Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.774
Filtrar
1.
Physiol Rep ; 12(11): e16055, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38872474

RESUMO

This study examined the effects of exercise and detraining at a young age on fat accumulation in various organs. Four-week-old male Otsuka Long-Evans Tokushima Fatty (OLETF) rats were assigned to either the non-exercise sedentary (OLETF Sed) or exercise groups. The exercise group was subdivided into two groups: exercise between 4 and 12 weeks of age (OLETF Ex) and exercise between 4 and 6 weeks of age followed by non-exercise between 6 and 12 weeks of age (OLETF DT). Body weight was significantly lower in the OLETF Ex group than in the OLETF Sed group at 12 weeks of age. Fat accumulation in the epididymal white adipose tissue, liver, and brown adipose tissue was suppressed in the OLETF Ex group. During the exercise period, body weight and food intake in the OLETF DT group were significantly lower than those in the OLETF Sed group. However, food intake was significantly higher in the OLETF DT group than in the OLETF Sed group after exercise cessation, resulting in extreme obesity with fatty liver and brown adipose tissue whitening. Detraining after early-onset exercise promotes hyperphagia, causing extreme obesity. Overeating should be avoided during detraining periods in cases of exercise cessation at a young age.


Assuntos
Tecido Adiposo Marrom , Fígado Gorduroso , Hiperfagia , Obesidade , Condicionamento Físico Animal , Ratos Endogâmicos OLETF , Animais , Masculino , Tecido Adiposo Marrom/metabolismo , Hiperfagia/fisiopatologia , Hiperfagia/metabolismo , Ratos , Fígado Gorduroso/metabolismo , Fígado Gorduroso/etiologia , Obesidade/metabolismo , Obesidade/fisiopatologia , Obesidade/etiologia , Ingestão de Alimentos , Fígado/metabolismo , Peso Corporal
2.
Appetite ; 200: 107533, 2024 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-38825014

RESUMO

Research has drawn contradictory conclusions as to whether humans adjust meal size based on meal energy density (ED) or exhibit 'passive overconsumption'. Recent observational research has suggested that meal EDs greater than 1.7-2 kcal/g are compensated for through consumption of smaller meal sizes. We tested the relationship between ED and meal size by examining energy intake of meals at three levels of ED: low (∼1.0 kcal/g), medium (1.7-2.0 kcal/g) and high (>3.0 kcal/g). Two randomised, crossover experiments were conducted with adult participants. In experiment 1 (n = 34, 62% female, mean age 37.4 years), participants were served a lunch including a familiar low, medium or high ED dessert to eat ad libitum. In experiment 2 (n = 32, 66% female, mean age 36.4 years), participants were served a lunch meal manipulated to be low, medium or high ED to eat ad libitum. For experiment 2, later energy intake (post-meal energy intake) was also measured. In experiment 1, participants consumed a similar amount of energy from the low vs. medium ED food. The high ED food was associated with an increased intake of approximately 240 kcals compared to medium (p < 0.001, Cohen's d = 2.31) and low (p < 0.001, Cohen's d = 4.42) ED foods. In experiment 2, there were no significant differences in meal size (grams) between ED meals, resulting in a largely linear relationship between meal ED and energy intake across the three ED conditions ('passive overconsumption'). There were no differences in later energy intake between ED conditions. Contrary to recent suggestions, foods higher in ED were not associated with adjustments to meal size and were associated with increased energy intake across two experiments. Reformulation of foods high in ED may be an effective population level approach to reducing energy intake and obesity. Clinical trial registry number: NCT05744050; https://clinicaltrials.gov/ct2/show/NCT05744050.


Assuntos
Estudos Cross-Over , Ingestão de Energia , Refeições , Tamanho da Porção , Humanos , Feminino , Adulto , Masculino , Adulto Jovem , Almoço , Pessoa de Meia-Idade , Hiperfagia/psicologia , Comportamento Alimentar/psicologia , Período Pós-Prandial
3.
Obesity (Silver Spring) ; 32(7): 1373-1388, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38932722

RESUMO

OBJECTIVE: Obesity is characterized by dysregulated homeostatic mechanisms resulting in positive energy balance; however, when this dysregulation occurs is unknown. We assessed the time course of alterations to behaviors promoting weight gain in male and female mice switched to an obesogenic high-fat diet (HFD). METHODS: Male and female C57BL/6J mice were housed in metabolic chambers and were switched from chow to a 60% or 45% HFD for 4 and 3 weeks, respectively. Food intake, meal patterns, energy expenditure (EE), and body weight were continuously measured. A separate cohort of male mice was switched from chow to a 60% HFD and was given access to locked or unlocked running wheels. RESULTS: Switching mice to obesogenic diets promotes transient bouts of hyperphagia during the first 2 weeks followed by persistent caloric hyperphagia. EE increases but not sufficiently enough to offset increased caloric intake, resulting in a sustained net positive energy balance. Hyperphagia is associated with consumption of calorically larger meals (impaired satiation) more frequently (impaired satiety), particularly during the light cycle. Running wheel exercise delays weight gain in male mice fed a 60% HFD by enhancing satiation and increasing EE. However, exercise effects on satiation are no longer apparent after 2 weeks, coinciding with weight gain. CONCLUSIONS: Exposure to obesogenic diets engages homeostatic regulatory mechanisms for ~2 weeks that ultimately fail, and consequent weight gain is characterized by impaired satiation and satiety. Insights into the etiology of obesity can be obtained by investigating changes to satiation and satiety mechanisms during the initial ~2 weeks of HFD exposure.


