine ,RC31-1245 - Abstract
Objective: Pro-opiomelanocortin (POMC)-derived peptides act on neurons expressing the Melanocortin 4 receptor (MC4R) to reduce body weight. Setmelanotide is a highly potent MC4R agonist that leads to weight loss in diet-induced obese animals and in obese individuals with complete POMC deficiency. While POMC deficiency is very rare, 1–5% of severely obese individuals harbor heterozygous mutations in MC4R. We sought to assess the efficacy of Setmelanotide in human MC4R deficiency. Methods: We studied the effects of Setmelanotide on mutant MC4Rs in cells and the weight loss response to Setmelanotide administration in rodent studies and a human clinical trial. We annotated the functional status of 369 published MC4R variants. Results: In cells, we showed that Setmelanotide is significantly more potent at MC4R than the endogenous ligand alpha-melanocyte stimulating hormone and can disproportionally rescue signaling by a subset of severely impaired MC4R mutants. Wild-type rodents appear more sensitive to Setmelanotide when compared to MC4R heterozygous deficient mice, while MC4R knockout mice fail to respond. In a 28-day Phase 1b clinical trial, Setmelanotide led to weight loss in obese MC4R variant carriers. Patients with POMC defects upstream of MC4R show significantly more weight loss with Setmelanotide than MC4R deficient patients or obese controls. Conclusions: Setmelanotide led to weight loss in obese people with MC4R deficiency; however, further studies are justified to establish whether Setmelanotide can elicit clinically meaningful weight loss in a subset of the MC4R deficient obese population.
- Published
- 2017
- Full Text
- View/download PDF
9. Oxytocin administration suppresses hypothalamic activation in response to visual food cues
- Author
-
Agatha A. van der Klaauw, Hisham Ziauddeen, Julia M. Keogh, Elana Henning, Sekesai Dachi, Paul C. Fletcher, and I. Sadaf Farooqi
- Subjects
Medicine ,Science - Abstract
Abstract The aim of this study was to use functional neuroimaging to investigate whether oxytocin modulates the neural response to visual food cues in brain regions involved in the control of food intake. Twenty-four normal weight volunteers received intranasal oxytocin (24 IU) or placebo in a double-blind, randomized crossover study. Measurements were made forty-five minutes after dosing. On two occasions, functional MRI (fMRI) scans were performed in the fasted state; the blood oxygen level-dependent (BOLD) response to images of high-calorie foods versus low-calorie foods was measured. Given its critical role in eating behaviour, the primary region of interest was the hypothalamus. Secondary analyses examined the parabrachial nuclei and other brain regions involved in food intake and food reward. Intranasal oxytocin administration suppressed hypothalamic activation to images of high-calorie compared to low-calorie food (P = 0.0125). There was also a trend towards suppression of activation in the parabrachial nucleus (P = 0.0683). No effects of intranasal oxytocin were seen in reward circuits or on ad libitum food intake. Further characterization of the effects of oxytocin on neural circuits in the hypothalamus is needed to establish the utility of targeting oxytocin signalling in obesity.
- Published
- 2017
- Full Text
- View/download PDF
10. Rare Variant Analysis of Human and Rodent Obesity Genes in Individuals with Severe Childhood Obesity
- Author
-
Audrey E. Hendricks, Elena G. Bochukova, Gaëlle Marenne, Julia M. Keogh, Neli Atanassova, Rebecca Bounds, Eleanor Wheeler, Vanisha Mistry, Elana Henning, Antje Körner, Dawn Muddyman, Shane McCarthy, Anke Hinney, Johannes Hebebrand, Robert A. Scott, Claudia Langenberg, Nick J. Wareham, Praveen Surendran, Joanna M. Howson, Adam S. Butterworth, John Danesh, Børge G Nordestgaard, Sune F Nielsen, Shoaib Afzal, Sofia Papadia, Sofie Ashford, Sumedha Garg, Glenn L. Millhauser, Rafael I. Palomino, Alexandra Kwasniewska, Ioanna Tachmazidou, Stephen O’Rahilly, Eleftheria Zeggini, Inês Barroso, I. Sadaf Farooqi, Understanding Society Scientific Group, EPIC-CVD Consortium, and UK10K Consortium
- Subjects
Medicine ,Science - Abstract
Abstract Obesity is a genetically heterogeneous disorder. Using targeted and whole-exome sequencing, we studied 32 human and 87 rodent obesity genes in 2,548 severely obese children and 1,117 controls. We identified 52 variants contributing to obesity in 2% of cases including multiple novel variants in GNAS, which were sometimes found with accelerated growth rather than short stature as described previously. Nominally significant associations were found for rare functional variants in BBS1, BBS9, GNAS, MKKS, CLOCK and ANGPTL6. The p.S284X variant in ANGPTL6 drives the association signal (rs201622589, MAF~0.1%, odds ratio = 10.13, p-value = 0.042) and results in complete loss of secretion in cells. Further analysis including additional case-control studies and population controls (N = 260,642) did not support association of this variant with obesity (odds ratio = 2.34, p-value = 2.59 × 10−3), highlighting the challenges of testing rare variant associations and the need for very large sample sizes. Further validation in cohorts with severe obesity and engineering the variants in model organisms will be needed to explore whether human variants in ANGPTL6 and other genes that lead to obesity when deleted in mice, do contribute to obesity. Such studies may yield druggable targets for weight loss therapies.
- Published
- 2017
- Full Text
- View/download PDF
11. Genetic architecture of human thinness compared to severe obesity.
- Author
-
Fernando Riveros-McKay, Vanisha Mistry, Rebecca Bounds, Audrey Hendricks, Julia M Keogh, Hannah Thomas, Elana Henning, Laura J Corbin, Understanding Society Scientific Group, Stephen O'Rahilly, Eleftheria Zeggini, Eleanor Wheeler, Inês Barroso, and I Sadaf Farooqi
- Subjects
Genetics ,QH426-470 - Abstract
The variation in weight within a shared environment is largely attributable to genetic factors. Whilst many genes/loci confer susceptibility to obesity, little is known about the genetic architecture of healthy thinness. Here, we characterise the heritability of thinness which we found was comparable to that of severe obesity (h2 = 28.07 vs 32.33% respectively), although with incomplete genetic overlap (r = -0.49, 95% CI [-0.17, -0.82], p = 0.003). In a genome-wide association analysis of thinness (n = 1,471) vs severe obesity (n = 1,456), we identified 10 loci previously associated with obesity, and demonstrate enrichment for established BMI-associated loci (pbinomial = 3.05x10-5). Simulation analyses showed that different association results between the extremes were likely in agreement with additive effects across the BMI distribution, suggesting different effects on thinness and obesity could be due to their different degrees of extremeness. In further analyses, we detected a novel obesity and BMI-associated locus at PKHD1 (rs2784243, obese vs. thin p = 5.99x10-6, obese vs. controls p = 2.13x10-6 pBMI = 2.3x10-13), associations at loci recently discovered with much larger sample sizes (e.g. FAM150B and PRDM6-CEP120), and novel variants driving associations at previously established signals (e.g. rs205262 at the SNRPC/C6orf106 locus and rs112446794 at the PRDM6-CEP120 locus). Our ability to replicate loci found with much larger sample sizes demonstrates the value of clinical extremes and suggest that characterisation of the genetics of thinness may provide a more nuanced understanding of the genetic architecture of body weight regulation and may inform the identification of potential anti-obesity targets.
- Published
- 2019
- Full Text
- View/download PDF
12. Divergent effects of central melanocortin signalling on fat and sucrose preference in humans
- Author
-
Agatha A. van der Klaauw, Julia M. Keogh, Elana Henning, Cheryl Stephenson, Sarah Kelway, Victoria M. Trowse, Naresh Subramanian, Stephen O’Rahilly, Paul C. Fletcher, and I. Sadaf Farooqi
- Subjects
Science - Abstract
Hypothalamic melanocortin-4-receptors (MC4R) regulate food preference in rodents, but their role in humans is unclear. Here, the authors perform food preference and liking tests in humans with MC4R mutations and find that they prefer fatty food more, but sweet food less, than people without MC4R mutations.
- Published
- 2016
- Full Text
- View/download PDF
13. The impact of acute nutritional interventions on the plasma proteome
- Author
-
Spyros I Vernardis, Vadim Demichev, Oliver Lemke, Nana-Maria Grüning, Christoph Messner, Matt White, Maik Pietzner, Alina Peluso, Tinh-Hai Collet, Elana Henning, Christoph Gille, Archie Campbell, Caroline Hayward, David J Porteous, Riccardo E Marioni, Michael Mülleder, Aleksej Zelezniak, Nicholas J Wareham, Claudia Langenberg, I Sadaf Farooqi, and Markus Ralser
- Subjects
Chemical Biology & High Throughput ,Endocrinology ,Metabolism ,Ecology,Evolution & Ethology ,Endocrinology, Diabetes and Metabolism ,Biochemistry (medical) ,Clinical Biochemistry ,Synthetic Biology ,Biochemistry ,Computational & Systems Biology - Abstract
ContextHumans respond profoundly to changes in diet, while nutrition and environment have a great impact on population health. It is therefore important to deeply characterize the human nutritional responses.ObjectiveEndocrine parameters and the metabolome of human plasma are rapidly responding to acute nutritional interventions such as caloric restriction or a glucose challenge. It is less well understood whether the plasma proteome would be equally dynamic, and whether it could be a source of corresponding biomarkers.MethodsWe used high-throughput mass spectrometry to determine changes in the plasma proteome of i) 10 healthy, young, male individuals in response to 2 days of acute caloric restriction followed by refeeding; ii) 200 individuals of the Ely epidemiological study before and after a glucose tolerance test at 4 time points (0, 30, 60, 120 minutes); and iii) 200 random individuals from the Generation Scotland study. We compared the proteomic changes detected with metabolome data and endocrine parameters.ResultsBoth caloric restriction and the glucose challenge substantially impacted the plasma proteome. Proteins responded across individuals or in an individual-specific manner. We identified nutrient-responsive plasma proteins that correlate with changes in the metabolome, as well as with endocrine parameters. In particular, our study highlights the role of apolipoprotein C1 (APOC1), a small, understudied apolipoprotein that was affected by caloric restriction and dominated the response to glucose consumption and differed in abundance between individuals with and without type 2 diabetes.ConclusionOur study identifies APOC1 as a dominant nutritional responder in humans and highlights the interdependency of acute nutritional response proteins and the endocrine system.
- Published
- 2023
- Full Text
- View/download PDF
14. Obesity-Associated GNAS Mutations and the Melanocortin Pathway
- Author
-
Aliki Perdikari, Jacek Mokrosinski, Tabitha Randell, I. Sadaf Farooqi, Sharon Lim, Fleur Talbot, Rebecca Bounds, Melanie Kershaw, Deepthi Jyothish, Edson Mendes de Oliveira, Tim Cheetham, Antoinette McAulay, Vikram Ayinampudi, Elizabeth C Crowne, Inês Barroso, Peter T Clayton, Praveen Partha, Cristina Matei, Sanjay Gupta, Louise C Wilson, Elana Henning, Keogh Jm, Rachel Ahmed, and Natalia Wasiluk
- Subjects
musculoskeletal diseases ,030213 general clinical medicine ,medicine.medical_specialty ,medicine.disease_cause ,Short stature ,Thyrotropin receptor ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,GNAS complex locus ,Medicine ,Pseudohypoparathyroidism ,Exome sequencing ,030304 developmental biology ,0303 health sciences ,Mutation ,biology ,business.industry ,General Medicine ,medicine.disease ,3. Good health ,Melanocortin 4 receptor ,Endocrinology ,biology.protein ,medicine.symptom ,Melanocortin ,business - Abstract
Background GNAS encodes the Gαs (stimulatory G-protein alpha subunit) protein, which mediates G protein-coupled receptor (GPCR) signaling. GNAS mutations cause developmental delay, short stature, and skeletal abnormalities in a syndrome called Albright's hereditary osteodystrophy. Because of imprinting, mutations on the maternal allele also cause obesity and hormone resistance (pseudohypoparathyroidism). Methods We performed exome sequencing and targeted resequencing in 2548 children who presented with severe obesity, and we unexpectedly identified 22 GNAS mutation carriers. We investigated whether the effect of GNAS mutations on melanocortin 4 receptor (MC4R) signaling explains the obesity and whether the variable clinical spectrum in patients might be explained by the results of molecular assays. Results Almost all GNAS mutations impaired MC4R signaling. A total of 6 of 11 patients who were 12 to 18 years of age had reduced growth. In these patients, mutations disrupted growth hormone-releasing hormone receptor signaling, but growth was unaffected in carriers of mutations that did not affect this signaling pathway (mean standard-deviation score for height, -0.90 vs. 0.75, respectively; P = 0.02). Only 1 of 10 patients who reached final height before or during the study had short stature. GNAS mutations that impaired thyrotropin receptor signaling were associated with developmental delay and with higher thyrotropin levels (mean [±SD], 8.4±4.7 mIU per liter) than those in 340 severely obese children who did not have GNAS mutations (3.9±2.6 mIU per liter; P = 0.004). Conclusions Because pathogenic mutations may manifest with obesity alone, screening of children with severe obesity for GNAS deficiency may allow early diagnosis, improving clinical outcomes, and melanocortin agonists may aid in weight loss. GNAS mutations that are identified by means of unbiased genetic testing differentially affect GPCR signaling pathways that contribute to clinical heterogeneity. Monogenic diseases are clinically more variable than their classic descriptions suggest. (Funded by Wellcome and others.).