Assuntos
Dieta Hiperlipídica , Ingestão de Energia , Metabolismo Energético , Comportamento Alimentar , Hiperfagia , Camundongos Endogâmicos C57BL , Obesidade , Aumento de Peso , Animais , Masculino , Camundongos , Dieta Hiperlipídica/efeitos adversos , Feminino , Obesidade/etiologia , Obesidade/metabolismo , Hiperfagia/etiologia , Comportamento Alimentar/fisiologia , Fatores de Tempo , Condicionamento Físico Animal , Saciação , Ingestão de Alimentos/fisiologia
4.
Mol Metab ; 86: 101975, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38925247

RESUMO

OBJECTIVE: The lactational period is associated with profound hyperphagia to accommodate the energy demands of nursing. These changes are important for the long-term metabolic health of the mother and children as altered feeding during lactation increases the risk of mothers and offspring developing metabolic disorders later in life. However, the specific behavioral mechanisms and neural circuitry mediating the hyperphagia of lactation are incompletely understood. METHODS: Here, we utilized home cage feeding devices to characterize the dynamics of feeding behavior in lactating mice. A combination of pharmacological and behavioral assays were utilized to determine how lactation alters meal structure, circadian aspects of feeding, hedonic feeding, and sensitivity to hunger and satiety signals in lactating mice. Finally, we utilized chemogenetic, immunohistochemical, and in vivo imaging approaches to characterize the role of hypothalamic agouti-related peptide (AgRP) neurons in lactational-hyperphagia. RESULTS: The lactational period is associated with increased meal size, altered circadian patterns of feeding, reduced sensitivity to gut-brain satiety signals, and enhanced sensitivity to negative energy balance. Hypothalamic AgRP neurons display increased sensitivity to negative energy balance and altered in vivo activity during the lactational state. Further, using in vivo imaging approaches we demonstrate that AgRP neurons are directly activated by lactation. Chemogenetic inhibition of AgRP neurons acutely reduces feeding in lactating mice, demonstrating an important role for these neurons in lactational-hyperphagia. CONCLUSIONS: Together, these results show that lactation collectively alters multiple components of feeding behavior and position AgRP neurons as an important cellular substrate mediating the hyperphagia of lactation.


Assuntos
Proteína Relacionada com Agouti , Comportamento Alimentar , Hiperfagia , Hipotálamo , Lactação , Neurônios , Animais , Proteína Relacionada com Agouti/metabolismo , Lactação/metabolismo , Hiperfagia/metabolismo , Feminino , Camundongos , Neurônios/metabolismo , Hipotálamo/metabolismo , Comportamento Alimentar/fisiologia , Metabolismo Energético , Camundongos Endogâmicos C57BL
5.
Cell Rep ; 43(6): 114343, 2024 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-38865247

RESUMO

Activation of prepronociceptin (PNOC)-expressing neurons in the arcuate nucleus (ARC) promotes high-fat-diet (HFD)-induced hyperphagia. In turn, PNOCARC neurons can inhibit the anorexic response of proopiomelanocortin (POMC) neurons. Here, we validate the necessity of PNOCARC activity for HFD-induced inhibition of POMC neurons in mice and find that PNOCARC-neuron-dependent inhibition of POMC neurons is mediated by gamma-aminobutyric acid (GABA) release. When monitoring individual PNOCARC neuron activity via Ca2+ imaging, we find a subpopulation of PNOCARC neurons that is inhibited upon gastrointestinal calorie sensing and disinhibited upon HFD feeding. Combining retrograde rabies tracing and circuit mapping, we find that PNOC neurons from the bed nucleus of the stria terminalis (PNOCBNST) provide inhibitory input to PNOCARC neurons, and this inhibitory input is blunted upon HFD feeding. This work sheds light on how an increase in caloric content of the diet can rewire a neuronal circuit, paving the way to overconsumption and obesity development.