- Published
- 2021
- Full Text
- View/download PDF
15. Human loss-of-function variants in the serotonin 2C receptor associated with obesity and maladaptive behavior
- Author
-
Yang He, Bas Brouwers, Hesong Liu, Hailan Liu, Katherine Lawler, Edson Mendes de Oliveira, Dong-Kee Lee, Yongjie Yang, Aaron R. Cox, Julia M. Keogh, Elana Henning, Rebecca Bounds, Aliki Perdikari, Vikram Ayinampudi, Chunmei Wang, Meng Yu, Longlong Tu, Nan Zhang, Na Yin, Junying Han, Nikolas A. Scarcelli, Zili Yan, Kristine M. Conde, Camille Potts, Jonathan C. Bean, Mengjie Wang, Sean M. Hartig, Lan Liao, Jianming Xu, Inês Barroso, Jacek Mokrosinski, Yong Xu, I. Sadaf Farooqi, Brouwers, Bas [0000-0001-6541-4835], Mendes de Oliveira, Edson [0000-0002-7330-7826], Cox, Aaron R [0000-0002-3330-5746], Conde, Kristine M [0000-0002-9525-4606], Bean, Jonathan C [0000-0002-8007-2383], Hartig, Sean M [0000-0002-2695-2072], Xu, Jianming [0000-0002-8208-9162], Barroso, Inês [0000-0001-5800-4520], Xu, Yong [0000-0002-4908-1572], Sadaf Farooqi, I [0000-0001-7609-3504], Apollo - University of Cambridge Repository, Cox, Aaron R. [0000-0002-3330-5746], Conde, Kristine M. [0000-0002-9525-4606], Bean, Jonathan C. [0000-0002-8007-2383], Hartig, Sean M. [0000-0002-2695-2072], and Sadaf Farooqi, I. [0000-0001-7609-3504]
- Subjects
Male ,Serotonin ,article ,631/208/1515 ,General Medicine ,General Biochemistry, Genetics and Molecular Biology ,Obesity, Morbid ,631/378/340 ,Mice ,HEK293 Cells ,Adaptation, Psychological ,Receptor, Serotonin, 5-HT2C ,Animals ,Humans ,Female ,Obesity ,631/443/319 ,Child ,Serotonin 5-HT2 Receptor Agonists - Abstract
Funder: National Institute for Health Research (NIHR), Serotonin reuptake inhibitors and receptor agonists are used to treat obesity, anxiety and depression. Here we studied the role of the serotonin 2C receptor (5-HT2CR) in weight regulation and behavior. Using exome sequencing of 2,548 people with severe obesity and 1,117 control individuals without obesity, we identified 13 rare variants in the gene encoding 5-HT2CR (HTR2C) in 19 unrelated people (3 males and 16 females). Eleven variants caused a loss of function in HEK293 cells. All people who carried variants had hyperphagia and some degree of maladaptive behavior. Knock-in male mice harboring a human loss-of-function HTR2C variant developed obesity and reduced social exploratory behavior; female mice heterozygous for the same variant showed similar deficits with reduced severity. Using the 5-HT2CR agonist lorcaserin, we found that depolarization of appetite-suppressing proopiomelanocortin neurons was impaired in knock-in mice. In conclusion, we demonstrate that 5-HT2CR is involved in the regulation of human appetite, weight and behavior. Our findings suggest that melanocortin receptor agonists might be effective in treating severe obesity in individuals carrying HTR2C variants. We suggest that HTR2C should be included in diagnostic gene panels for severe childhood-onset obesity.
- Published
- 2022
16. Leptin-Mediated Changes in the Human Metabolome
- Author
-
I. Sadaf Farooqi, Katherine Lawler, Leonardo Bottolo, Takuhiro Sonoyama, Isabel Huang-Doran, Julia M. Keogh, Tinh-Hai Collet, Stephen O'Rahilly, and Elana Henning
- Subjects
Leptin ,Male ,0301 basic medicine ,obesity ,Endocrinology, Diabetes and Metabolism ,Clinical Biochemistry ,030204 cardiovascular system & hematology ,Severity of Illness Index ,Biochemistry ,0302 clinical medicine ,Endocrinology ,Loss of Function Mutation ,Tandem Mass Spectrometry ,Medicine ,Child ,Beta oxidation ,Clinical Research Article ,Leptin Deficiency ,digestive, oral, and skin physiology ,Recombinant Proteins ,Treatment Outcome ,Child, Preschool ,Metabolome ,Female ,hormones, hormone substitutes, and hormone antagonists ,AcademicSubjects/MED00250 ,medicine.medical_specialty ,Adolescent ,Hormone Replacement Therapy ,Lipolysis ,Context (language use) ,lipids ,03 medical and health sciences ,Internal medicine ,Humans ,Metabolomics ,bile acids ,business.industry ,Biochemistry (medical) ,Metabolism ,medicine.disease ,Obesity ,030104 developmental biology ,Energy Intake ,Energy Metabolism ,business ,Chromatography, Liquid - Abstract
Context While severe obesity due to congenital leptin deficiency is rare, studies in patients before and after treatment with leptin can provide unique insights into the role that leptin plays in metabolic and endocrine function. Objective The aim of this study was to characterize changes in peripheral metabolism in people with congenital leptin deficiency undergoing leptin replacement therapy, and to investigate the extent to which these changes are explained by reduced caloric intake. Design Ultrahigh performance liquid chromatography-tandem mass spectroscopy (UPLC-MS/MS) was used to measure 661 metabolites in 6 severely obese people with congenital leptin deficiency before, and within 1 month after, treatment with recombinant leptin. Data were analyzed using unsupervised and hypothesis-driven computational approaches and compared with data from a study of acute caloric restriction in healthy volunteers. Results Leptin replacement was associated with class-wide increased levels of fatty acids and acylcarnitines and decreased phospholipids, consistent with enhanced lipolysis and fatty acid oxidation. Primary and secondary bile acids increased after leptin treatment. Comparable changes were observed after acute caloric restriction. Branched-chain amino acids and steroid metabolites decreased after leptin, but not after acute caloric restriction. Individuals with severe obesity due to leptin deficiency and other genetic obesity syndromes shared a metabolomic signature associated with increased BMI. Conclusion Leptin replacement was associated with changes in lipolysis and substrate utilization that were consistent with negative energy balance. However, leptin’s effects on branched-chain amino acids and steroid metabolites were independent of reduced caloric intake and require further exploration.
- Published
- 2020
- Full Text
- View/download PDF
17. Visualization of sympathetic neural innervation in human white adipose tissue
- Author
-
I. Sadaf Farooqi, Aliki Perdikari, Julia M. Keogh, Elana Henning, Tessa M. Cacciottolo, Edson Mendes de Oliveira, Perdikari, Aliki [0000-0001-6841-4187], Mendes de Oliveira, Edson [0000-0002-7330-7826], Farooqi, I. Sadaf [0000-0001-7609-3504], Apollo - University of Cambridge Repository, Farooqi, I Sadaf [0000-0001-7609-3504], and Farooqi, Ismaa [0000-0001-7609-3504]
- Subjects
Pathology ,medicine.medical_specialty ,obesity ,Sympathetic Nervous System ,Adipose Tissue, White ,Immunology ,Adipose tissue ,Sympathetic nerve ,Methods and techniques ,White adipose tissue ,three-dimensional microscopy ,Computer Science::Digital Libraries ,General Biochemistry, Genetics and Molecular Biology ,law.invention ,Immunolabeling ,High Energy Physics::Theory ,Confocal microscopy ,law ,Mathematics::Quantum Algebra ,Adipocytes ,medicine ,sympathetic innervation ,Humans ,Astrophysics::Solar and Stellar Astrophysics ,whole tissue immunolabelling ,human adipose tissue ,Quantitative Biology::Neurons and Cognition ,business.industry ,General Neuroscience ,Adipose Tissue ,Large lipid droplets ,Tissue staining ,business ,Immunostaining - Abstract
Peer reviewed: True, Funder: Bernard Wolfe Health Neuroscience Endowment, Funder: Botnar Fondation, Funder: NIHR Cambridge Biomedical Research Center, Funder: NIHR Senior Investigator Award, Obesity, defined as an excess of adipose tissue that adversely affects health, is a major cause of morbidity and mortality. However, to date, understanding the structure and function of human adipose tissue has been limited by the inability to visualize cellular components due to the innate structure of adipocytes, which are characterized by large lipid droplets. Combining the iDISCO and the CUBIC protocols for whole tissue staining and optical clearing, we developed a protocol to enable immunostaining and clearing of human subcutaneous white adipose tissue (WAT) obtained from individuals with severe obesity. We were able to perform immunolabelling of sympathetic nerve terminals in whole WAT and subsequent optical clearing by eliminating lipids to render the opaque tissue completely transparent. We then used light sheet confocal microscopy to visualize sympathetic innervation of human WAT from obese individuals in a three-dimensional manner. We demonstrate the visualization of sympathetic nerve terminals in human WAT. This protocol can be modified to visualize other structures such as blood vessels involved in the development, maintenance and function of human adipose tissue in health and disease.
- Published
- 2022
- Full Text
- View/download PDF
18. Obesity due to Steroid Receptor Coactivator-1 deficiency is associated with endocrine and metabolic abnormalities
- Author
-
Tessa M Cacciottolo, Elana Henning, Julia M Keogh, Pierre Bel Lassen, Katherine Lawler, Rebecca Bounds, Rachel Ahmed, Aliki Perdikari, Edson Mendes de Oliveira, Miriam Smith, Edmund M Godfrey, Elspeth Johnson, Leanne Hodson, Karine Clément, Agatha A van der Klaauw, I Sadaf Farooqi, University of Cambridge [UK] (CAM), Nutrition et obésités: approches systémiques (UMR-S 1269) (Nutriomics), Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU), Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), University of Oxford, and Gestionnaire, HAL Sorbonne Université 5
- Subjects
Male ,[SDV.AEN] Life Sciences [q-bio]/Food and Nutrition ,Nuclear Receptor Coactivator 1 ,Endocrinology ,Endocrinology, Diabetes and Metabolism ,Biochemistry (medical) ,Clinical Biochemistry ,Humans ,Female ,Fibrosis ,Biochemistry ,[SDV.AEN]Life Sciences [q-bio]/Food and Nutrition ,Obesity, Morbid - Abstract
Context Genetic variants affecting the nuclear hormone receptor coactivator steroid receptor coactivator, SRC-1, have been identified in people with severe obesity and impair melanocortin signaling in cells and mice. As a result, obese patients with SRC-1 deficiency are being treated with a melanocortin 4 receptor agonist in clinical trials. Objective Here, our aim was to comprehensively describe and characterize the clinical phenotype of SRC-1 variant carriers to facilitate diagnosis and clinical management. Methods In genetic studies of 2462 people with severe obesity, we identified 23 rare heterozygous variants in SRC-1. We studied 29 adults and 18 children who were SRC-1 variant carriers and performed measurements of metabolic and endocrine function, liver imaging, and adipose tissue biopsies. Findings in adult SRC-1 variant carriers were compared to 30 age- and body mass index (BMI)-matched controls. Results The clinical spectrum of SRC-1 variant carriers included increased food intake in children, normal basal metabolic rate, multiple fractures with minimal trauma (40%), persistent diarrhea, partial thyroid hormone resistance, and menorrhagia. Compared to age-, sex-, and BMI-matched controls, adult SRC-1 variant carriers had more severe adipose tissue fibrosis (46.2% vs 7.1% respectively, P = .03) and a suggestion of increased liver fibrosis (5/13 cases vs 2/13 in controls, odds ratio = 3.4), although this was not statistically significant. Conclusion SRC-1 variant carriers exhibit hyperphagia in childhood, severe obesity, and clinical features of partial hormone resistance. The presence of adipose tissue fibrosis and hepatic fibrosis in young patients suggests that close monitoring for the early development of obesity-associated metabolic complications is warranted.
- Published
- 2022
- Full Text
- View/download PDF
19. Author response for 'Visualization of sympathetic neural innervation in human white adipose tissue'
- Author
-
null Aliki Perdikari, null Tessa Cacciottolo, null Elana Henning, null Edson Mendes de Oliveira, null Julia M. Keogh, and null I. Sadaf Farooqi
- Published
- 2022
- Full Text
- View/download PDF
20. Obesity due to melanocortin 4 receptor (MC4R) deficiency is associated with delayed gastric emptying
- Author
-
I. Sadaf Farooqi, Eleonora Seelig, Evelyn Shin, John R. Buscombe, Agatha A. van der Klaauw, Julia M. Keogh, Daniel Gillett, Elana Henning, Gillett, Daniel [0000-0002-9773-6502], van der Klaauw, Agatha A [0000-0001-6971-8828], Farooqi, I Sadaf [0000-0001-7609-3504], and Apollo - University of Cambridge Repository
- Subjects
medicine.medical_specialty ,obesity ,Gastroparesis ,Endocrinology, Diabetes and Metabolism ,Enteroendocrine cell ,MC4R ,Endocrinology ,gastric emptying ,Internal medicine ,medicine ,Humans ,Peptide YY ,Meal ,Gastric emptying ,PYY ,business.industry ,digestive, oral, and skin physiology ,Area under the curve ,medicine.disease ,Postprandial Period ,Obesity ,Melanocortin 4 receptor ,Postprandial ,Receptor, Melanocortin, Type 4 ,business ,hormones, hormone substitutes, and hormone antagonists ,Hormone - Abstract
Objective People who are severely obese due to melanocortin-4 receptor (MC4R) deficiency experience hyperphagia and impaired fullness after a meal (satiety). Meal-induced satiety is influenced by hormones, such as peptide-YY (PYY), which are released by enteroendocrine cells upon nutrient delivery to the small intestine. Design We investigated whether gastric emptying and PYY levels are altered in MC4R deficiency. Methods Gastric emptying was measured with a gastric scintigraphy protocol using technetium-99m (99 Tcm )-Tin Colloid for 3.5 h in individuals with loss of function MC4R variants and a control group of similar age and weight. In a separate study, we measured plasma PYY levels before and at multiple time points after three standardised meals given to individuals with MC4R deficiency and controls. Fasting PYY (basal secretion) and postprandial PYY levels were measured and the area under the curve and inter-meal peak were calculated. Results We found that gastric emptying time was significantly delayed and percentage meal retention increased in individuals with MC4R deficiency compared to obese controls. In addition, fasting and mean PYY secretion throughout the day were decreased in MC4R deficiency, whereas postprandial PYY secretion was unaltered. Conclusion Delayed gastric emptying and reduced basal PYY secretion may contribute to impaired satiety in people with obesity due to MC4R deficiency.
- Published
- 2021
- Full Text
- View/download PDF
21. Neural networks associated with body composition in frontotemporal dementia
- Author
-
Cheng T. Liang, Julia M. Keogh, Cherie Strikwerda-Brown, Matthew C. Kiernan, Emma Devenney, John R. Hodges, I. Sadaf Farooqi, Rebekah M. Ahmed, Ramon Landin-Romero, Elana Henning, and Olivier Piguet
- Subjects
0301 basic medicine ,Male ,medicine.medical_specialty ,media_common.quotation_subject ,Thalamus ,Neurosciences. Biological psychiatry. Neuropsychiatry ,Nucleus accumbens ,Amygdala ,03 medical and health sciences ,0302 clinical medicine ,Absorptiometry, Photon ,Internal medicine ,Medicine ,Humans ,Gray Matter ,skin and connective tissue diseases ,RC346-429 ,Research Articles ,media_common ,Aged ,medicine.diagnostic_test ,business.industry ,General Neuroscience ,Putamen ,Brain ,Magnetic resonance imaging ,Appetite ,Feeding Behavior ,Middle Aged ,medicine.disease ,Magnetic Resonance Imaging ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,Adipose Tissue ,Frontotemporal Dementia ,Body Composition ,Female ,Neurology (clinical) ,sense organs ,Neurology. Diseases of the nervous system ,Atrophy ,Nerve Net ,business ,Insula ,030217 neurology & neurosurgery ,Frontotemporal dementia ,Research Article ,RC321-571 - Abstract
Background Frontotemporal dementia (FTD) is associated with complex changes in eating behavior and metabolism, which potentially affect disease pathogenesis and survival. It is currently not known if body composition changes and changes in fat deposition also exist in FTD, the relationship of these changes in eating behavior and appetite, and whether these changes are centrally mediated. Methods Body composition was measured in 28 people with behavioral‐variant frontotemporal dementia (bvFTD), 16 with Alzheimer’s disease (AD), and 19 healthy controls, using dual energy x‐ray absorptiometry. Changes in body composition were correlated to brain grey matter atrophy using voxel‐based morphometry on high‐resolution magnetic resonance imaging. Results Behavioral‐variant FTD was characterized by changes in body composition, with increased total fat mass, visceral adipose tissue area (VAT area), and android: gynoid ratio compared to control and AD participants (all P values
- Published
- 2019
22. Neural correlates of fat preference in frontotemporal dementia: translating insights from the obesity literature
- Author
-
Yu Chen, Elana Henning, Muireann Irish, John R. Hodges, Rebekah M. Ahmed, Matthew C. Kiernan, Olivier Piguet, I. Sadaf Farooqi, Nga Yan Tse, Ahmed, Rebekah M [0000-0001-6996-8317], Chen, Yu [0000-0001-5863-7002], Kiernan, Matthew C [0000-0001-9054-026X], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,Male ,medicine.medical_specialty ,Neurosciences. Biological psychiatry. Neuropsychiatry ,Audiology ,Affect (psychology) ,03 medical and health sciences ,Food Preferences ,0302 clinical medicine ,Atrophy ,Rating scale ,Alzheimer Disease ,medicine ,Humans ,Obesity ,RC346-429 ,Research Articles ,Aged ,Cerebral Cortex ,Neural correlates of consciousness ,business.industry ,General Neuroscience ,Putamen ,Patient Acuity ,Middle Aged ,medicine.disease ,Amygdala ,Dietary Fats ,Magnetic Resonance Imaging ,Preference ,030104 developmental biology ,Frontotemporal Dementia ,Female ,Neurology (clinical) ,Neurology. Diseases of the nervous system ,Nerve Net ,business ,030217 neurology & neurosurgery ,Frontotemporal dementia ,RC321-571 ,Research Article - Abstract
Funder: Royal Australasian College of Physicians; Id: http://dx.doi.org/10.13039/501100001232, Funder: MND Research Institute of Australia; Id: http://dx.doi.org/10.13039/100008714, Funder: Australian Research Council Future Fellowship; Id: http://dx.doi.org/10.13039/501100000923, OBJECTIVE: Alterations in eating behaviour are one of the diagnostic features of behavioural variant frontotemporal dementia (bvFTD). It is hypothesised that underlying brain network disturbances and atrophy to key structures may affect macronutrient preference in bvFTD. We aimed to establish whether a preference for dietary fat exists in bvFTD, its association with cognitive symptoms and the underlying neural mechanisms driving these changes. METHODS: Using a test meal paradigm, adapted from the obesity literature, with variable fat content (low 20%, medium 40% and high 60%), preference for fat in 20 bvFTD was compared to 16 Alzheimer's disease (AD) and 13 control participants. MRI brain scans were analysed to determine the neural correlates of fat preference. RESULTS: Behavioural variant FTD patients preferred the high-fat meal compared to both AD (U = 61.5; p = 0.001) and controls (U = 41.5; p = 0.001), with 85% of bvFTD participants consistently rating the high-fat content meal as their preferred option. This increased preference for the high-fat meal was associated with total behavioural change (Cambridge Behavioural Inventory: rs = 0.462; p = 0.001), as well as overall functional decline (Frontotemporal Dementia Rating Scale: rs = -0.420; p = 0.03). A preference for high-fat content in bvFTD was associated with atrophy in an extended brain network including frontopolar, anterior cingulate, insular cortices, putamen and amygdala extending into lateral temporal, posteromedial parietal and occipital cortices. CONCLUSIONS: Increased preference for fat content is associated with many of the canonical features of bvFTD. These findings offer new insights into markers of disease progression and pathogenesis, providing potential treatment targets.
- Published
- 2021
23. Human MC4R variants affect endocytosis, trafficking and dimerization revealing multiple cellular mechanisms involved in weight regulation
- Author
-
M. Madan Babu, Elana Henning, Maria Marti-Solano, Bianca Plouffe, Carole A. Daly, Julia M. Keogh, Michel Bouvier, Bas Brouwers, Rebecca Bounds, Jacek Mokrosinski, Fabiola B.F. Monteiro, I. Sadaf Farooqi, Suli-Anne Laurin, Vikram Ayinampudi, David Clarke, Natalia Wasiluk, Edson Mendes de Oliveira, Shane Houston, Farooqi, Ismaa [0000-0001-7609-3504], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,obesity ,MC4R ,Energy homeostasis ,Gα(s) ,0302 clinical medicine ,Chlorocebus aethiops ,Cyclic AMP ,GTP-Binding Protein alpha Subunits, Gs ,Phosphorylation ,Internalization ,Extracellular Signal-Regulated MAP Kinases ,lcsh:QH301-705.5 ,beta-Arrestins ,media_common ,Chemistry ,Effector ,16. Peace & justice ,Endocytosis ,Cell biology ,COS Cells ,Receptor, Melanocortin, Type 4 ,Signal Transduction ,Gs alpha subunit ,media_common.quotation_subject ,Allosteric regulation ,Biology ,Affect (psychology) ,Models, Biological ,General Biochemistry, Genetics and Molecular Biology ,Article ,GPCRs ,03 medical and health sciences ,SDG 3 - Good Health and Well-being ,Animals ,Humans ,melanocortin ,MSH ,G protein-coupled receptor ,therapy ,β-arrestin ,Gαs ,Body Weight ,Cell Membrane ,Genetic Variation ,030104 developmental biology ,HEK293 Cells ,lcsh:Biology (General) ,Mutation ,Mutant Proteins ,weight loss ,Protein Multimerization ,030217 neurology & neurosurgery ,Function (biology) - Abstract
Summary The Melanocortin-4 Receptor (MC4R) plays a pivotal role in energy homeostasis. We used human MC4R mutations associated with an increased or decreased risk of obesity to dissect mechanisms that regulate MC4R function. Most obesity-associated mutations impair trafficking to the plasma membrane (PM), whereas obesity-protecting mutations either accelerate recycling to the PM or decrease internalization, resulting in enhanced signaling. MC4R mutations that do not affect canonical Gαs protein-mediated signaling, previously considered to be non-pathogenic, nonetheless disrupt agonist-induced internalization, β-arrestin recruitment, and/or coupling to Gαs, establishing their causal role in severe obesity. Structural mapping reveals ligand-accessible sites by which MC4R couples to effectors and residues involved in the homodimerization of MC4R, which is disrupted by multiple obesity-associated mutations. Human genetic studies reveal that endocytosis, intracellular trafficking, and homodimerization regulate MC4R function to a level that is physiologically relevant, supporting the development of chaperones, agonists, and allosteric modulators of MC4R for weight loss therapy., Graphical abstract, Highlights • Obesity-associated MC4R mutations that do not reduce cAMP disrupt other processes • MC4R mutations impact receptor homodimerization, endocytosis, and trafficking • Obesity-protecting mutations increase plasma membrane MC4Rs and enhance signaling • Multiple mechanisms regulate melanocortin tone to a physiologically relevant level, Using mutations in the human Melanocortin-4 Receptor (MC4R), Brouwers et al. identify receptor trafficking and endocytosis, coupling to Gαs/β-arrestins, and homodimerization as mechanisms involved in the regulation of body weight that may be targeted for weight loss therapy.
- Published
- 2021
- Full Text
- View/download PDF
24. Human BDNF/TrkB variants impair hippocampal synaptogenesis and associate with neurobehavioural abnormalities
- Author
-
Mingyan Zhu, Elana Henning, Beata K. Blaszczyk, Julia M. Keogh, Gary A. Wayman, Takuhiro Sonoyama, Peter Kirwan, Suzanne M. Appleyard, Magdalena Jura, Inês Barroso, I. Sadaf Farooqi, Bas Brouwers, Florian T. Merkle, Marko Hyvönen, David C. DeWitt, Lukas K. J. Stadler, Fuki M. Hisama, Apollo - University of Cambridge Repository, Stadler, Lukas [0000-0002-7028-4390], Barroso, Ines [0000-0001-5800-4520], Merkle, Florian [0000-0002-8513-2998], and Farooqi, Ismaa [0000-0001-7609-3504]
- Subjects
0301 basic medicine ,Male ,Neurite ,Adolescent ,Molecular biology ,Neurogenesis ,Neuronal Outgrowth ,Synaptogenesis ,lcsh:Medicine ,Tropomyosin receptor kinase B ,Hippocampal formation ,Biology ,Hippocampus ,03 medical and health sciences ,Glutamatergic ,631/208 ,0302 clinical medicine ,Endocrinology ,Neurotrophic factors ,Genetics ,Humans ,Receptor, trkB ,Phosphorylation ,692/163 ,Receptor ,lcsh:Science ,Child ,Loss function ,Neurons ,Multidisciplinary ,Brain-Derived Neurotrophic Factor ,musculoskeletal, neural, and ocular physiology ,692/617 ,lcsh:R ,article ,030104 developmental biology ,Neurology ,nervous system ,Child, Preschool ,lcsh:Q ,Female ,631/378 ,Neuroscience ,Protein Kinases ,030217 neurology & neurosurgery ,631/337 ,Signal Transduction - Abstract
Brain-derived neurotrophic factor (BDNF) signals through its high affinity receptor Tropomyosin receptor kinase-B (TrkB) to regulate neuronal development, synapse formation and plasticity. In rodents, genetic disruption of Bdnf and TrkB leads to weight gain and a spectrum of neurobehavioural phenotypes. Here, we functionally characterised a de novo missense variant in BDNF and seven rare variants in TrkB identified in a large cohort of people with severe, childhood-onset obesity. In cells, the E183K BDNF variant resulted in impaired processing and secretion of the mature peptide. Multiple variants in the kinase domain and one variant in the extracellular domain of TrkB led to a loss of function through multiple signalling pathways, impaired neurite outgrowth and dominantly inhibited glutamatergic synaptogenesis in hippocampal neurons. BDNF/TrkB variant carriers exhibited learning difficulties, impaired memory, hyperactivity, stereotyped and sometimes, maladaptive behaviours. In conclusion, human loss of function BDNF/TrkB variants that impair hippocampal synaptogenesis may contribute to a spectrum of neurobehavioural disorders.