Assuntos
Dieta Hiperlipídica , Hiperfagia , Núcleos Septais , Animais , Hiperfagia/metabolismo , Camundongos , Núcleos Septais/metabolismo , Neurônios/metabolismo , Masculino , Ácido gama-Aminobutírico/metabolismo , Pró-Opiomelanocortina/metabolismo , Neurônios GABAérgicos/metabolismo , Núcleo Arqueado do Hipotálamo/metabolismo , Camundongos Endogâmicos C57BL , Precursores de Proteínas , Receptores Opioides
6.
Endocrinology ; 165(7)2024 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-38815086

RESUMO

The serotonin 2C receptor (5-HT2CR)-melanocortin pathway plays well-established roles in the regulation of feeding behavior and body weight homeostasis. Dysfunctions in this system, such as loss-of-function mutations in the Htr2c gene, can lead to hyperphagia and obesity. In this study, we aimed to investigate the potential therapeutic strategies for ameliorating hyperphagia, hyperglycemia, and obesity associated with a loss-of-function mutation in the Htr2c gene (Htr2cF327L/Y). We demonstrated that reexpressing functional 5-HT2CR solely in hypothalamic pro-opiomelanocortin (POMC) neurons is sufficient to reduce food intake and body weight in Htr2cF327L/Y mice subjected to a high-fat diet (HFD). In addition, 5-HT2CR expression restores the responsiveness of POMC neurons to lorcaserin, a selective agonist for 5-HT2CR. Similarly, administration of melanotan II, an agonist of the melanocortin receptor 4 (MC4R), effectively suppresses feeding and weight gain in Htr2cF327L/Y mice. Strikingly, promoting wheel-running activity in Htr2cF327L/Y mice results in a decrease in HFD consumption and improved glucose homeostasis. Together, our findings underscore the crucial role of the melanocortin system in alleviating hyperphagia and obesity related to dysfunctions of the 5-HT2CR, and further suggest that MC4R agonists and lifestyle interventions might hold promise in counteracting hyperphagia, hyperglycemia, and obesity in individuals carrying rare variants of the Htr2c gene.


Assuntos
Dieta Hiperlipídica , Hiperfagia , Obesidade , Pró-Opiomelanocortina , Receptor Tipo 4 de Melanocortina , Receptor 5-HT2C de Serotonina , Animais , Receptor 5-HT2C de Serotonina/metabolismo , Receptor 5-HT2C de Serotonina/genética , Masculino , Camundongos , Hiperfagia/metabolismo , Hiperfagia/genética , Pró-Opiomelanocortina/metabolismo , Pró-Opiomelanocortina/genética , Obesidade/metabolismo , Obesidade/genética , Receptor Tipo 4 de Melanocortina/genética , Receptor Tipo 4 de Melanocortina/metabolismo , Receptor Tipo 4 de Melanocortina/agonistas , alfa-MSH/farmacologia , alfa-MSH/análogos & derivados , Mutação com Perda de Função , Hipotálamo/metabolismo , Peso Corporal/efeitos dos fármacos , Ingestão de Alimentos/efeitos dos fármacos , Ingestão de Alimentos/fisiologia , Ingestão de Alimentos/genética , Neurônios/metabolismo , Neurônios/efeitos dos fármacos , Modelos Animais de Doenças , Hiperglicemia/metabolismo , Hiperglicemia/genética , Camundongos Endogâmicos C57BL , Benzazepinas , Peptídeos Cíclicos
7.
Endocrinology ; 165(7)2024 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-38815068

RESUMO

The growth hormone secretagogue receptor (GHSR), primarily known as the receptor for the hunger hormone ghrelin, potently controls food intake, yet the specific Ghsr-expressing cells mediating the orexigenic effects of this receptor remain incompletely characterized. Since Ghsr is expressed in gamma-aminobutyric acid (GABA)-producing neurons, we sought to investigate whether the selective expression of Ghsr in a subset of GABA neurons is sufficient to mediate GHSR's effects on feeding. First, we crossed mice that express a tamoxifen-dependent Cre recombinase in the subset of GABA neurons that express glutamic acid decarboxylase 2 (Gad2) enzyme (Gad2-CreER mice) with reporter mice, and found that ghrelin mainly targets a subset of Gad2-expressing neurons located in the hypothalamic arcuate nucleus (ARH) and that is predominantly segregated from Agouti-related protein (AgRP)-expressing neurons. Analysis of various single-cell RNA-sequencing datasets further corroborated that the primary subset of cells coexpressing Gad2 and Ghsr in the mouse brain are non-AgRP ARH neurons. Next, we crossed Gad2-CreER mice with reactivable GHSR-deficient mice to generate mice expressing Ghsr only in Gad2-expressing neurons (Gad2-GHSR mice). We found that ghrelin treatment induced the expression of the marker of transcriptional activation c-Fos in the ARH of Gad2-GHSR mice, yet failed to induce food intake. In contrast, food deprivation-induced refeeding was higher in Gad2-GHSR mice than in GHSR-deficient mice and similar to wild-type mice, suggesting that ghrelin-independent roles of GHSR in a subset of GABA neurons is sufficient for eliciting full compensatory hyperphagia in mice.