- Published
- 2020
25. Exome Sequencing Identifies Genes and Gene Sets Contributing to Severe Childhood Obesity, Linking PHIP Variants to Repressed POMC Transcription
- Author
-
Rebecca Bounds, Inês Barroso, Eleftheria Zeggini, Felicity Payne, Nicholas J. Wareham, Caroline Hayward, Elana Henning, Stephen O'Rahilly, Gaëlle Marenne, Elena G. Bochukova, Audrey E. Hendricks, Sofie Ashford, Allan Daly, Julia M. Keogh, I. Sadaf Farooqi, Saad Pathan, Aliki Perdikari, Eleanor Wheeler, Vanisha Mistry, Christopher J. Lelliott, Claudia Langenberg, The Wellcome Trust Sanger Institute [Cambridge], Génétique, génomique fonctionnelle et biotechnologies (UMR 1078) (GGB), EFS-Université de Brest (UBO)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Institut Brestois Santé Agro Matière (IBSAM), Université de Brest (UBO), University of Colorado [Denver], Metabolic Research Laboratories [Cambridge, UK] (University of Cambridge), Wellcome Trust - MRC Cambridge-Addenbrooke’s Hospital [Cambridge, UK], MRC Human Genetics Unit, University of Edinburgh-Western General Hospital, Generation Scotland [Edinburgh, UK] (Centre for Genomic and Experimental Medicine), University of Edinburgh-Institute of Genetics and Molecular Medicine [Edinburgh, UK], MRC Epidemiology Unit [Cambridge, UK] (Wellcome Trust-MRC Institute of Metabolic Science), University of Cambridge [UK] (CAM)-Addenbrooke’s Hospital [Cambridge, UK]-Wellcome Trust-MRC Institute of Metabolic Science (IMS), MRC Metabolic Diseases Unit [Cambridge, UK] (Metabolic Research Laboratories), University of Cambridge [UK] (CAM), Helmholtz Zentrum München = German Research Center for Environmental Health, marenne, gaelle, Institut Brestois Santé Agro Matière (IBSAM), Université de Brest (UBO)-Université de Brest (UBO)-EFS-Institut National de la Santé et de la Recherche Médicale (INSERM), and Helmholtz-Zentrum München (HZM)
- Subjects
Male ,0301 basic medicine ,Pediatric Obesity ,Pro-Opiomelanocortin ,Physiology ,Endocrinology, Diabetes and Metabolism ,[SDV]Life Sciences [q-bio] ,Bioinformatics ,Mice ,0302 clinical medicine ,Endocrinology ,Missing heritability problem ,Chlorocebus aethiops ,Transcriptional regulation ,Medicine ,Exome ,genetics ,Child ,Cells, Cultured ,Exome sequencing ,Genetics ,Intracellular Signaling Peptides and Proteins ,Imidazoles ,POMC ,Middle Aged ,Penetrance ,Obesity, Morbid ,[SDV] Life Sciences [q-bio] ,Female ,Adult ,MEDLINE ,Biology ,Article ,Childhood obesity ,03 medical and health sciences ,gene set ,Animals ,Humans ,severe childhood obesity ,Molecular Biology ,Gene ,Genetic association ,function ,Genetic heterogeneity ,business.industry ,association ,Genetic Variation ,Cell Biology ,medicine.disease ,Mice, Inbred C57BL ,030104 developmental biology ,business ,030217 neurology & neurosurgery - Abstract
Summary Obesity is genetically heterogeneous with monogenic and complex polygenic forms. Using exome and targeted sequencing in 2,737 severely obese cases and 6,704 controls, we identified three genes (PHIP, DGKI, and ZMYM4) with an excess burden of very rare predicted deleterious variants in cases. In cells, we found that nuclear PHIP (pleckstrin homology domain interacting protein) directly enhances transcription of pro-opiomelanocortin (POMC), a neuropeptide that suppresses appetite. Obesity-associated PHIP variants repressed POMC transcription. Our demonstration that PHIP is involved in human energy homeostasis through transcriptional regulation of central melanocortin signaling has potential diagnostic and therapeutic implications for patients with obesity and developmental delay. Additionally, we found an excess burden of predicted deleterious variants involving genes nearest to loci from obesity genome-wide association studies. Genes and gene sets influencing obesity with variable penetrance provide compelling evidence for a continuum of causality in the genetic architecture of obesity, and explain some of its missing heritability., Graphical Abstract, Highlights • Three genes (PHIP, DGKI, and ZMYM4) are linked to severe childhood obesity • Wild-type PHIP enhances POMC transcription, but variants in PHIP repress POMC • Rare variants in BMI-associated loci from GWAS are enriched in severe obesity • Genetic architecture of severe childhood obesity reveals a continuum of causality, Childhood obesity can be caused by penetrant mutations in a number of genes controlling appetite and body weight. Marenne et al. identify three genes with mutations with variable penetrance in a continuum of causality in childhood obesity, and demonstrate that variants in PHIP repress POMC transcription.
- Published
- 2020
- Full Text
- View/download PDF
26. Disruption of the homeodomain transcription factor orthopedia homeobox (Otp) is associated with obesity and anxiety
- Author
-
Elena G. Bochukova, Rasneer Sonia Bains, Susan Kirsch, Julia M. Keogh, Patrick M. Nolan, Audrey E. Hendricks, Michelle Simon, Inês Barroso, I. Sadaf Farooqi, Gareth Banks, Cheryl L. Scudamore, Kimberly A. Watson, Rebecca Dumbell, Lee Moir, Roger D. Cox, Elana Henning, Adrienne E. Sullivan, Murray L. Whitelaw, David C. Bersten, Stephen O'Rahilly, and Elizabeth Bentley
- Subjects
Male ,0301 basic medicine ,lcsh:Internal medicine ,OTP ,Hypothalamus ,Gene Expression ,Mutagenesis (molecular biology technique) ,Nerve Tissue Proteins ,Energy balance ,Anxiety ,Bioinformatics ,Oxytocin ,Energy homeostasis ,Mouse model ,Mice ,03 medical and health sciences ,Databases, Genetic ,Animals ,Humans ,Medicine ,Missense mutation ,Amino Acid Sequence ,Obesity ,lcsh:RC31-1245 ,Molecular Biology ,Transcription factor ,Loss function ,Homeodomain Proteins ,2. Zero hunger ,Genetics ,Base Sequence ,business.industry ,Genes, Homeobox ,Brain ,Chromosome Mapping ,Gene Expression Regulation, Developmental ,Cell Biology ,Neurosecretory Systems ,Phenotype ,Human mutation ,030104 developmental biology ,Chromosomal region ,Homeobox ,Female ,Original Article ,Transcriptome ,business ,Vasopressin ,Transcription Factors - Abstract
Objective Genetic studies in obese rodents and humans can provide novel insights into the mechanisms involved in energy homeostasis. Methods In this study, we genetically mapped the chromosomal region underlying the development of severe obesity in a mouse line identified as part of a dominant N-ethyl-N-nitrosourea (ENU) mutagenesis screen. We characterized the metabolic and behavioral phenotype of obese mutant mice and examined changes in hypothalamic gene expression. In humans, we examined genetic data from people with severe early onset obesity. Results We identified an obese mouse heterozygous for a missense mutation (pR108W) in orthopedia homeobox (Otp), a homeodomain containing transcription factor required for the development of neuroendocrine cell lineages in the hypothalamus, a region of the brain important in the regulation of energy homeostasis. OtpR108W/+ mice exhibit increased food intake, weight gain, and anxiety when in novel environments or singly housed, phenotypes that may be partially explained by reduced hypothalamic expression of oxytocin and arginine vasopressin. R108W affects the highly conserved homeodomain, impairs DNA binding, and alters transcriptional activity in cells. We sequenced OTP in 2548 people with severe early-onset obesity and found a rare heterozygous loss of function variant in the homeodomain (Q153R) in a patient who also had features of attention deficit disorder. Conclusions OTP is involved in mammalian energy homeostasis and behavior and appears to be necessary for the development of hypothalamic neural circuits. Further studies will be needed to investigate the contribution of rare variants in OTP to human energy homeostasis., Highlights • A mouse Otp mutation alters hypothalamic neuropeptide expression leading to increased food intake, obesity and anxiety. • In severe early onset obesity, we found a heterozygous LOF variant in a patient with attention deficit disorder features. • These studies show for the first time that mutations in the Otp/OTP gene cause obesity.
- Published
- 2017
27. Steroid receptor coactivator-1 modulates the function of Pomc neurons and energy homeostasis
- Author
-
Saito K, Inês Barroso, Liao L, Elana Henning, Qingchun Tong, Jiake Xu, van der Klaauw Aa, Elena G. Bochukova, Bert W. O'Malley, Tessa M. Cacciottolo, Katherine Lawler, Matthew C. Banton, Yan X, Julia M. Keogh, Pingwen Xu, Chunmei Wang, Zhu L, Jr. Hinton A, Yanlin He, I. S. Farooqi, Stadler Lkj, Yang Y, Audrey E. Hendricks, Stephen O'Rahilly, Mistry, and Yong Xu
- Subjects
Leptin ,Male ,endocrine system ,Heterozygote ,Hypothalamus ,Mutation, Missense ,Mice, Transgenic ,Biology ,Article ,Energy homeostasis ,Membrane Potentials ,Mice ,Nuclear Receptor Coactivator 1 ,Cell Line, Tumor ,Animals ,Homeostasis ,Humans ,Gene Knock-In Techniques ,Obesity ,Alleles ,Crosses, Genetic ,Neurons ,digestive, oral, and skin physiology ,Body Weight ,Genetic Variation ,Cell biology ,Steroid Receptor Coactivator 1 ,HEK293 Cells ,Phenotype ,nervous system ,hormones, hormone substitutes, and hormone antagonists ,Gene Deletion ,Function (biology) - Abstract
Hypothalamic neurons expressing the anorectic peptide Pro-opiomelanocortin (Pomc) regulate food intake and body weight. Here, we show that Steroid Receptor Coactivator-1 (SRC-1) interacts with a target of leptin receptor activation, phosphorylated STAT3, to potentiate Pomc transcription. Deletion of SRC-1 in Pomc neurons in mice attenuates their depolarization by leptin, decreases Pomc expression and increases food intake leading to high-fat diet-induced obesity. In humans, fifteen rare heterozygous variants in SRC-1 found in severely obese individuals impair leptin-mediated Pomc reporter activity in cells, whilst four variants found in non-obese controls do not. In a knock-in mouse model of a loss of function human variant (SRC-1L1376P), leptin-induced depolarization of Pomc neurons and Pomc expression are significantly reduced, and food intake and body weight are increased. In summary, we demonstrate that SRC-1 modulates the function of hypothalamic Pomc neurons, and suggest that targeting SRC-1 may represent a useful therapeutic strategy for weight loss., Neurons expressing pro-opiomelanocortin (Pomc) regulate food intake and body weight. Here the authors show that Steroid Receptor Coactivator-1 (SRC-1) regulates the function of Pomc expressing neurons, and that rare heterozygous variants found in obese individuals lead to loss of SRC-1 function.