Assuntos
Núcleo Arqueado do Hipotálamo , Privação de Alimentos , Neurônios GABAérgicos , Grelina , Glutamato Descarboxilase , Hiperfagia , Receptores de Grelina , Animais , Masculino , Camundongos , Neurônios GABAérgicos/metabolismo , Receptores de Grelina/genética , Receptores de Grelina/metabolismo , Hiperfagia/metabolismo , Grelina/metabolismo , Grelina/farmacologia , Núcleo Arqueado do Hipotálamo/metabolismo , Privação de Alimentos/fisiologia , Glutamato Descarboxilase/metabolismo , Glutamato Descarboxilase/genética , Camundongos Transgênicos , Proteína Relacionada com Agouti/metabolismo , Proteína Relacionada com Agouti/genética , Camundongos Endogâmicos C57BL
8.
Nat Metab ; 6(6): 1076-1091, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38777856

RESUMO

Nutrient handling is an essential function of the gastrointestinal tract. Hormonal responses of small intestinal enteroendocrine cells (EECs) have been extensively studied but much less is known about the role of colonic EECs in metabolic regulation. To address this core question, we investigated a mouse model deficient in colonic EECs. Here we show that colonic EEC deficiency leads to hyperphagia and obesity. Furthermore, colonic EEC deficiency results in altered microbiota composition and metabolism, which we found through antibiotic treatment, germ-free rederivation and transfer to germ-free recipients, to be both necessary and sufficient for the development of obesity. Moreover, studying stool and blood metabolomes, we show that differential glutamate production by intestinal microbiota corresponds to increased appetite and that colonic glutamate administration can directly increase food intake. These observations shed light on an unanticipated host-microbiota axis in the colon, part of a larger gut-brain axis, that regulates host metabolism and body weight.


Assuntos
Colo , Células Enteroendócrinas , Microbioma Gastrointestinal , Obesidade , Animais , Células Enteroendócrinas/metabolismo , Camundongos , Colo/microbiologia , Colo/metabolismo , Obesidade/metabolismo , Obesidade/microbiologia , Camundongos Endogâmicos C57BL , Ácido Glutâmico/metabolismo , Eixo Encéfalo-Intestino , Hiperfagia/metabolismo
9.
Obes Res Clin Pract ; 18(3): 209-215, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38705820

RESUMO

BACKGROUND: Eating-related problems (e.g., binge eating (BE)) and impaired quality of life (QoL) is more prevalent in children with overweight and obesity. This study aimed to investigate changes in self-reported overeating (OE), BE, and QoL in children with overweight or obesity attending multicomponent 10-week lifestyle camps with a 52-weeks follow-up. Additionally, the study sought to investigate whether self-reported OE/BE before camp was associated with changes in QoL. METHODS: Children aged 7 to 14-years could attend camp if they had overweight/obesity, were lonely, unhappy, or had social or family-related problems. In this study only children with overweight and obesity were included (n:185). OE, BE, and QoL were measured using self-reported questionnaires. RESULTS: In total, 38 % of the children reported regular BE at baseline. Regular OE, occasional BE, and occasional OE was reported by 14 %, 13 %, and 11 %, respectively, while 24 % reported no eating-related problems. The relative risk of experiencing eating-related problems decreased at 10-weeks compared to baseline. Additionally, the probability of regular OE (RR 0.12 (95 % CI 0.04;0.38) (X2 = 8.44, p = 0.004)) and regular BE (RR 0.01 (95 % CI 0.00;0.11) (X2 = 9.91, p = 0.002)) remained lower at 52-weeks relative to baseline. All QoL dimensions improved after camp, and the presence of self-reported OE and regular BE at baseline was significantly associated with lower QoL at baseline, 10 and 52-weeks. CONCLUSION: Children self-reporting OE and BE may be a particular vulnerable group that needs more support from camp staff and healthcare professionals to improve QoL. CLINICAL TRIAL REGISTRATION: clinicaltrials.gov with ID: NCT04522921.


Assuntos
Obesidade Infantil , Qualidade de Vida , Adolescente , Criança , Feminino , Humanos , Masculino , Bulimia/psicologia , Acampamento , Comportamento Alimentar/psicologia , Hiperfagia/psicologia , Estilo de Vida , Sobrepeso/psicologia , Obesidade Infantil/psicologia , Autorrelato , Inquéritos e Questionários
10.
J Psychiatr Pract ; 30(3): 242-244, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38819249

RESUMO

Catatonia is a complex syndrome with unique cognitive, psychomotor, and mood features. Mannerisms and stereotypies are catatonic signs that have been extensively observed and described in the literature, mostly in the context of movements or motor acts. Stereotypies are commonly described as repetitive psychomotor or verbal acts with the abnormality not inherent in the act but in its frequency. Mannerisms, like stereotypies, are repetitive psychomotor or verbal acts, but they are fundamentally odd in nature. Recently, several reports have described these phenomena in the context of complex behaviors, such as eating and drinking. Identification and appreciation of personal and cultural norms, in addition to a careful analysis of behavioral processes and actions, are important tools for clinicians to identify these potentially elusive and often missed patterns of behavior in patients with catatonia. We present the case of a 30-year-old male with a psychiatric history of treatment-resistant, recurrent major depressive disorder with psychotic features who presented to the inpatient psychiatric unit with signs of catatonia, including repeated, purposeless eating. The patient's chart was reviewed, and a literature review was conducted using PubMed with the keywords catatonia, stereotypies, mannerisms, and hyperphagia. The patient, who was diagnosed with catatonia and expressed hyperphagia as a stereotypy, responded to lorazepam. This case shows that hyperphagia may present as a stereotypy in patients with catatonia.