- Published
- 2019
- Full Text
- View/download PDF
28. Exome Sequencing Identifies Multiple Genes and Gene-Sets Contributing to Severe Childhood Obesity
- Author
-
Elena G. Bochukova, Allan Daly, Claudia Langenberg, Julia M. Keogh, Stephen O'Rahilly, Sofie Ashford, Caroline Hayward, Vanisha Mistry, Eleftheria Zeggini, Rebecca Bounds, Felicity Payne, Gaëlle Marenne, Christopher J. Lelliott, Audrey E. Hendricks, Saad Pathan, Eleanor Wheeler, I. Sadaf Farooqi, Nicholas J. Wareham, Elana Henning, Interval, Inês Barroso, and Aliki Perdikari
- Subjects
Pleckstrin homology domain ,Genetics ,Transcription (biology) ,Genetic heterogeneity ,In silico ,medicine ,Biology ,medicine.disease ,Gene ,Childhood obesity ,Exome sequencing ,Genetic association - Abstract
Obesity is genetically heterogeneous with monogenic and complex polygenic forms. Using exome sequencing followed by targeted sequencing in 2,737 severely obese cases and 6,704 controls, we identified three genes (PHIP, DGKI, ZMYM4) with an excess burden in cases of variants (MAF
- Published
- 2019
- Full Text
- View/download PDF
29. Genetic architecture of human thinness compared to severe obesity
- Author
-
Julia M. Keogh, Fernando Riveros-Mckay, Laura J Corbin, I. Sadaf Farooqi, Eleanor Wheeler, Rebecca Bounds, Stephen O'Rahilly, Hannah Thomas, Audrey E. Hendricks, Vanisha Mistry, Elana Henning, Inês Barroso, Eleftheria Zeggini, Wolke, Dieter, Farooqi, Ismaa [0000-0001-7609-3504], O'Rahilly, Stephen [0000-0003-2199-4449], Wheeler, Eleanor [0000-0002-8616-6444], Barroso, Ines [0000-0001-5800-4520], and Apollo - University of Cambridge Repository
- Subjects
Male ,obesity ,genome signal processing ,Cancer Research ,Physiology ,Muscle Proteins ,Genome-wide association study ,QH426-470 ,Mathematical and Statistical Techniques ,0302 clinical medicine ,Cell Signaling ,Medicine and Health Sciences ,Genetics (clinical) ,2. Zero hunger ,Genetics ,0303 health sciences ,Statistics ,Genomics ,Middle Aged ,Metaanalysis ,ALSPAC ,humanities ,Neoplasm Proteins ,Obesity, Morbid ,3. Good health ,Physiological Parameters ,Physical Sciences ,Female ,Genomic Signal Processing ,childhood obesity ,Research Article ,Signal Transduction ,Adult ,human genetics ,Receptors, Cell Surface ,body mass index ,Locus (genetics) ,Biology ,Research and Analysis Methods ,Polymorphism, Single Nucleotide ,Childhood obesity ,03 medical and health sciences ,Thinness ,medicine ,Humans ,Genetic Predisposition to Disease ,Statistical Methods ,Allele ,Molecular Biology ,Alleles ,Ecology, Evolution, Behavior and Systematics ,030304 developmental biology ,Genetic association ,Body Weight ,Biology and Life Sciences ,Computational Biology ,genetic loci ,Cell Biology ,Heritability ,Genome Analysis ,medicine.disease ,Obesity ,Genetic architecture ,meta-analysis ,genome-wide association studies ,Mathematics ,030217 neurology & neurosurgery ,Genome-Wide Association Study ,Transcription Factors ,RC - Abstract
The variation in weight within a shared environment is largely attributable to genetic factors. Whilst many genes/loci confer susceptibility to obesity, little is known about the genetic architecture of healthy thinness. Here, we characterise the heritability of thinness which we found was comparable to that of severe obesity (h2 = 28.07 vs 32.33% respectively), although with incomplete genetic overlap (r = -0.49, 95% CI [-0.17, -0.82], p = 0.003). In a genome-wide association analysis of thinness (n = 1,471) vs severe obesity (n = 1,456), we identified 10 loci previously associated with obesity, and demonstrate enrichment for established BMI-associated loci (pbinomial = 3.05x10-5). Simulation analyses showed that different association results between the extremes were likely in agreement with additive effects across the BMI distribution, suggesting different effects on thinness and obesity could be due to their different degrees of extremeness. In further analyses, we detected a novel obesity and BMI-associated locus at PKHD1 (rs2784243, obese vs. thin p = 5.99x10-6, obese vs. controls p = 2.13x10-6 pBMI = 2.3x10-13), associations at loci recently discovered with much larger sample sizes (e.g. FAM150B and PRDM6-CEP120), and novel variants driving associations at previously established signals (e.g. rs205262 at the SNRPC/C6orf106 locus and rs112446794 at the PRDM6-CEP120 locus). Our ability to replicate loci found with much larger sample sizes demonstrates the value of clinical extremes and suggest that characterisation of the genetics of thinness may provide a more nuanced understanding of the genetic architecture of body weight regulation and may inform the identification of potential anti-obesity targets., Author summary Obesity-associated disorders are amongst the leading causes of morbidity and mortality worldwide. Most genome-wide association studies (GWAS) have focused on body mass index (BMI = weight in Kg divided by height squared (m2)) and obesity, but to date no genetic association study testing thin and healthy individuals has been performed. In this study, we recruited a first of its kind cohort of 1,471 clinically ascertained thin and healthy individuals and contrasted the genetic architecture of the trait with that of severe early onset obesity. We show that thinness, like obesity, is a heritable trait with a polygenic component. In a GWAS of persistent healthy thinness vs. severe obesity with a total sample size of 2,927, we are able to find evidence of association in loci that have only been recently discovered using large cohorts with >40,000 individuals. We also find a novel BMI-associated locus at PKHD1 in UK Biobank highlighted by our association study. This work illustrates the value and increased power brought upon by using clinically ascertained extremes to study complex traits and provides a valuable resource on which to study resistance to obesity in an increasingly obesogenic environment.
- Published
- 2019
- Full Text
- View/download PDF
30. The Sleep/Wake Cycle is Directly Modulated by Changes in Energy Balance
- Author
-
Sekesai V Dachi, Síle Dunbar, Elana Henning, Suzanne L. Dickson, Agatha A. van der Klaauw, Sebastian M. Schmid, I. Sadaf Farooqi, Tinh-Hai Collet, Julia M. Keogh, Sarah Kelway, Diane Suddaby, van der Klaauw, Agatha [0000-0001-6971-8828], Henning, Elana [0000-0002-0399-4114], Keogh, Julia [0000-0002-0399-4114], Farooqi, Ismaa [0000-0001-7609-3504], and Apollo - University of Cambridge Repository
- Subjects
Adult ,Male ,0301 basic medicine ,Calorie restricted diet ,Gerontology ,Time Factors ,Polysomnography ,Library science ,leptin ,German ,03 medical and health sciences ,0302 clinical medicine ,Physiology (medical) ,Homeostasis ,Humans ,Medicine ,media_common.cataloged_instance ,Obesity ,sleep ,European union ,media_common ,business.industry ,European research ,Healthy Volunteers ,language.human_language ,3. Good health ,030104 developmental biology ,orexin ,Research centre ,language ,Sleep and Metabolism ,caloric restriction ,Sleep Stages ,Neurology (clinical) ,Energy Metabolism ,business ,030217 neurology & neurosurgery ,Sleep duration - Abstract
STUDY OBJECTIVES: The rise in obesity has been paralleled by a decline in sleep duration in epidemiological studies. However, the potential mechanisms linking energy balance and the sleep/wake cycle are not well understood. We aimed to examine the effects of manipulating energy balance on the sleep/wake cycle. METHODS: Twelve healthy normal weight men were housed in a clinical research facility and studied at three time points: baseline, after energy balance was disrupted by 2 days of caloric restriction to 10% of energy requirements, and after energy balance was restored by 2 days of ad libitum/free feeding. Sleep architecture, duration of sleep stages, and sleep-associated respiratory parameters were measured by polysomnography. RESULTS: Two days of caloric restriction significantly increased the duration of deep (stage 4) sleep (16.8% to 21.7% of total sleep time; P = 0.03); an effect which was entirely reversed upon free feeding (P = 0.01). Although the apnea-hypopnea index stayed within the reference range (< 5 events per hour), it decreased significantly from caloric restriction to free feeding (P = 0.03). Caloric restriction was associated with a marked fall in leptin (P < 0.001) and insulin levels (P = 0.002). The fall in orexin levels from baseline to caloric restriction correlated positively with duration of stage 4 sleep (Spearman rho = 0.83, P = 0.01) and negatively with the number of awakenings in caloric restriction (Spearman rho = -0.79, P = 0.01). CONCLUSIONS: We demonstrate that changes in energy homeostasis directly and reversibly impact on the sleep/wake cycle. These findings provide a mechanistic framework for investigating the association between sleep duration and obesity risk.
- Published
- 2016
- Full Text
- View/download PDF
31. The clinical and molecular spectrum associated with obesity-associated GNAS1 mutations
- Author
-
Fleur Talbot, Elana Henning, Keogh Jm, Mendes Edson de, and Sadaf Farooqi
- Subjects
biology ,business.industry ,GNAS complex locus ,biology.protein ,Medicine ,business ,Bioinformatics ,medicine.disease ,Spectrum (topology) ,Obesity - Published
- 2017
- Full Text
- View/download PDF
32. Leptin Mediates the Increase in Blood Pressure Associated with Obesity
- Author
-
Michael A. Cowley, Julio Licinio, Jack T. Pryor, Joel K. Elmquist, Stephen O'Rahilly, Scott M. Sternson, Russell D. Brown, Martin G. Myers, Jaspreet K. Bassi, Eric Ravussin, Pablo J. Enriori, Elana Henning, Frank L. Greenway, David Spanswick, Ralph J. DiLeone, Julia M. Keogh, Stephanie E. Simonds, Andrew M. Allen, I. Sadaf Farooqi, Kevin L. Grove, Keogh, Julia [0000-0002-0399-4114], Henning, Elana [0000-0002-0399-4114], O'Rahilly, Stephen [0000-0003-2199-4449], Farooqi, Ismaa [0000-0001-7609-3504], and Apollo - University of Cambridge Repository
- Subjects
Leptin ,medicine.medical_specialty ,endocrine system ,030204 cardiovascular system & hematology ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,0302 clinical medicine ,Central melanocortin system ,Internal medicine ,Brown adipose tissue ,medicine ,Animals ,Obesity ,Receptor ,Dorsomedial hypothalamic nucleus ,030304 developmental biology ,Neurons ,0303 health sciences ,Leptin receptor ,Biochemistry, Genetics and Molecular Biology(all) ,digestive, oral, and skin physiology ,medicine.disease ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Endocrinology ,Hypothalamus ,Mutation ,Hypertension ,Receptors, Leptin ,hormones, hormone substitutes, and hormone antagonists ,Signal Transduction ,RC - Abstract
Summary Obesity is associated with increased blood pressure (BP), which in turn increases the risk of cardiovascular diseases. We found that the increase in leptin levels seen in diet-induced obesity (DIO) drives an increase in BP in rodents, an effect that was not seen in animals deficient in leptin or leptin receptors (LepR). Furthermore, humans with loss-of-function mutations in leptin and the LepR have low BP despite severe obesity. Leptin’s effects on BP are mediated by neuronal circuits in the dorsomedial hypothalamus (DMH), as blocking leptin with a specific antibody, antagonist, or inhibition of the activity of LepR-expressing neurons in the DMH caused a rapid reduction of BP in DIO mice, independent of changes in weight. Re-expression of LepRs in the DMH of DIO LepR-deficient mice caused an increase in BP. These studies demonstrate that leptin couples changes in weight to changes in BP in mammalian species., Graphical Abstract, Highlights • Leptin is the link between obesity and increased blood pressure • Leptin acts through the dorsomedial hypothalamus to increase blood pressure • Blockade of leptin signaling reduces blood pressure in obese mice • Humans with defects in leptin signaling are protected from obesity hypertension, Leptin is found to be the link between obesity and increased blood pressure. Blocking leptin action reduces blood pressure in obese mice with clinical studies in humans, suggesting that defects in leptin signaling may protect against hypertension associated with obesity.
- Published
- 2014
- Full Text
- View/download PDF
33. KSR2 Mutations Are Associated with Obesity, Insulin Resistance, and Impaired Cellular Fuel Oxidation
- Author
-
Laura R, Pearce, Neli, Atanassova, Matthew C, Banton, Bill, Bottomley, Agatha A, van der Klaauw, Jean-Pierre, Revelli, Audrey, Hendricks, Julia M, Keogh, Elana, Henning, Deon, Doree, Sabrina, Jeter-Jones, Sumedha, Garg, Elena G, Bochukova, Rebecca, Bounds, Sofie, Ashford, Emma, Gayton, Peter C, Hindmarsh, Julian P H, Shield, Elizabeth, Crowne, David, Barford, Nick J, Wareham, Stephen, O'Rahilly, Michael P, Murphy, David R, Powell, Ines, Barroso, and I Sadaf, Farooqi
- Subjects
Male ,Models, Molecular ,Proto-Oncogene Proteins B-raf ,MAP Kinase Signaling System ,Biochemistry, Genetics and Molecular Biology(all) ,Fatty Acids ,Molecular Sequence Data ,Age Factors ,Hyperphagia ,Protein Serine-Threonine Kinases ,Protein Structure, Tertiary ,Mice ,Glucose ,Animals ,Humans ,Female ,Amino Acid Sequence ,Obesity ,Age of Onset ,Insulin Resistance ,Child ,Energy Metabolism ,Oxidation-Reduction ,Sequence Alignment - Abstract
SummaryKinase suppressor of Ras 2 (KSR2) is an intracellular scaffolding protein involved in multiple signaling pathways. Targeted deletion of Ksr2 leads to obesity in mice, suggesting a role in energy homeostasis. We explored the role of KSR2 in humans by sequencing 2,101 individuals with severe early-onset obesity and 1,536 controls. We identified multiple rare variants in KSR2 that disrupt signaling through the Raf-MEK-ERK pathway and impair cellular fatty acid oxidation and glucose oxidation in transfected cells; effects that can be ameliorated by the commonly prescribed antidiabetic drug, metformin. Mutation carriers exhibit hyperphagia in childhood, low heart rate, reduced basal metabolic rate and severe insulin resistance. These data establish KSR2 as an important regulator of energy intake, energy expenditure, and substrate utilization in humans. Modulation of KSR2-mediated effects may represent a novel therapeutic strategy for obesity and type 2 diabetes.PaperFlick
- Published
- 2013
- Full Text
- View/download PDF
34. Rare variants in single-minded 1 (SIM1) are associated with severe obesity
- Author
-
Shwetha, Ramachandrappa, Anne, Raimondo, Anna M G, Cali, Julia M, Keogh, Elana, Henning, Sadia, Saeed, Amanda, Thompson, Sumedha, Garg, Elena G, Bochukova, Soren, Brage, Victoria, Trowse, Eleanor, Wheeler, Adrienne E, Sullivan, Mehul, Dattani, Peter E, Clayton, Vipan, Datta, Vippan, Datta, John B, Bruning, Nick J, Wareham, Stephen, O'Rahilly, Daniel J, Peet, Ines, Barroso, Murray L, Whitelaw, and I Sadaf, Farooqi
- Subjects
Male ,Models, Molecular ,Transcriptional Activation ,Heterozygote ,medicine.medical_specialty ,Aryl hydrocarbon receptor nuclear translocator ,Adolescent ,DNA Mutational Analysis ,Mutation, Missense ,Gene Expression ,030209 endocrinology & metabolism ,Biology ,Energy homeostasis ,03 medical and health sciences ,0302 clinical medicine ,Genes, Reporter ,Internal medicine ,Basic Helix-Loop-Helix Transcription Factors ,medicine ,Humans ,Obesity ,Child ,Genetic Association Studies ,Luciferases, Renilla ,030304 developmental biology ,2. Zero hunger ,Genetics ,0303 health sciences ,Infant ,Heterozygote advantage ,General Medicine ,Penetrance ,Body Height ,Pedigree ,Repressor Proteins ,Melanocortin 4 receptor ,HEK293 Cells ,Endocrinology ,Case-Control Studies ,Child, Preschool ,SIM1 ,Receptor, Melanocortin, Type 4 ,Female ,Erratum ,Melanocortin ,Haploinsufficiency ,Research Article - Abstract
Single-minded 1 (SIM1) is a basic helix-loop-helix transcription factor involved in the development and function of the paraventricular nucleus of the hypothalamus. Obesity has been reported in Sim1 haploinsufficient mice and in a patient with a balanced translocation disrupting SIM1. We sequenced the coding region of SIM1 in 2,100 patients with severe, early onset obesity and in 1,680 controls. Thirteen different heterozygous variants in SIM1 were identified in 28 unrelated severely obese patients. Nine of the 13 variants significantly reduced the ability of SIM1 to activate a SIM1-responsive reporter gene when studied in stably transfected cells coexpressing the heterodimeric partners of SIM1 (ARNT or ARNT2). SIM1 variants with reduced activity cosegregated with obesity in extended family studies with variable penetrance. We studied the phenotype of patients carrying variants that exhibited reduced activity in vitro. Variant carriers exhibited increased ad libitum food intake at a test meal, normal basal metabolic rate, and evidence of autonomic dysfunction. Eleven of the 13 probands had evidence of a neurobehavioral phenotype. The phenotypic similarities between patients with SIM1 deficiency and melanocortin 4 receptor (MC4R) deficiency suggest that some of the effects of SIM1 deficiency on energy homeostasis are mediated by altered melanocortin signaling.
- Published
- 2013
- Full Text
- View/download PDF
35. High protein intake stimulates postprandial GLP1 and PYY release
- Author
-
Mohammad A. Ghatei, Elana Henning, Waljit S. Dhillo, Victoria M. Trowse, Julia M. Keogh, I. Sadaf Farooqi, and Agatha A. van der Klaauw
- Subjects
2. Zero hunger ,0303 health sciences ,Meal ,Nutrition and Dietetics ,Calorie ,030309 nutrition & dietetics ,Chemistry ,Endocrinology, Diabetes and Metabolism ,digestive, oral, and skin physiology ,Medicine (miscellaneous) ,030209 endocrinology & metabolism ,Glucagon-like peptide-1 ,Crossover study ,03 medical and health sciences ,0302 clinical medicine ,Endocrinology ,Animal science ,Postprandial ,Peptide YY ,Ghrelin ,Hormone - Abstract
Objective Meals high in protein induce greater intermeal satiety than meals high in fat and carbohydrates. We studied the gut hormone response and subsequent food intake after breakfasts high in protein, carbohydrate or high in fat controlled for volume, calories and appearance. Design and Methods Eight healthy volunteers participated in this randomized three-way crossover study. Study breakfasts were calculated to provide 20% of daily energy requirements and provided either 60% of energy from protein, fat or carbohydrate. Blood was drawn half-hourly for 4 h; energy intake at a subsequent ad libitum meal was measured. Results Total ghrelin decreased after food intake equally with the three breakfasts. PYY levels were highest after the high protein breakfast (P = 0.005). Indeed, PYY at 240 min was highest after the high protein breakfast compared to the high fat breakfast and to the high carbohydrate breakfast (P = 0.011 and P = 0.012, respectively). GLP-1 levels were highest after the high protein breakfast (P = 0.041) at 120 min and remained higher throughout the study. These differences in gut hormones did not translate into differences in food intake (1023 ± 390 kcal after high protein, 1016 ± 388 kcal after high fat and 1158 ± 433 kcal after high carbohydrate). Conclusion We conclude that a high protein meal increases circulating concentrations of the gut hormones PYY and GLP-1, but when meals are matched for volume, appearance and caloric value, these gut hormone changes do not translate into a reduction in ad libitum food intake.
- Published
- 2013
- Full Text
- View/download PDF
36. A deletion of the HBII-85 class of small nucleolar RNAs (snoRNAs) is associated with hyperphagia, obesity and hypogonadism
- Author
-
Julia M. Keogh, Richard J. Ellis, Mehul T. Dattani, Robin G. Walters, Susan E. Holder, I. Sadaf Farooqi, Stephen O'Rahilly, Mieke M. van Haelst, Giles S.H. Yeo, U. L. Fairbrother, Carolin Purmann, Philippe Froguel, Angela F. Brady, Elana Henning, Adam J. de Smith, Alexandra I. F. Blakemore, University of Groningen, Other departments, Human genetics, Amsterdam Neuroscience - Complex Trait Genetics, and Amsterdam Reproduction & Development (AR&D)
- Subjects
Male ,Molecular Sequence Data ,RECOMBINATION ,DEFINE ,Biology ,Hyperphagia ,Young Adult ,Hypogonadotropic hypogonadism ,Genetics ,medicine ,Humans ,RNA, Small Nucleolar ,Amino Acid Sequence ,Obesity ,BREAKPOINTS ,Small nucleolar RNA ,Molecular Biology ,Genotyping ,Gene ,Genetics (clinical) ,Chromosomes, Human, Pair 15 ,Hypogonadism ,Breakpoint ,Chromosome ,Chromosome Mapping ,CLUSTER ,General Medicine ,Articles ,medicine.disease ,Phenotype ,GENE ,DEFICIENCY ,PRADER-WILLI-SYNDROME ,Sequence Alignment ,Comparative genomic hybridization ,ALU ELEMENTS - Abstract
Genetic studies in patients with severe early-onset obesity have provided insights into the molecular and physiological pathways that regulate body weight in humans. We report a 19-year-old male with hyperphagia and severe obesity, mild learning difficulties and hypogonadism, in whom diagnostic tests for Prader-Willi syndrome (PWS) had been negative. We carried out detailed clinical and metabolic phenotyping of this patient and investigated the genetic basis of this obesity syndrome using Agilent 185 k array comparative genomic hybridization (aCGH) and Affymetrix 6.0 genotyping arrays. The identified deletion was validated using multiplex ligation-dependent probe amplification and long-range PCR, followed by breakpoint sequencing which enabled precise localization of the deletion. We identified a approximately 187 kb microdeletion at chromosome 15q11-13 that encompasses non-coding small nucleolar RNAs (including HBII-85 snoRNAs) which were not expressed in peripheral lymphocytes from the patient. Characterization of the clinical phenotype revealed increased ad libitum food intake, normal basal metabolic rate when adjusted for fat-free mass, partial hypogonadotropic hypogonadism and growth failure. We have identified a novel deletion on chromosome 15q11-13 in an individual with hyperphagia, obesity, hypogonadism and other features associated with PWS, which is normally caused by deficiency of several paternally expressed imprinted transcripts within chromosome 15q11-13, a region that includes multiple protein-coding genes as well as several non-coding snoRNAs. These findings provide direct evidence for the role of a particular family of non-coding RNAs, the HBII-85 snoRNA cluster, in human energy homeostasis, growth and reproduction.
- Published
- 2016
- Full Text
- View/download PDF
37. Prolonged successful therapy for hyperinsulinaemic hypoglycaemia after gastric bypass: the pathophysiological role of GLP1 and its response to a somatostatin analogue
- Author
-
K S Myint, Jerry R. Greenfield, Nick Finer, Elana Henning, I S Farooqi, and Jens J. Holst
- Subjects
Adult ,medicine.medical_specialty ,Time Factors ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Gastric Bypass ,Octreotide ,Incretin ,Lanreotide ,medicine.disease_cause ,chemistry.chemical_compound ,Endocrinology ,Glucagon-Like Peptide 1 ,Internal medicine ,medicine ,Humans ,business.industry ,Gastric bypass surgery ,Insulin ,General Medicine ,Nesidioblastosis ,Somatostatin ,chemistry ,Pancreatectomy ,Congenital Hyperinsulinism ,Female ,business ,medicine.drug ,Hormone - Abstract
Background: Spontaneous hyperinsulinaemic hypoglycaemia following gastric bypass surgery (GBS) is increasingly recognised. However, its pathophysiology remains unclear. Some patients require pancreatectomy. Medical therapy with calcium channel blockers, acarbose and diazoxide has been reported to be beneficial but has variable adherence and response. Method: We demonstrate the role of GLP1, counter-regulatory hormones and the subsequent response of GLP1 to somatostatin analogue therapy in a 42-year-old woman with persistent neuroglycopaenia 6 years after GBS. Plasma GLP1, insulin and glucose were measured for 5 h on three settings: i) a 75 g oral glucose tolerance test (OGTT); ii) a standard liquid test meal (LTM); and iii) an OGTT 30 min after a s.c. injection of 100 mg octreotide. Results: In comparison with obese non-diabetic controls, the patient had an elevated fasting and a markedly enhanced GLP1 response during the OGTT, followed by an exaggerated insulin response and a subsequent low glucose level. The GLP1 response to a LTM was similar but greater. Octreotide given prior to the OGTT attenuated both the GLP1 and insulin responses and abolished hypoglycaemia. Octreotide therapy significantly improved the patient’s neuroglycopaenic symptoms. The hormone profile was reassessed after 6 months following the LTM preceded by octreotide injection. Peak GLP1 and insulin responses were less pronounced than pretreatment responses and without hypoglycaemia. The patient was treated with lanreotide and had remained symptom-free and euglycaemic for 4 years. Conclusion: An exaggerated incretin response following altered gastrointestinal anatomy was the likely cause of hypoglycaemia in our GBS patient. Somatostatin successfully suppressed this response acutely and in the long term, thereby avoiding pancreatectomy and its sequelae.
- Published
- 2012
- Full Text
- View/download PDF
38. Resistance to thyroid hormone is associated with raised energy expenditure, muscle mitochondrial uncoupling, and hyperphagia
- Author
-
Nadia Schoenmakers, Julia M. Keogh, Penny J. D. Owen, I. Sadaf Farooqi, Sylvie Dufour, Philippa Raymond-Barker, Elana Henning, Catherine Mitchell, Kitt Falk Petersen, Krishna Chatterjee, Samantha Northcott, Peter R. Murgatroyd, John Lazarus, David B. Savage, Douglas L. Rothman, David Halsall, Gerald I. Shulman, Suzanne Curran, and Douglas E. Befroy
- Subjects
Adult ,Thyroid Hormone Resistance Syndrome ,endocrine system ,medicine.medical_specialty ,Citric Acid Cycle ,Hyperphagia ,Biology ,Thyroid hormone receptor beta ,Eating ,Adenosine Triphosphate ,Hyperthyroxinemia ,Internal medicine ,medicine ,Humans ,Resting energy expenditure ,Child ,Muscle, Skeletal ,Thyroid hormone receptor ,digestive, oral, and skin physiology ,Thyroid ,Skeletal muscle ,Thyroid Hormone Receptors beta ,General Medicine ,Middle Aged ,medicine.disease ,Mitochondria, Muscle ,Endocrinology ,medicine.anatomical_structure ,Hormone receptor ,Insulin Resistance ,Energy Metabolism ,Research Article ,Hormone - Abstract
Resistance to thyroid hormone (RTH), a dominantly inherited disorder usually associated with mutations in thyroid hormone receptor beta (THRB), is characterized by elevated levels of circulating thyroid hormones (including thyroxine), failure of feedback suppression of thyrotropin, and variable tissue refractoriness to thyroid hormone action. Raised energy expenditure and hyperphagia are recognized features of hyperthyroidism, but the effects of comparable hyperthyroxinemia in RTH patients are unknown. Here, we show that resting energy expenditure (REE) was substantially increased in adults and children with THRB mutations. Energy intake in RTH subjects was increased by 40%, with marked hyperphagia particularly evident in children. Rates of muscle TCA cycle flux were increased by 75% in adults with RTH, whereas rates of ATP synthesis were unchanged, as determined by 13C/31P magnetic resonance spectroscopy. Mitochondrial coupling index between ATP synthesis and mitochondrial rates of oxidation (as estimated by the ratio of ATP synthesis to TCA cycle flux) was significantly decreased in RTH patients. These data demonstrate that basal mitochondrial substrate oxidation is increased and energy production in the form of ATP synthesis is decreased in the muscle of RTH patients and that resting oxidative phosphorylation is uncoupled in this disorder. Furthermore, these observations suggest that mitochondrial uncoupling in skeletal muscle is a major contributor to increased REE in patients with RTH, due to tissue selective retention of thyroid hormone receptor alpha sensitivity to elevated thyroid hormone levels.
- Published
- 2010
- Full Text
- View/download PDF
39. Modulation of Blood Pressure by Central Melanocortinergic Pathways
- Author
-
Soren Brage, Elana Henning, John P. Mayer, Jerry R. Greenfield, Gregory S Cameron, Jeffrey W Miller, Teik Choon See, Stephen O'Rahilly, I. Sadaf Farooqi, Julia M. Keogh, David J. Lomas, Beatrice Astruc, and Julie Satterwhite
- Subjects
Adult ,Male ,Heterozygote ,medicine.medical_specialty ,Genotype ,Urinary system ,Blood Pressure ,Overweight ,Autonomic Nervous System ,Catecholamines ,Double-Blind Method ,Heart Rate ,Weight loss ,Internal medicine ,Prevalence ,medicine ,Humans ,Obesity ,Setmelanotide ,Cross-Over Studies ,Dose-Response Relationship, Drug ,business.industry ,General Medicine ,Melanocortin 4 receptor ,Blood pressure ,Endocrinology ,Hypertension ,Mutation ,Receptor, Melanocortin, Type 4 ,Female ,medicine.symptom ,Melanocortin ,Sleep ,business ,Weight gain ,Signal Transduction - Abstract
Weight gain and weight loss are associated with changes in blood pressure through unknown mechanisms. Central melanocortinergic signaling is implicated in the control of energy balance and blood pressure in rodents, but there is no information regarding such an association with blood pressure in humans.We assessed blood pressure, heart rate, and urinary catecholamines in overweight or obese subjects with a loss-of-function mutation in MC4R, the gene encoding the melanocortin 4 receptor, and in equally overweight control subjects. We also examined the effects of an MC4R agonist administered for 7 days in 28 overweight or obese volunteers.The prevalence of hypertension was markedly lower in the MC4R-deficient subjects than in the control subjects (24% vs. 53%, P=0.009). After the exclusion of subjects taking antihypertensive medications, blood-pressure levels were significantly lower in MC4R-deficient subjects than in control subjects, with mean (+/-SE) systolic blood pressures of 123+/-14 mm Hg and 131+/-12 mm Hg, respectively (P=0.02), and mean diastolic blood pressures of 73+/-10 mm Hg and 79+/-7 mm Hg, respectively (P=0.03). As compared with control subjects, MC4R-deficient subjects had a lower increase in heart rate on waking (P=0.007), a lower heart rate during euglycemic hyperinsulinemia (P0.001), and lower 24-hour urinary norepinephrine excretion (P=0.04). The maximum tolerated daily dose of 1.0 mg of the MC4R agonist led to significant increases of 9.3+/-1.9 mm Hg in systolic blood pressure and of 6.6+/-1.1 mm Hg in diastolic blood pressure (P0.001 for both comparisons) at 24 hours, as compared with placebo. Differences in blood pressure were not explained by changes in insulin levels; there were no significant adverse events.Results of our genetic and pharmacologic studies implicate melanocortinergic signaling in the control of human blood pressure through an insulin-independent mechanism.
- Published
- 2009
- Full Text
- View/download PDF
40. Acute energy balance alteration modifies sleep organization in healthy men
- Author
-
Sadaf Farooqi, Sebastian M. Schmid, Sekesai V Dachi, A. A. van der Klaauw, Tinh-Hai Collet, Elana Henning, M. Mölle, Síle Dunbar, Keogh Jm, Diane Suddaby, and Rodrigo Chamorro Melo
- Subjects
medicine.medical_specialty ,Physical medicine and rehabilitation ,business.industry ,medicine ,Energy balance ,General Medicine ,business ,Sleep in non-human animals - Published
- 2017
- Full Text
- View/download PDF
41. Dopamine Modulates the Neural Representation of Subjective Value of Food in Hungry Subjects
- Author
-
Wolfram Schultz, Martin D. Vestergaard, Paul C. Fletcher, Hisham Ziauddeen, I. Sadaf Farooqi, Elana Henning, Nenad Medic, Fletcher, Paul C [0000-0001-8257-1517], and Apollo - University of Cambridge Repository
- Subjects
Adult ,Male ,Agonist ,Hunger ,medicine.drug_class ,Dopamine ,Decision Making ,Intraparietal sulcus ,decision ,Young Adult ,03 medical and health sciences ,value ,0302 clinical medicine ,Double-Blind Method ,Reward ,Reaction Time ,medicine ,Humans ,Dopaminergic modulation ,030304 developmental biology ,0303 health sciences ,food ,General Neuroscience ,Dopaminergic ,Representation (systemics) ,Antagonist ,Brain ,Articles ,Magnetic Resonance Imaging ,Female ,Psychology ,Value (mathematics) ,Neuroscience ,Photic Stimulation ,Psychomotor Performance ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Although there is a rich literature on the role of dopamine in value learning, much less is known about its role in using established value estimations to shape decision-making. Here we investigated the effect of dopaminergic modulation on value-based decision-making for food items in fasted healthy human participants. The Becker-deGroot-Marschak auction, which assesses subjective value, was examined in conjunction with pharmacological fMRI using a dopaminergic agonist and an antagonist. We found that dopamine enhanced the neural response to value in the inferior parietal gyrus/intraparietal sulcus, and that this effect predominated toward the end of the valuation process when an action was needed to record the value. Our results suggest that dopamine is involved in acting upon the decision, providing additional insight to the mechanisms underlying impaired decision-making in healthy individuals and clinical populations with reduced dopamine levels.
- Published
- 2014
- Full Text
- View/download PDF
42. Obesity-associated melanocortin-4 receptor mutations are associated with changes in the brain response to food cues
- Author
-
Agatha A, van der Klaauw, Elisabeth A H, von dem Hagen, Julia M, Keogh, Elana, Henning, Stephen, O'Rahilly, Andrew D, Lawrence, Andrew J, Calder, and I Sadaf, Farooqi
- Subjects
Adult ,Male ,Adolescent ,JCEM Online: Advances in Genetics ,digestive, oral, and skin physiology ,Appetite ,Feeding Behavior ,Hyperphagia ,Middle Aged ,Weight Gain ,Magnetic Resonance Imaging ,Neostriatum ,Oxygen ,Young Adult ,Food ,Mutation ,Humans ,Receptor, Melanocortin, Type 4 ,Female ,Obesity ,Signal Transduction - Abstract
Context: Mutations in the melanocortin-4 receptor (MC4R) represent the commonest genetic form of obesity and are associated with hyperphagia. Objective: The aim of this study was to investigate whether melanocortin signaling modulates anticipatory food reward by studying the brain activation response to food cues in individuals with MC4R mutations. Design/Setting/Participants/Main Outcome Measure: We used functional magnetic resonance imaging to measure blood oxygen level-dependent responses to images of highly palatable, appetizing foods, bland foods, and non-food objects in eight obese individuals with MC4R mutations, 10 equally obese controls, and eight lean controls with normal MC4R genotypes. Based on previous evidence, we performed a region-of-interest analysis centered on the caudate/putamen (dorsal striatum) and ventral striatum. Results: Compared to non-foods, appetizing foods were associated with activation in the dorsal and ventral striatum in lean controls and in MC4R-deficient individuals. Surprisingly, we observed reduced activation of the dorsal and ventral striatum in obese controls relative to MC4R-deficient patients and lean controls. There were no group differences for the contrast of disgusting foods with bland foods or non-foods, suggesting that the effects observed in response to appetizing foods were not related to arousal. Conclusion: We identified differences in the striatal response to food cues between two groups of obese individuals, those with and those without MC4R mutations. These findings are consistent with a role for central melanocortinergic circuits in the neural response to visual food cues.
- Published
- 2014
43. Functional characterization of obesity-associated variants involving the α and β isoforms of human SH2B1
- Author
-
Keogh Jm, Joel M. Cline, Lawrence S. Argetsinger, Stephen O'Rahilly, Hsiao Wen Su, Sadia Saeed, Elana Henning, Sumedha Garg, Liangyou Rui, Ray Joe, Steven Shoelson, Laura R. Pearce, Michael E. Doche, I. Sadaf Farooqi, Inês Barroso, Elena G. Bochukova, and Christin Carter-Su
- Subjects
Gene isoform ,Adult ,Leptin ,Male ,medicine.medical_specialty ,Neurite ,Adolescent ,medicine.medical_treatment ,Mutation, Missense ,030209 endocrinology & metabolism ,Biology ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Endocrinology ,SH2B1 ,Internal medicine ,medicine ,Humans ,Insulin ,Protein Isoforms ,Obesity ,Child ,030304 developmental biology ,Adaptor Proteins, Signal Transducing ,Genetics ,0303 health sciences ,Brief Report ,Alternative splicing ,Signal transducing adaptor protein ,IRS2 ,3. Good health ,Alternative Splicing ,Female ,Signal transduction ,Signal Transduction - Abstract
We have previously reported rare variants in sarcoma (Src) homology 2 (SH2) B adaptor protein 1 (SH2B1) in individuals with obesity, insulin resistance, and maladaptive behavior. Here, we identify 4 additional SH2B1 variants by sequencing 500 individuals with severe early-onset obesity. SH2B1 has 4 alternatively spliced isoforms. One variant (T546A) lies within the N-terminal region common to all isoforms. As shown for past variants in this region, T546A impairs SH2B1β enhancement of nerve growth factor-induced neurite outgrowth, and the individual with the T546A variant exhibits mild developmental delay. The other 3 variants (A663V, V695M, and A723V) lie in the C-terminal tail of SH2B1α. SH2B1α variant carriers were hyperinsulinemic but did not exhibit the behavioral phenotype observed in individuals with SH2B1 variants that disrupt all isoforms. In in vitro assays, SH2B1α, like SH2B1β, enhances insulin- and leptin-induced insulin receptor substrate 2 (IRS2) phosphorylation and GH-induced cell motility. None of the variants affect SH2B1α enhancement of insulin- and leptin-induced IRS2 phosphorylation. However, T546A, A663V, and A723V all impair the ability of SH2B1α to enhance GH-induced cell motility. In contrast to SH2B1β, SH2B1α does not enhance nerve growth factor-induced neurite outgrowth. These studies suggest that genetic variants that disrupt isoforms other than SH2B1β may be functionally significant. Further studies are needed to understand the mechanism by which the individual isoforms regulate energy homeostasis and behavior.
- Published
- 2014
44. Postprandial total ghrelin suppression is modulated by melanocortin signaling in humans
- Author
-
Andrea M. Haqq, Julia M. Keogh, Jonathan Q. Purnell, Agatha A. van der Klaauw, Anthea Blackwood, I. Sadaf Farooqi, and Elana Henning
- Subjects
Adult ,Blood Glucose ,Male ,medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Clinical Biochemistry ,Biology ,Biochemistry ,Endocrinology ,Internal medicine ,medicine ,Humans ,Insulin ,Obesity ,Meal ,digestive, oral, and skin physiology ,Biochemistry (medical) ,Area under the curve ,Fasting ,JCEM Online: Brief Reports ,Postprandial Period ,Ghrelin ,Melanocortin 4 receptor ,Postprandial ,Mutation ,Receptor, Melanocortin, Type 4 ,Female ,Melanocortin ,hormones, hormone substitutes, and hormone antagonists ,Ghrelin secretion - Abstract
Ghrelin is implicated in meal initiation because circulating levels increase before and fall after meal consumption. In rodents, ghrelin stimulates food intake via hypothalamic circuits expressing the melanocortin 4 receptor (MC4R).The aim of the study was to investigate the impact of central melanocortinergic tone on ghrelin secretion in humans. DESIGN/SETTING/PATIENTS/MAIN OUTCOME MEASURE: We measured plasma total ghrelin before and after 3 standardized meals in patients with MC4R mutations and obese and lean controls. Fasting total ghrelin, area under the curve, and early (30-min) and intermeal postprandial total ghrelin suppression (the percentage difference between the premeal and the 30-min postprandial or intermeal nadir total ghrelin concentration) were calculated.Fasting total ghrelin concentrations and area under the curve for total ghrelin concentrations were significantly decreased in both obese groups compared to lean controls (fasting total ghrelin: lean controls, 1083 ± 203 pg/ml; and obese controls, 696 ± 81 pg/ml; MC4R: 609 ± 99 pg/ml, P.05). Early and intermeal postprandial total ghrelin suppression was attenuated in MC4R-deficient patients compared to lean controls (P.05); a similar pattern was observed for early postprandial suppression in comparison with obese controls, but this difference did not reach statistical significance (P = .09).These findings are consistent with a role for central melanocortinergic signaling in modulating meal-related total ghrelin suppression.
- Published
- 2013
45. Modulation of the sleep–wake cycle by changes in energy balance
- Author
-
Agatha A. van der Klaauw, Sekesai V Dachi, Sarah Kelway, I. Sadaf Farooqi, Suzanne L. Dickson, Elana Henning, Tinh-Hai Collet, Julia M. Keogh, Diane Suddaby, Sebastian M. Schmid, and Síle Dunbar
- Subjects
medicine.medical_specialty ,Calorie ,medicine.diagnostic_test ,business.industry ,Leptin ,Caloric theory ,General Medicine ,Polysomnography ,medicine.disease ,Obesity ,Orexin ,Endocrinology ,Internal medicine ,medicine ,media_common.cataloged_instance ,Circadian rhythm ,European union ,business ,media_common - Abstract
Background The rise in obesity has been paralleled by a decline in sleep duration according to results of epidemiological studies over the past 30 years. The potential mechanisms linking energy balance and the sleep–wake cycle are not well understood. We aimed to directly assess the effects of manipulating energy balance on the sleep–wake cycle. Methods The study included 12 healthy normal weight men. All assessments were done at baseline, after 2 days of caloric restriction to about 200 calories per day, and after 2 days of free feeding to restore energy balance. We measured energy balance with indirect calorimetry; the sleep–wake cycle (total sleep time [TST] and time spent in various stages of sleep [%TST]); the apnoea–hypopnoea index, a marker of hypoventilation, with polysomnography; and serum leptin, insulin, and orexin concentrations. Data are given as mean (SD). Findings The energy deficit induced by acute caloric restriction was fully compensated for after 2 days of free feeding when energy balance was restored. Caloric restriction significantly increased the duration of deep (stage 4) sleep (from 16·8 %TST [1·8] at baseline to 21·7 [1·8] during caloric restriction, p=0·03), which was entirely reversed to 16·1 (1·9) upon free feeding (p=0·01). The apnoea–hypopnoea index was increased from 1·5 (0·5) at baseline to 2·2 (0·7) during caloric restriction and returned to baseline values upon free feeding (1·1 [0·2], p=0·05). Caloric restriction was associated with a significant fall in leptin (from 3·35 ng/mL [2·06] to 0·70 [0·52], p=0·002) and insulin concentrations (from 45·6 pmol/L [19·7] to 16·1 [9·2], p=0·005); the change in orexin concentrations from baseline to caloric restriction correlated positively with duration of stage 4 sleep in caloric restriction (Pearson r=0·71, p=0·03). Changes in energy balance also led to changes in sympathetic tone and the pulsatile secretion of thyroid stimulating hormone and growth hormone. Interpretation We have demonstrated that changes in energy homoeostasis directly and reversibly impact on the sleep–wake cycle. These findings provide a mechanistic framework for investigating the association between sleep duration and obesity risk. Funding Wellcome Trust, National Institute for Health Research Cambridge Biomedical Research Centre, European Research Council, Bernard Wolfe Health Neuroscience Fund, Swiss National Science Foundation, European Society of Endocrinology, German Research Foundation, European Union Seventh Framework programme NeuroFAST consortium.
- Published
- 2016
- Full Text
- View/download PDF
46. Genome-wide SNP and CNV analysis identifies common and low-frequency variants associated with severe early-onset obesity
- Author
-
Elana Henning, Ruth J. F. Loos, Eleanor Wheeler, Nicholas J. Wareham, Hannah Blackburn, Sumedha Garg, Sarah J. Lindsay, I. Sadaf Farooqi, Ni Huang, Matthew E. Hurles, Stephen O'Rahilly, Inês Barroso, Elena G. Bochukova, and Julia M. Keogh
- Subjects
DNA Copy Number Variations ,Genotype ,Quantitative Trait Loci ,030209 endocrinology & metabolism ,Locus (genetics) ,Single-nucleotide polymorphism ,Genome-wide association study ,Quantitative trait locus ,Biology ,Polymorphism, Single Nucleotide ,Article ,Body Mass Index ,03 medical and health sciences ,0302 clinical medicine ,Genetics ,medicine ,SNP ,Humans ,Genetic Predisposition to Disease ,Obesity ,Child ,030304 developmental biology ,2. Zero hunger ,0303 health sciences ,Genome, Human ,medicine.disease ,Phenotype ,Case-Control Studies ,RMST ,Body mass index ,Genome-Wide Association Study - Abstract
Common and rare variants associated with body mass index (BMI) and obesity account for5% of the variance in BMI. We performed SNP and copy number variation (CNV) association analyses in 1,509 children with obesity at the extreme tail (3 s.d. from the mean) of the BMI distribution and 5,380 controls. Evaluation of 29 SNPs (P1 × 10(-5)) in an additional 971 severely obese children and 1,990 controls identified 4 new loci associated with severe obesity (LEPR, PRKCH, PACS1 and RMST). A previously reported 43-kb deletion at the NEGR1 locus was significantly associated with severe obesity (P = 6.6 × 10(-7)). However, this signal was entirely driven by a flanking 8-kb deletion; absence of this deletion increased risk for obesity (P = 6.1 × 10(-11)). We found a significant burden of rare, single CNVs in severely obese cases (P0.0001). Integrative gene network pathway analysis of rare deletions indicated enrichment of genes affecting G protein-coupled receptors (GPCRs) involved in the neuronal regulation of energy homeostasis.
- Published
- 2012
47. A mutation in the thyroid hormone receptor alpha gene
- Author
-
Elena, Bochukova, Nadia, Schoenmakers, Maura, Agostini, Erik, Schoenmakers, Odelia, Rajanayagam, Julia M, Keogh, Elana, Henning, Jana, Reinemund, Evelien, Gevers, Margarita, Sarri, Kate, Downes, Amaka, Offiah, Assunta, Albanese, David, Halsall, John W R, Schwabe, Murray, Bain, Keith, Lindley, Francesco, Muntoni, Faraneh, Vargha-Khadem, Faraneh Vargha, Khadem, Mehul, Dattani, I Sadaf, Farooqi, Mark, Gurnell, and Krishna, Chatterjee
- Subjects
Models, Molecular ,endocrine system ,medicine.medical_specialty ,Heterozygote ,Thyroid Hormones ,endocrine system diseases ,Growth-hormone-releasing hormone receptor ,Protein Conformation ,Biology ,Thyroid hormone receptor beta ,Hypothyroidism ,Internal medicine ,medicine ,Humans ,Child ,Growth Disorders ,Insulin-like growth factor 1 receptor ,Thyroid hormone receptor ,Thyroid ,General Medicine ,medicine.disease ,Thyroid hormone resistance ,Thyroxine ,medicine.anatomical_structure ,Endocrinology ,Thyroid hormone receptor alpha ,Codon, Nonsense ,Triiodothyronine ,Female ,Hormone ,Thyroid Hormone Receptors alpha - Abstract
Thyroid hormones exert their effects through alpha (TRα1) and beta (TRβ1 and TRβ2) receptors. Here we describe a child with classic features of hypothyroidism (growth retardation, developmental retardation, skeletal dysplasia, and severe constipation) but only borderline-abnormal thyroid hormone levels. Using whole-exome sequencing, we identified a de novo heterozygous nonsense mutation in a gene encoding thyroid hormone receptor alpha (THRA) and generating a mutant protein that inhibits wild-type receptor action in a dominant negative manner. Our observations are consistent with defective human TRα-mediated thyroid hormone resistance and substantiate the concept of hormone action through distinct receptor subtypes in different target tissues.
- Published
- 2011
48. High protein intake stimulates postprandial GLP1 and PYY release
- Author
-
Agatha A, van der Klaauw, Julia M, Keogh, Elana, Henning, Victoria M, Trowse, Waljit S, Dhillo, Mohammad A, Ghatei, and I Sadaf, Farooqi
- Subjects
Adult ,Male ,Cross-Over Studies ,Hunger ,digestive, oral, and skin physiology ,Middle Aged ,Satiation ,Postprandial Period ,Dietary Fats ,Ghrelin ,Healthy Volunteers ,Article ,Young Adult ,Glucagon-Like Peptide 1 ,Dietary Carbohydrates ,Humans ,Female ,Peptide YY ,Dietary Proteins ,Energy Intake ,Meals - Abstract
OBJECTIVE: Meals high in protein induce greater intermeal satiety than meals high in fat and carbohydrates. We studied the gut hormone response and subsequent food intake after breakfasts high in protein, carbohydrate or high in fat controlled for volume, calories and appearance. DESIGN AND METHODS: Eight healthy volunteers participated in this randomized three-way crossover study. Study breakfasts were calculated to provide 20% of daily energy requirements and provided either 60% of energy from protein, fat or carbohydrate. Blood was drawn half-hourly for 4 h; energy intake at a subsequent ad libitum meal was measured. RESULTS: Total ghrelin decreased after food intake equally with the three breakfasts. PYY levels were highest after the high protein breakfast (P = 0.005). Indeed, PYY at 240 min was highest after the high protein breakfast compared to the high fat breakfast and to the high carbohydrate breakfast (P = 0.011 and P = 0.012, respectively). GLP-1 levels were highest after the high protein breakfast (P = 0.041) at 120 min and remained higher throughout the study. These differences in gut hormones did not translate into differences in food intake (1023 ± 390 kcal after high protein, 1016 ± 388 kcal after high fat and 1158 ± 433 kcal after high carbohydrate). CONCLUSION: We conclude that a high protein meal increases circulating concentrations of the gut hormones PYY and GLP-1, but when meals are matched for volume, appearance and caloric value, these gut hormone changes do not translate into a reduction in ad libitum food intake.
- Published
- 2011
49. Missense Mutations in the Basic Helix-Loop-Helix Transcription Factor Single-Minded 1 Result in Severe, Early-Onset Obesity
- Author
-
Shwetha Ramachandrappa, Anne Raimondo, Julia Keogh, Elana Henning, Anna Cali, Soren Brage, Nick Wareham, Stephen O'Rahilly, Ines Barroso, Murray Whitelaw, and I Sadaf Farooqi
- Published
- 2011
- Full Text
- View/download PDF
50. Obesity due to Melanocortin 4 Receptor (MC4R) Deficiency Is Associated with Increased Linear Growth and Final Height, Fasting Hyperinsulinemia, and Incompletely Suppressed Growth Hormone Secretion
- Author
-
Stephen O'Rahilly, Cecilia Camacho-Hübner, Hanno Pijl, I. Sadaf Farooqi, Julia M. Keogh, Elana Henning, Ferdinand Roelfsema, Carlos Eduardo Martinelli, Agatha A. van der Klaauw, Jerry R. Greenfield, and Anthea Blackwood
- Subjects
INSULINA ,Male ,medicine.medical_specialty ,Fasting hyperinsulinemia ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Clinical Biochemistry ,Context (language use) ,Biochemistry ,Severity of Illness Index ,Statistics, Nonparametric ,Endocrinology ,Insulin-Like Growth Factor II ,Internal medicine ,Hyperinsulinism ,medicine ,Humans ,Insulin ,Obesity ,Insulin-Like Growth Factor I ,Glycoproteins ,Immunoassay ,Anthropometry ,business.industry ,Human Growth Hormone ,Biochemistry (medical) ,medicine.disease ,Growth hormone secretion ,Body Height ,Melanocortin 4 receptor ,Insulin-Like Growth Factor Binding Protein 1 ,Insulin-Like Growth Factor Binding Protein 3 ,Mutation ,Receptor, Melanocortin, Type 4 ,Female ,Melanocortin ,business ,Carrier Proteins - Abstract
Melanocortin receptor 4 (MC4R) deficiency is characterized by increased linear growth greater than expected for the degree of obesity.The objective of the investigation was to study the somatotroph axis in obese MC4R-deficient patients and equally obese controls.We obtained anthropometric measurements and insulin concentrations in 153 MC4R-deficient subjects and 1392 controls matched for age and severity of obesity. We measured fasting IGF-I, IGF-II, IGF binding protein (IGFBP)-1, IGFBP-3, and acid-labile subunit levels in a subset of 33 MC4R-deficient patients and 36 control subjects. We examined pulsatile GH secretion in six adult MC4R-deficient subjects and six obese controls.Height sd score was significantly greater in MC4R-deficient children under 5 yr of age compared with controls (mean ± SEM: 2.3 ± 0.06 vs. 1.8 ± 0.04, P0.001), an effect that persisted throughout childhood. Final height (cm) was greater in MC4R-deficient men (mean ± SEM 173 ± 2.5 vs. 168 ± 2.1, P0.001) and women (mean 165 ± 2.1 vs. 158 ± 1.9, P0.001). Fasting IGF-I, IGF-II, acid-labile subunit, and IGFBP-3 concentrations were similar in the two groups. GH levels were markedly suppressed in obese controls, but pulsatile GH secretion was retained in MC4R deficiency. The mean maximal GH secretion rate per burst (P0.05) and mass per burst (P0.05) were increased in MC4R deficiency, consistent with increased pulsatile and total GH secretion. Fasting insulin levels were markedly elevated in MC4R-deficient children.In MC4R deficiency, increased linear growth in childhood leads to increased adult final height, greater than predicted by obesity alone. GH pulsatility is maintained in MC4R deficiency, a finding consistent with animal studies, suggesting a role for MC4R in controlling hypothalamic somatostatinergic tone. Fasting insulin levels are significantly higher in children carrying MC4R mutations. Both of these factors may contribute to the accelerated growth phenotype characteristic of MC4R deficiency.
- Published
- 2011
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.