Assuntos
Catatonia , Hiperfagia , Humanos , Catatonia/etiologia , Catatonia/tratamento farmacológico , Masculino , Hiperfagia/psicologia , Hiperfagia/etiologia , Adulto , Comportamento Estereotipado , Transtorno Depressivo Maior , Lorazepam/uso terapêutico , Lorazepam/administração & dosagem
11.
Genes (Basel) ; 15(4)2024 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-38674329

RESUMO

Childhood obesity is a significant public health concern, particularly among Hispanic populations. This study aimed to elucidate the genetic predisposition to obesity in Puerto Rican children of Hispanic descent, addressing a notable gap in existing research. A cohort of 103 children with obesity and hyperphagia underwent genetic screening for rare obesity-related variants. Clinical assessments and family history evaluations were conducted to characterize the demographic and clinical characteristics of the cohort. Genetic testing revealed a high prevalence of variants, with 73% of subjects having at least one reported variant. Pathogenic variants, predominantly associated with obesity-related ciliopathies, were identified in 7% of cases. Additionally, 90% of cases had variants of uncertain significance, highlighting the complexity of genetic contributions to obesity. This study emphasizes the critical need for further investigation into the genetic foundations of obesity, particularly within Hispanic communities. The findings emphasize the importance of early medical evaluation, vigilant monitoring for hyperphagia onset, and targeted interventions tailored to the unique genetic landscape of Puerto Rican children. This research provides a foundational framework for future studies to mitigate the impact of genetic obesity within this population.


Assuntos
Predisposição Genética para Doença , Hispânico ou Latino , Obesidade Infantil , Humanos , Criança , Masculino , Feminino , Obesidade Infantil/genética , Obesidade Infantil/epidemiologia , Obesidade Infantil/etnologia , Hispânico ou Latino/genética , Porto Rico/epidemiologia , Genótipo , Adolescente , Pré-Escolar , Testes Genéticos/métodos , Hiperfagia/genética
12.
Am J Intellect Dev Disabil ; 129(3): 175-190, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38657964

RESUMO

Hyperphagia is highly penetrant in Prader-Willi syndrome (PWS) and has increasingly been reported in other neurogenetic conditions (NGC). The Hyperphagia Questionnaire (HQ) was completed by caregivers of 4-8-year-olds with PWS (n = 17), Angelman syndrome (AS; n = 22), Williams syndrome (WS; n = 25), or low-risk controls (LRC; n = 35). All NGC groups were significantly elevated in HQ Total and Behavior scores compared to LRC. Only AS and WS were significantly elevated in the Drive domain, and only PWS in the Severity domain. After controlling for externalizing behavior, HQ Total scores were higher for PWS relative to other groups. Hyperphagic symptoms may not differentiate PWS from other NGCs in early childhood. However, hyperphagic phenotypes may be most severe in PWS. Further investigation of these profiles may inform etiology and syndrome-specific treatments.


Assuntos
Síndrome de Angelman , Hiperfagia , Síndrome de Prader-Willi , Humanos , Pré-Escolar , Masculino , Feminino , Síndrome de Prader-Willi/diagnóstico , Criança , Síndrome de Angelman/fisiopatologia , Síndrome de Angelman/diagnóstico
13.
Clin Obes ; 14(4): e12661, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38596856

RESUMO

Most patients with GNB1 encephalopathy have developmental delay and/or intellectual disability, brain anomalies and seizures. Recently, two cases with GNB1 encephalopathy caused by haploinsufficiency have been reported that also show a Prader-Willi-like phenotype of childhood hypotonia and severe obesity. Here we present three new cases from our expert centre for genetic obesity in which GNB1 truncating and splice variants, probably leading to haploinsufficiency, were identified. They all have obesity, hyperphagia and intellectual deficit. The clinical cases and their weight courses are presented, together with a review of all 68 published cases with GNB1 encephalopathy. Information on weight was not mentioned in most of these articles, so we contacted authors for additional clinical information on weight status and hyperphagia. Of the 42 patients whose weight status we could determine, obesity was present in 8 patients (19%). Obesity is significantly over-represented in the group with truncating and splicing variants. In this group, we see an obesity prevalence of 75%. Since GNB1 has been linked to several key genes in the hypothalamic leptin-melanocortin pathway, which regulates satiety and energy expenditure, our data support the potential association between GNB1 haploinsufficiency and genetic obesity. We also suggest GNB1 is a candidate gene for the known obesity phenotype of the 1p36 microdeletion syndrome given this chromosomal region includes the GNB1 gene. Knowledge of an additional obesity phenotype is important for prognosis, early interventions against obesity and awareness when prescribing weight-inducing medication.


Assuntos
Subunidades beta da Proteína de Ligação ao GTP , Haploinsuficiência , Obesidade , Humanos , Masculino , Feminino , Subunidades beta da Proteína de Ligação ao GTP/genética , Obesidade/genética , Criança , Deficiência Intelectual/genética , Pré-Escolar , Fenótipo , Adolescente , Hiperfagia/genética , Adulto
14.
Clin Obes ; 14(3): e12659, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38602039

RESUMO

Nearly 90 clinicians and researchers from around the world attended the first IMPROVE 2022 International Meeting on Pathway-Related Obesity. Delegates attended in person or online from across Europe, Argentina and Israel to hear the latest scientific and clinical developments in hyperphagia and severe, early-onset obesity, and set out a vision of excellence for the future for improving the diagnosis, treatment, and care of patients with melanocortin-4 receptor (MC4R) pathway-related obesity. The meeting co-chair Peter Kühnen, Charité Universitätsmedizin Berlin, Germany, indicated that change was needed with the rapidly increasing prevalence of obesity and the associated complications to improve the understanding of the underlying mechanisms and acknowledge that monogenic forms of obesity can play an important role, providing insights that can be applied to a wider group of patients with obesity. World-leading experts presented the latest research and led discussions on the underlying science of obesity, diagnosis (including clinical and genetic approaches such as the role of defective MC4R signalling), and emerging clinical data and research with targeted pharmacological approaches. The aim of the meeting was to agree on the questions that needed to be addressed in future research and to ensure that optimised diagnostic work-up was used with new genetic testing tools becoming available. This should aid the planning of new evidence-based treatment strategies for the future, as explained by co-chair Martin Wabitsch, Ulm University Medical Center, Germany.


Assuntos
Obesidade , Receptor Tipo 4 de Melanocortina , Humanos , Hiperfagia , Obesidade/terapia , Receptor Tipo 4 de Melanocortina/genética , Receptor Tipo 4 de Melanocortina/metabolismo , Transdução de Sinais
15.
J Neurodev Disord ; 16(1): 22, 2024 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-38671361

RESUMO

BACKGROUND: Prader-Willi syndrome (PWS) is a rare neurobehavioral-metabolic disease caused by the lack of paternally expressed genes in the chromosome 15q11-q13 region, characterized by hypotonia, neurocognitive problems, behavioral difficulties, endocrinopathies, and hyperphagia resulting in severe obesity if energy intake is not controlled. Diazoxide choline extended-release (DCCR) tablets have previously been evaluated for their effects on hyperphagia and other behavioral complications of people with PWS in a Phase 3 placebo-controlled study of participants with PWS, age 4 and older with hyperphagia (C601) and in an open label extension study, C602. METHODS: To better understand the longer-term impact of DCCR, a cohort from PATH for PWS, a natural history study that enrolled participants with PWS age 5 and older, who met the C601 age, weight and baseline hyperphagia inclusion criteria and had 2 hyperphagia assessments ≥ 6 months apart, were compared to the C601/C602 cohort. Hyperphagia was measured using the Hyperphagia Questionnaire for Clinical Trials (HQ-CT, range 0-36). The primary analysis used observed values with no explicit imputation of missing data. A sensitivity analysis was conducted in which all missing HQ-CT assessments in the C601/C602 cohort were assigned the highest possible value (36), representing the worst-case scenario. Other behavioral changes were assessed using the Prader-Willi Syndrome Profile questionnaire (PWSP). RESULTS: Relative to the PATH for PWS natural history study cohort, the DCCR-treated C601/C602 cohort showed significant improvements in HQ-CT score at 26 weeks (LSmean [SE] -8.3 [0.75] vs. -2.5 [0.43], p < 0.001) and 52 weeks (LSmean [SE] -9.2 [0.77] vs. -3.4 [0.47], p < 0.001). The comparison between the cohorts remained significant in the worst-case imputation sensitivity analysis. There were also significant improvements in all domains of the PWSP at 26 weeks (all p < 0.001) and 52 weeks (all p ≤ 0.003) for C601/C602 participants compared to the PATH for PWS participants. CONCLUSION: Long-term administration of DCCR to people with PWS resulted in changes in hyperphagia and other behavioral complications of PWS that are distinct from the natural history of the syndrome as exemplified by the cohort from PATH for PWS. The combined effects of administration of DCCR should reduce the burden of the syndrome on the patient, caregivers and their families, and thereby may benefit people with PWS and their families. TRIAL REGISTRATION: Clinical study C601 was originally registered on ClinicalTrials.gov on February 22, 2018 (NCT03440814). Clinical study C602 was originally registered on ClinicalTrials.gov on October 22, 2018 (NCT03714373). PATH for PWS was originally registered on ClinicalTrials.gov on October 24, 2018 (NCT03718416).


Assuntos
Preparações de Ação Retardada , Diazóxido , Hiperfagia , Síndrome de Prader-Willi , Humanos , Síndrome de Prader-Willi/complicações , Síndrome de Prader-Willi/tratamento farmacológico , Feminino , Masculino , Hiperfagia/tratamento farmacológico , Hiperfagia/etiologia , Criança , Adulto , Adolescente , Diazóxido/administração & dosagem , Diazóxido/farmacologia , Adulto Jovem , Pré-Escolar , Estudos de Coortes
16.
Eat Behav ; 53: 101874, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38636439

RESUMO

OBJECTIVE: To assess whether attentional bias to food cues and appetitive traits are independently and interactively associated with adiposity in adolescents. METHOD: Eighty-five adolescents, 14-17-years had their attentional bias to food images measured in a sated state by computing eye tracking measures of attention (first fixation duration, cumulative fixation duration) to food and control distractor images that bordered a computer game. Parents reported adolescent appetitive traits including the food approach domains of enjoyment of food, food responsiveness, emotional overeating, and the food avoidance domains of satiety responsiveness and emotional overeating through the Children's Eating Behavior Questionnaire. RESULTS: First fixation bias to food cues was positively associated with enjoyment of food, and negatively associated with satiety responsiveness. In a series of regression models adjusted for relevant covariates, first fixation bias to food cues (ß = 0.83, p = 0.007), higher food responsiveness (ß = 0.74, p < 0.001), higher emotional overeating (ß = 0.51, p = 0.002), and a composite appetite score (ß = 1.42, p < 0.001) were each significantly associated with greater BMI z-scores. In models assessing the interactive effects between attentional bias and appetitive traits, higher first fixation bias to food cues interacted synergistically with food responsiveness and emotional overeating in relation to BMI z-score. A synergistic interaction between first fixation bias to food cues and the composite appetite score in relation to BMI z-score was also observed. CONCLUSION: Individuals with high attentional bias to food cues and obesogenic appetitive traits may be particularly susceptible to weight gain.


Assuntos
Adiposidade , Viés de Atenção , Sinais (Psicologia) , Humanos , Adolescente , Feminino , Masculino , Viés de Atenção/fisiologia , Adiposidade/fisiologia , Apetite/fisiologia , Comportamento Alimentar/psicologia , Alimentos , Hiperfagia/psicologia , Pais/psicologia , Inquéritos e Questionários , Índice de Massa Corporal , Emoções/fisiologia
17.
Mol Metab ; 84: 101933, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38583571

RESUMO

OBJECTIVE: Alström Syndrome (AS), caused by biallelic ALMS1 mutations, includes obesity with disproportionately severe insulin resistant diabetes, dyslipidemia, and fatty liver. Prior studies suggest that hyperphagia is accounted for by loss of ALMS1 function in hypothalamic neurones, whereas disproportionate metabolic complications may be due to impaired adipose tissue expandability. We tested this by comparing the metabolic effects of global and mesenchymal stem cell (MSC)-specific Alms1 knockout. METHODS: Global Alms1 knockout (KO) mice were generated by crossing floxed Alms1 and CAG-Cre mice. A Pdgfrα-Cre driver was used to abrogate Alms1 function selectively in MSCs and their descendants, including preadipocytes. We combined metabolic phenotyping of global and Pdgfrα+ Alms1-KO mice on a 45% fat diet with measurements of body composition and food intake, and histological analysis of metabolic tissues. RESULTS: Assessed on 45% fat diet to promote adipose expansion, global Alms1 KO caused hyperphagia, obesity, insulin resistance, dyslipidaemia, and fatty liver. Pdgfrα-cre driven KO of Alms1 (MSC KO) recapitulated insulin resistance, fatty liver, and dyslipidaemia in both sexes. Other phenotypes were sexually dimorphic: increased fat mass was only present in female Alms1 MSC KO mice. Hyperphagia was not evident in male Alms1 MSC KO mice, but was found in MSC KO females, despite no neuronal Pdgfrα expression. CONCLUSIONS: Mesenchymal deletion of Alms1 recapitulates metabolic features of AS, including fatty liver. This confirms a key role for Alms1 in the adipose lineage, where its loss is sufficient to cause systemic metabolic effects and damage to remote organs. Hyperphagia in females may depend on Alms1 deficiency in oligodendrocyte precursor cells rather than neurones. AS should be regarded as a forme fruste of lipodystrophy.


Assuntos
Síndrome de Alstrom , Células-Tronco Mesenquimais , Camundongos Knockout , Animais , Camundongos , Masculino , Feminino , Células-Tronco Mesenquimais/metabolismo , Síndrome de Alstrom/metabolismo , Síndrome de Alstrom/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Resistência à Insulina , Fígado Gorduroso/metabolismo , Fígado Gorduroso/genética , Obesidade/metabolismo , Obesidade/genética , Hiperfagia/metabolismo , Hiperfagia/genética , Tecido Adiposo/metabolismo , Camundongos Endogâmicos C57BL , Composição Corporal
18.
Appetite ; 198: 107336, 2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38574819

RESUMO

Studies examining preconception eating behaviours with longitudinal dietary patterns from preconception to late pregnancy as well as gestational weight gain (GWG) are limited. We derived dietary pattern trajectories from preconception to late-pregnancy, and related preconception eating behaviours to these trajectories and GWG. Preconception eating behaviours were assessed using the Three-Factor Eating Questionnaire measuring cognitive restraint (CR) - conscious restriction of food intake, emotional eating (EE) - overeating in response to negative emotions, and uncontrolled eating (UE) - overeating with a feeling of lack of control. Dietary intakes were measured at preconception, 20-21 and 34-36 weeks' gestation with food frequency questionnaires. Dietary patterns were determined using factor analysis, and trajectories derived using group-based trajectory modelling. Inadequate and excessive GWG were defined according to Institute of Medicine guidelines based on weights at preconception and the last antenatal visit (median: 38 weeks' gestation). Two dietary patterns were derived: 'Fast Food, Fried Snacks and Desserts (FFD)' and 'Soup, Fish and Vegetables (SFV)'. Adherence trajectories from preconception to late-pregnancy were characterised as consistently high ("stable-high") and low ("stable-low"). Women with higher UE scores had higher odds of being in the "stable-high" trajectory (n = 34) of the FFD pattern [Odds Ratio (OR): 1.25, 95% Confidence Interval (CI): 1.03, 1.51], compared to "stable-low" (n = 260). Percentages of women with inadequate, adequate or excessive GWG were 21.7% (n = 70), 25.8% (n = 83), and 52.5% (n = 169), respectively; women with higher EE scores had a higher likelihood of excessive GWG [Relative Risk Ratio (RRR): 1.35, 95% CI: 1.02, 1.80], but this association was attenuated after adjusting for preconception body mass index. Eating behaviour interventions to improve dietary patterns among pregnant women may need to start as early as preconception, incorporating strategies to manage UE.


Assuntos
Dieta , Comportamento Alimentar , Ganho de Peso na Gestação , Humanos , Feminino , Gravidez , Adulto , Comportamento Alimentar/psicologia , Dieta/psicologia , Inquéritos e Questionários , Adulto Jovem , Índice de Massa Corporal , Hiperfagia/psicologia , Estudos Longitudinais , Padrões Dietéticos
19.
Appetite ; 198: 107355, 2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38621593

RESUMO

Associative learning can drive many different types of behaviors, including food consumption. Previous studies have shown that cues paired with food delivery while mice are hungry will lead to increased consumption in the presence of those cues at later times. We previously showed that overconsumption can be driven in male mice by contextual cues, using chow pellets. Here we extended our findings by examining other parameters that may influence the outcome of context-conditioned overconsumption training. We found that the task worked equally well in males and females, and that palatable substances such as high-fat diet and Ensure chocolate milkshake supported learning and induced overconsumption. Surprisingly, mice did not overconsume when sucrose was used as the reinforcer during training, suggesting that nutritional content is a critical factor. Interestingly, we also observed that diet-induced obese mice did not learn the task. Overall, we find that context-conditioned overconsumption can be studied in lean male and female mice, and with multiple reinforcer types.


Assuntos
Sinais (Psicologia) , Dieta Hiperlipídica , Camundongos Endogâmicos C57BL , Obesidade , Animais , Masculino , Feminino , Obesidade/etiologia , Obesidade/psicologia , Camundongos , Reforço Psicológico , Camundongos Obesos , Hiperfagia/psicologia , Comportamento Alimentar/psicologia , Sacarose/administração & dosagem , Magreza/psicologia
20.
BMC Public Health ; 24(1): 958, 2024 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-38575925

RESUMO

BACKGROUND: About 40% of people respond to stress by consuming more unhealthy foods. This behavior is associated with increased energy intake and the risk of obesity. As mobile health (mHealth) applications (apps) have been shown to be an easy-to-use intervention tool, the characterization of potential app users is necessary to develop target group-specific apps and to increase adherence rates. METHODS: This cross-sectional online survey was conducted in the spring of 2021 in Germany. Sociodemographic data and data on personality (Big Five Inventory, BFI-10), stress-eating (Salzburg Stress Eating Scale, SSES), and technology behavior (Personal Innovativeness in the Domain of Information Technology, PIIT; Technology Acceptance Model 3, TAM 3) were collected. RESULTS: The analysis included 1228 participants (80.6% female, mean age: 31.4 ± 12.8 years, mean body mass index (BMI): 23.4 ± 4.3 kg/m2). Based on the TAM score, 33.3% (409/1228) of the participants had a high intention to use a hypothetical mHealth app to avoid stress-overeating. These persons are characterized by a higher BMI (24.02 ± 4.47 kg/m2, p < 0.001), by being stress-overeaters (217/409, 53.1%), by the personality trait "neuroticism" (p < 0.001), by having specific eating reasons (all p < 0.01), and by showing a higher willingness to adopt new technologies (p < 0.001). CONCLUSION: This study suggests that individuals who are prone to stress-overeating are highly interested in adopting an mHealth app as support. Participants with a high intention to use an mHealth app seem to have a general affinity towards new technology (PIIT) and appear to be more insecure with conflicting motives regarding their diet. TRIAL REGISTRATION: This survey was registered in the German Clinical Trials Register (Registration number: DRKS00023984).


Assuntos
Aplicativos Móveis , Telemedicina , Adolescente , Adulto , Feminino , Humanos , Masculino , Adulto Jovem , Estudos Transversais , Hiperfagia , Obesidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA