Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 318
Filter
1.
Am J Physiol Endocrinol Metab ; 327(1): E69-E80, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38717361

ABSTRACT

Acylglycerophosphate acyltransferases (AGPATs) catalyze the de novo formation of phosphatidic acid to synthesize glycerophospholipids and triglycerides. AGPATs demonstrate unique physiological roles despite a similar biochemical function. AGPAT3 is highly expressed in the testis, kidney, and liver, with intermediate expression in adipose tissue. Loss of AGPAT3 is associated with reproductive abnormalities and visual dysfunction. However, the role of AGPAT3 in adipose tissue and whole body metabolism has not been investigated. We found that male Agpat3 knockout (KO) mice exhibited reduced body weights with decreased white and brown adipose tissue mass. Such changes were less pronounced in the female Agpat3-KO mice. Agpat3-KO mice have reduced plasma insulin growth factor 1 (IGF1) and insulin levels and diminished circulating lipid metabolites. They manifested intact glucose homeostasis and insulin sensitivity despite a lean phenotype. Agpat3-KO mice maintained an energy balance with normal food intake, energy expenditure, and physical activity, except for increased water intake. Their adaptive thermogenesis was also normal despite reduced brown adipose mass and triglyceride content. Mechanistically, Agpat3 was elevated during mouse and human adipogenesis and enriched in adipocytes. Agpat3-knockdown 3T3-L1 cells and Agpat3-deficient mouse embryonic fibroblasts (MEFs) have impaired adipogenesis in vitro. Interestingly, pioglitazone treatment rescued the adipogenic deficiency in Agpat3-deficient cells. We conclude that AGPAT3 regulates adipogenesis and adipose development. It is possible that adipogenic impairment in Agpat3-deficient cells potentially leads to reduced adipose mass. Findings from this work support the unique role of AGPAT3 in adipose tissue.NEW & NOTEWORTHY AGPAT3 deficiency results in male-specific growth retardation. It reduces adipose tissue mass but does not significantly impact glucose homeostasis or energy balance, except for influencing water intake in mice. Like AGPAT2, AGPAT3 is upregulated during adipogenesis, potentially by peroxisome proliferator-activated receptor gamma (PPARγ). Loss of AGPAT3 impairs adipocyte differentiation, which could be rescued by pioglitazone. Overall, AGPAT3 plays a significant role in regulating adipose tissue mass, partially involving its influence on adipocyte differentiation.


Subject(s)
1-Acylglycerol-3-Phosphate O-Acyltransferase , Adipocytes , Mice, Knockout , Animals , Female , Male , Mice , 1-Acylglycerol-3-Phosphate O-Acyltransferase/genetics , 1-Acylglycerol-3-Phosphate O-Acyltransferase/metabolism , Adipocytes/metabolism , Adipogenesis/genetics , Adipogenesis/physiology , Adipose Tissue, Brown/metabolism , Cell Differentiation , Energy Metabolism/genetics , Insulin Resistance/genetics , Mice, Inbred C57BL , Phenotype , Thermogenesis/genetics , Thinness/metabolism , Thinness/genetics
2.
Genes Genomics ; 46(5): 557-575, 2024 05.
Article in English | MEDLINE | ID: mdl-38483771

ABSTRACT

BACKGROUND: Analysing genomes of animal model organisms is widely used for understanding the genetic basis of complex traits and diseases, such as obesity, for which only a few mouse models exist, however, without their lean counterparts. OBJECTIVE: To analyse genetic differences in the unique mouse models of polygenic obesity (Fat line) and leanness (Lean line) originating from the same base population and established by divergent selection over more than 60 generations. METHODS: Genetic variability was analysed using WGS. Variants were identified with GATK and annotated with Ensembl VEP. g.Profiler, WebGestalt, and KEGG were used for GO and pathway enrichment analysis. miRNA seed regions were obtained with miRPathDB 2.0, LncRRIsearch was used to predict targets of identified lncRNAs, and genes influencing adipose tissue amount were searched using the IMPC database. RESULTS: WGS analysis revealed 6.3 million SNPs, 1.3 million were new. Thousands of potentially impactful SNPs were identified, including within 24 genes related to adipose tissue amount. SNP density was highest in pseudogenes and regulatory RNAs. The Lean line carries SNP rs248726381 in the seed region of mmu-miR-3086-3p, which may affect fatty acid metabolism. KEGG analysis showed deleterious missense variants in immune response and diabetes genes, with food perception pathways being most enriched. Gene prioritisation considering SNP GERP scores, variant consequences, and allele comparison with other mouse lines identified seven novel obesity candidate genes: 4930441H08Rik, Aff3, Fam237b, Gm36633, Pced1a, Tecrl, and Zfp536. CONCLUSION: WGS revealed many genetic differences between the lines that accumulated over the selection period, including variants with potential negative impacts on gene function. Given the increasing availability of mouse strains and genetic polymorphism catalogues, the study is a valuable resource for researchers to study obesity.


Subject(s)
Obesity , Thinness , Animals , Mice , Thinness/genetics , Thinness/metabolism , Obesity/genetics , Obesity/metabolism , Genome , Whole Genome Sequencing , Adipose Tissue/metabolism
3.
Eur J Hum Genet ; 32(1): 117-124, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37474786

ABSTRACT

Thinness and anorexia nervosa are both characterised by persistent low weight. Individuals with anorexia nervosa concurrently report distorted perceptions of their body and engage in weight-loss behaviours, whereas individuals with thinness often wish to gain weight. Both conditions are heritable and share genomics with BMI, but are not genetically correlated with each other. Based on their pattern of genetic associations with other traits, we explored differences between thinness and anorexia nervosa on a genomic level. In Part 1, using publicly available data, we compared genetic correlations of persistent thinness/anorexia nervosa with eleven psychiatric disorders. In Part 2, we identified individuals with adolescent persistent thinness in the Avon Longitudinal Study of Parents and Children (ALSPAC) by latent class growth analysis of measured BMI from 10 to 24 years (n = 6594) and evaluated associations with psychiatric and anthropometric polygenic scores. In Part 1, in contrast to the positive genetic correlations of anorexia nervosa with various psychiatric disorders, persistent thinness showed negative genetic correlations with attention deficit hyperactivity disorder (rgAN = 0.08 vs. rgPT = -0.30), alcohol dependence (rgAN = 0.07 vs. rgPT = -0.44), major depressive disorder (rgAN = 0.27 vs. rgPT = -0.18) and post-traumatic stress disorder (rgAN = 0.26 vs. rgPT = -0.20). In Part 2, individuals with adolescent persistent thinness in the ALSPAC had lower borderline personality disorder polygenic scores (OR = 0.77; Q = 0.01). Overall, results suggest that genetic variants associated with thinness are negatively associated with psychiatric disorders and therefore thinness may be differentiable from anorexia nervosa on a genomic level.


Subject(s)
Anorexia Nervosa , Depressive Disorder, Major , Adolescent , Child , Humans , Anorexia Nervosa/genetics , Anorexia Nervosa/psychology , Thinness/genetics , Longitudinal Studies , Genomics
4.
Philos Trans R Soc Lond B Biol Sci ; 378(1888): 20220205, 2023 10 23.
Article in English | MEDLINE | ID: mdl-37661743

ABSTRACT

Genetic disruption of key molecular components of the hypothalamic leptin-melanocortin pathway causes severe obesity in mice and humans. Physiological studies in people who carry these mutations have shown that the adipose tissue-derived hormone leptin primarily acts to defend against starvation. A lack of leptin causes an intense drive to eat and increases the rewarding properties of food, demonstrating that human appetite has a strong biological basis. Genetic studies in clinical- and population-based cohorts of people with obesity or thinness continue to provide new insights into the physiological mechanisms involved in weight regulation and identify molecular targets for weight loss therapy. This article is part of a discussion meeting issue 'Causes of obesity: theories, conjectures and evidence (Part II)'.


Subject(s)
Leptin , Thinness , Humans , Animals , Mice , Thinness/genetics , Obesity/genetics , Adipose Tissue , Mutation
5.
J Hum Nutr Diet ; 36(4): 1451-1467, 2023 08.
Article in English | MEDLINE | ID: mdl-36632775

ABSTRACT

BACKGROUND: Some persons are genetically resistant to obesity, but only a few studies have evaluated thinness genes for preventing obesity. We aimed to investigate the association of polygenic variants with being underweight and their interaction with the lifestyles of middle-aged and elderly persons and identify potential new genetic approaches for managing body weight. METHODS: In total, 58,701 participants aged 40-77 years were recruited from urban hospitals in Korea. Underweight (case) was defined as body mass index (BMI) < 18.5 kg m2 (n = 991) and normal weight (control, n = 21,921) was defined as 18.5 ≤ BMI < 23 kg m2 . A genome-wide association study was run to identify thinness-related single nucleotide polymorphisms (SNPs) after adjustment for compound factors using Gplink. The generalised multifactor dimensionality reduction program was used to identify the genetic variants with SNP-SNP interactions. The polygenic risk score (PRS) was calculated by summing up the number of risk alleles in each SNP and classifying them into low-, medium- and high-PRS. RESULTS: The best model included the ANK2_rs7656666, CAST_rs28042, SLC1A3_rs928431867, CHST12_rs2906173, ALOX5_rs1051713, RGS6_rs17180754, ST8SIA5_rs79491311 and DCC_rs35721894 alleles. The participants with high-PRS had a lower BMI (p < 0.0001) than those with low-PRS and were 3.834 (2.58-5.70) times more likely to be underweight after multivariate adjustment (p < 0.001). The selected SNPs were correlated with each other and highly expressed in brain-related genes. The genes with minor alleles of CAST_rs28042 and CHST12_rs2906173 exhibited a higher expression frequency in brain-related tissues. PRS had significant interactions with protein, sodium, indigestible carbohydrates, calcium intake and exercise (p < 0.05), influencing the underweight state. People with a high-PRS were more underweight than those with low-PRS under high protein, sodium, high calcium, low indigestible carbohydrate intake and low exercise by 3.75, 3.88, 7.05, 3.18 and 3.80 times, respectively (p < 0.0001). CONCLUSIONS: In conclusion, adults having a high-PRS were significantly correlated with being underweight, especially in combination with a particular nutritional status. These results show the potential for thinness genes to be applied to personalised nutrition for preventing obesity through targeted gene therapy.


Subject(s)
Genome-Wide Association Study , Thinness , Aged , Middle Aged , Humans , Adult , Thinness/genetics , Calcium , Obesity/genetics , Risk Factors , Body Mass Index , Sodium
6.
Genome Biol ; 23(1): 230, 2022 10 31.
Article in English | MEDLINE | ID: mdl-36316722

ABSTRACT

BACKGROUND: Overweight and obesity are defined by an anomalous or excessive fat accumulation that may compromise health. To find single-nucleotide polymorphisms (SNPs) influencing metabolic phenotypes associated with the obesity state, we analyze multiple anthropometric and clinical parameters in a cohort of 790 healthy volunteers and study potential associations with 48 manually curated SNPs, in metabolic genes functionally associated with the mechanistic target of rapamycin (mTOR) pathway. RESULTS: We identify and validate rs2291007 within a conserved region in the 3'UTR of folliculin-interacting protein FNIP2 that correlates with multiple leanness parameters. The T-to-C variant represents the major allele in Europeans and disrupts an ancestral target sequence of the miRNA miR-181b-5p, thus resulting in increased FNIP2 mRNA levels in cancer cell lines and in peripheral blood from carriers of the C allele. Because the miRNA binding site is conserved across vertebrates, we engineered the T-to-C substitution in the endogenous Fnip2 allele in mice. Primary cells derived from Fnip2 C/C mice show increased mRNA stability, and more importantly, Fnip2 C/C mice replicate the decreased adiposity and increased leanness observed in human volunteers. Finally, expression levels of FNIP2 in both human samples and mice negatively associate with leanness parameters, and moreover, are the most important contributor in a multifactorial model of body mass index prediction. CONCLUSIONS: We propose that rs2291007 influences human leanness through an evolutionarily conserved modulation of FNIP2 mRNA levels.


Subject(s)
MicroRNAs , Overweight , Humans , Animals , Mice , 3' Untranslated Regions , Overweight/genetics , Thinness/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Polymorphism, Single Nucleotide , RNA, Messenger/genetics , RNA, Messenger/metabolism , Obesity/genetics , Carrier Proteins/metabolism
7.
Int J Mol Sci ; 23(19)2022 Sep 29.
Article in English | MEDLINE | ID: mdl-36232772

ABSTRACT

Endometrial cancer is the most common gynaecological malignancy in developed countries. One of the largest risk factors for endometrial cancer is obesity. The aim of this study was to determine whether there are differences in the transcriptome of endometrial cancers from obese vs. lean women. Here we investigate the transcriptome of endometrial cancer between obese and lean postmenopausal women using rRNA-depleted RNA-Seq data from endometrial cancer tissues and matched adjacent non-cancerous endometrial tissues. Differential expression analysis identified 12,484 genes (6370 up-regulated and 6114 down-regulated) in endometrial cancer tissues from obese women, and 6219 genes (3196 up-regulated and 3023 down-regulated) in endometrial cancer tissues from lean women (adjusted p-value < 0.1). A gene ontology enrichment analysis revealed that the top 1000 up-regulated genes (by adjusted p-value) were enriched for growth and proliferation pathways while the top 1000 down-regulated genes were enriched for cytoskeleton restructure networks in both obese and lean endometrial cancer tissues. In this study, we also show perturbations in the expression of protein coding genes (HIST1H2BL, HIST1H3F, HIST1H2BH, HIST1H1B, TTK, PTCHD1, ASPN, PRELP, and CDH13) and the lncRNA MBNL1-AS1 in endometrial cancer tissues. Overall, this study has identified gene expression changes that are similar and also unique to endometrial cancers from obese vs. lean women. Furthermore, some of these genes may serve as prognostic biomarkers or, possibly, therapeutic targets for endometrial cancer.


Subject(s)
Endometrial Neoplasms , Obesity , RNA, Long Noncoding , Thinness , Transcriptome , Biomarkers/metabolism , Endometrial Neoplasms/genetics , Endometrial Neoplasms/metabolism , Female , Humans , Obesity/genetics , Obesity/metabolism , RNA, Long Noncoding/genetics , Thinness/genetics , Thinness/metabolism
8.
Proc Natl Acad Sci U S A ; 119(16): e2200476119, 2022 04 19.
Article in English | MEDLINE | ID: mdl-35412887

ABSTRACT

Augmentor α and ß (Augα and Augß) are newly discovered ligands of the receptor tyrosine kinases Alk and Ltk. Augα functions as a dimeric ligand that binds with high affinity and specificity to Alk and Ltk. However, a monomeric Augα fragment and monomeric Augß also bind to Alk and potently stimulate cellular responses. While previous studies demonstrated that oncogenic Alk mutants function as important drivers of a variety of human cancers, the physiological roles of Augα and Augß are poorly understood. Here, we investigate the physiological roles of Augα and Augß by exploring mice deficient in each or both Aug ligands. Analysis of mutant mice showed that both Augα single knockout and double knockout of Augα and Augß exhibit a similar thinness phenotype and resistance to diet-induced obesity. In the Augα-knockout mice, the leanness phenotype is coupled to increased physical activity. By contrast, Augß-knockout mice showed similar weight curves as the littermate controls. Experiments are presented demonstrating that Augα is robustly expressed and metabolically regulated in agouti-related peptide (AgRP) neurons, cells that control whole-body energy homeostasis in part via their projections to the paraventricular nucleus (PVN). Moreover, both Alk and melanocortin receptor-4 are expressed in discrete neuronal populations in the PVN and are regulated by projections containing Augα and AgRP, respectively, demonstrating that two distinct mechanisms that regulate pigmentation operate in the hypothalamus to control body weight. These experiments show that Alk-driven cancers were co-opted from a neuronal pathway in control of body weight, offering therapeutic opportunities for metabolic diseases and cancer.


Subject(s)
Anaplastic Lymphoma Kinase , Body Weight , Cytokines , Hypothalamus , Animals , Mice , Anaplastic Lymphoma Kinase/genetics , Anaplastic Lymphoma Kinase/metabolism , Cytokines/genetics , Cytokines/metabolism , Hypothalamus/metabolism , Ligands , Metabolic Networks and Pathways , Mice, Knockout , Neoplasms/enzymology , Protein Kinase Inhibitors/pharmacology , Thinness/genetics
9.
J Anim Physiol Anim Nutr (Berl) ; 106(4): 881-887, 2022 Jul.
Article in English | MEDLINE | ID: mdl-34643002

ABSTRACT

The study was performed to investigate protein digestibility and utilization in an F2 cross (M2-F2 cross) between the selected Fat (F) line and an M2 congenic line. The congenic M2 line carried the Fob3b2 quantitative trait locus (QTL) from the selected Lean (L) line previously shown to contain the Tst gene with leanness, anti-diabetic and resistance to diet-induced obesity effects. The main objective of the study was to test if some of the effects on leanness and obesity resistance of the L-line Fob3b2 could also be due to the effect of this QTL on nutrient digestibility and bioavailability. The F2 littermates carrying either the Fat line segment within the Fob3b2 region or the L-line were compared when fed the high-fat diet. Eleven mice per genotype were individually housed in metabolic cages. In 5-day experimental period, body mass and diet intake were measured. The part of study was done on the F and L line and tested the difference in apparent protein digestibility on low-fat (LFD) and high-fat (HFD) diet. The nitrogen content was determined in the diet, faeces, and urine based on which, the apparent protein digestibility, apparent protein biological value and apparent net protein utilization were calculated There were no significant differences in any of these parameters on congenic line, confirming that the phenotypic effect on adiposity between the genotypes in the M2-F2 population was not due to the differential effect of the Fob3b2 locus carrying the Tst gene on protein utilization. We conclude that the observed phenotypic effects of this gene region are due to direct metabolic actions rather than the effects on nutrient absorption and nitrogen utilization since there were no differences in apparent protein digestibility between L and F lines, irrespective to HFD or LFD. The age of animals had significant effect on the level of digestibility.


Subject(s)
Rodent Diseases , Thinness , Alleles , Animals , Biological Availability , Diet, High-Fat , Mice , Nitrogen/metabolism , Obesity/genetics , Obesity/veterinary , Proteins/metabolism , Rodent Diseases/genetics , Thinness/genetics , Thinness/veterinary
10.
J Nutr Biochem ; 99: 108857, 2022 01.
Article in English | MEDLINE | ID: mdl-34520852

ABSTRACT

Nutritional insults early in life have been associated with metabolic diseases in adulthood. We aimed to evaluate the effects of maternal food restriction during the suckling period on metabolism and interscapular brown adipose tissue (iBAT) thermogenically involved proteins in adult rat offspring. Wistar rats underwent food restriction by 50% during the first two-thirds of lactation (FR50 group). Control rats were fed ad libitum throughout lactation (CONT group). At birth, the litter size was adjusted to eight pups, and weaning was performed at 22 days old. Body weight and food and water intake were assessed every two days. High- (HCD, 4,589 cal) and normal-caloric diet (NCD, 3,860 cal) preferences, as well as food intake during the dark part of the cycle, were assessed. At 100 days old, the rats were euthanized, and blood and tissues were removed for further analyses. Adult FR50 rats, although hyperphagic and preferring to eat HCD (P<.001), were leaner (P<.001) than the CONT group. The FR50 rats, were normoglycemic (P=.962) and had hypertriglyceridemia (P<.01). In addition, the FR50 rats were dyslipidemic (P<.01), presenting with a high atherogenic risk by the Castelli indexes (P<.01), had a higher iBAT mass (P<.01), fewer ß3 adrenergic receptors (ß3-AR, P<.05) and higher iBAT expression of uncoupled protein 1 (UCP1, P<.05) and peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α, P<.001) than the CONT rats. In conclusion, maternal food restriction during early breastfeeding programs rat offspring to have a lean phenotype, despite hyperphagia, and increased iBAT UCP1 and PGC-1α protein expression.


Subject(s)
Adipose Tissue, Brown/metabolism , Breast Feeding , Lactation/metabolism , Thermogenesis , Thinness/metabolism , Animals , Caloric Restriction , Energy Metabolism , Female , Humans , Male , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Phenotype , Rats , Rats, Wistar , Receptors, Adrenergic, beta-3/genetics , Receptors, Adrenergic, beta-3/metabolism , Thinness/genetics , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
11.
Nutrients ; 13(11)2021 Oct 24.
Article in English | MEDLINE | ID: mdl-34836017

ABSTRACT

Body mass index (BMI) influences the prognosis of patients with non-small cell lung cancer (NSCLC), including both early-stage and late-stage NSCLC patients that are undergoing chemotherapies. However, earlier research on the relationship between BMI and survival in patients taking epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitors (TKIs) yielded contradictory results. These publications either had a limited number of patients or were getting TKIs in various lines of therapy, which might explain why the outcomes were contradictory. As a result, we undertook retrospective study to examine the effect of BMI on survival outcomes in patients with advanced EGFR mutant NSCLC receiving first-line EGFR-TKIs. We also compared the findings to those with wild-type EGFR. Between November 2010 and March 2014, 513 patients with advanced NSCLC were enrolled in the study. According to the adjusted BMI cut-off point for Asia, 35 out of 513 (6.8%) patients were underweight (BMI < 18.5 kg/m2), whereas 197 (38.4%) were overweight (BMI > 24 kg/m2). Overweight patients with wild-type EGFR exhibited longer progression-free survival (4.6 vs. 2.1 months, p = 0.003) and overall survival (OS) (8.9 vs. 4.3 months, p = 0.003) than underweight patients. Overweight patients with EGFR mutations had a longer OS than normal-weight patients (23.0 vs. 20.2 months, p = 0.025). Bodyweight reduction was related to a shorter OS in both the mutant EGFR patients (17.1 vs. 30.5 months, p < 0.001) and the wild-type EGFR patients (7.8 vs. 18.7 months, p < 0.001). In conclusion, advanced stages NSCLC patients with a lower BMI and early weight loss had a worse outcome that was independent of EGFR mutation status.


Subject(s)
Body Mass Index , Carcinoma, Non-Small-Cell Lung/mortality , Lung Neoplasms/mortality , Weight Loss/genetics , Aged , Carcinoma, Non-Small-Cell Lung/genetics , ErbB Receptors/genetics , Female , Humans , Lung Neoplasms/genetics , Lung Neoplasms/physiopathology , Male , Middle Aged , Mutation , Overweight/genetics , Overweight/mortality , Retrospective Studies , Survival Rate , Thinness/genetics , Thinness/mortality
12.
Front Endocrinol (Lausanne) ; 12: 698115, 2021.
Article in English | MEDLINE | ID: mdl-34646232

ABSTRACT

Previous studies have shown that agonists of GPR17 stimulate, while antagonists inhibit feeding. However, whole body knockout of GPR17 in mice of the C57Bl/6 strain did not affect energy balance, whereas selective knockout in oligodendrocytes or pro-opiomelanocortin neurons provided protection from high fat diet-induced obesity and impaired glucose homeostasis. We reasoned that whole body knockout of GPR17 in mice of the 129 strain might elicit more marked effects because the 129 strain is more susceptible than the C57Bl/6 strain to increased sympathetic activity and less susceptible to high fat diet-induced obesity. Consistent with this hypothesis, compared to wild-type mice, and when fed on either a chow or a high fat diet, GPR17 -/- mice of the 129 strain displayed increased expression of uncoupling protein-1 in white adipose tissue, lower body weight and fat content, reduced plasma leptin, non-esterified fatty acids and triglycerides, and resistance to high fat diet-induced glucose intolerance. Not only energy expenditure, but also energy intake was raised. Administration of leptin did not suppress the increased food intake in GPR17 -/- mice of the 129 strain, whereas it did suppress food intake in GPR17 +/+ mice. The only difference between GPR17 +/- and GPR17 +/+ mice of the C57Bl/6 strain was that the body weight of the GPR17 -/- mice was lower than that of the GPR17 +/+ mice when the mice were fed on a standard chow diet. We propose that the absence of GPR17 raises sympathetic activity in mice of the 129 strain in response to a low plasma fuel supply, and that the consequent loss of body fat is partly mitigated by increased energy intake.


Subject(s)
Energy Intake , Leptin/blood , Leptin/pharmacology , Nerve Tissue Proteins/genetics , Receptors, G-Protein-Coupled/genetics , Thinness/genetics , Adipose Tissue/metabolism , Animals , Body Composition/genetics , Energy Intake/drug effects , Energy Intake/physiology , Female , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Species Specificity , Thinness/blood
13.
JAMA Netw Open ; 4(7): e2118824, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34323981

ABSTRACT

Importance: Neurodevelopmental disorders have been proposed to involve alterations to epigenetic regulation, and epigenetic effects may extend to germline cells to affect later generations. Weight status may affect DNA methylation, and maternal weight before and during pregnancy has been associated with offspring DNA methylation as well as attention-deficit/hyperactivity disorder (ADHD). Objective: To assess whether a woman's weight before and during pregnancy is associated with ADHD in her grandchild. Design, Setting, and Participants: This cohort study analyzed data from 19 835 grandmother-mother dyads and 44 720 grandchildren in the Nurses' Health Study II (NHS-II) cohort (2001-2013), a population-based prospective cohort study. Cluster-weighted generalized estimating equations were modeled to estimate the association of grandmother's prepregnancy body mass index (BMI) and gestational weight gain with grandchild risk of ADHD. Data analyses were conducted from May 2018 to April 2021. Grandmothers reported their height and weight before, and weight gain during, their pregnancy with the NHS-II participants. Mothers self-reported height and weight prior to pregnancy. From those data, grandmother BMI and mother BMI were calculated as weight in kilograms divided by height in meters squared and categorized as underweight (<18.5), healthy/normal (18.5-24.9), overweight (25.0-29.9), or obese (≥30). Main Outcomes and Measures: Cases of ADHD identified by maternal report of having a child with a diagnosis of ADHD. Results: In total, 19 835 grandmothers (97.6% White race/ethnicity; 2113 [10.7%] prepregnancy underweight and 1391 [7.0%] prepregnancy overweight or obese) were included in this cohort study. Of 44 720 grandchildren, 3593 (8%) received a diagnosis of ADHD. Higher odds of ADHD among grandchildren were found for those whose grandmother was underweight compared with healthy weight prior to pregnancy with the NHS-II participant (adjusted odds ratio, 1.25; 95% CI, 1.10-1.42). By contrast, grandmother gestational weight gain was not significantly associated with risk of grandchild ADHD (adjusted odds ratio for <20 lbs [9.1 kg], 1.06; 95% CI, 0.96-1.16; adjusted odds ratio for >29 lbs [13.2 kg], 1.01; 95% CI, 0.91-1.13). Mother prepregnancy BMI showed an association with ADHD among offspring, with a stronger association detected for obese status (adjusted odds ratio, 1.27; 95% CI, 1.07-1.49) than for overweight status (adjusted odds ratio, 1.13; 95% CI, 1.02-1.26) compared with normal weight as a reference group. The positive association between grandmother prepregnancy underweight and ADHD risk among the grandchildren remained unchanged after further adjustment for potential mediators, including maternal prepregnancy BMI. Conclusions and Relevance: The results of this cohort study indicate that grandmother underweight prior to pregnancy is associated with an increased risk of ADHD among grandchildren, independent of grandmother gestational weight gain and independent of maternal prepregnancy weight status.


Subject(s)
Attention Deficit Disorder with Hyperactivity/genetics , Body Weight/genetics , Gestational Weight Gain/genetics , Grandparents , Maternal Health/statistics & numerical data , Aged , Body Height , Body Mass Index , Child , Child, Preschool , DNA Methylation , Epigenesis, Genetic , Female , Humans , Middle Aged , Mothers , Odds Ratio , Overweight/genetics , Prospective Studies , Thinness/genetics
14.
Front Endocrinol (Lausanne) ; 12: 628907, 2021.
Article in English | MEDLINE | ID: mdl-34248836

ABSTRACT

Obesity is an excess accumulation of body fat. Its progression rate has remained high in recent years. Therefore, the aim of this study was to diagnose important differentially expressed genes (DEGs) associated in its development, which may be used as novel biomarkers or potential therapeutic targets for obesity. The gene expression profile of E-MTAB-6728 was downloaded from the database. After screening DEGs in each ArrayExpress dataset, we further used the robust rank aggregation method to diagnose 876 significant DEGs including 438 up regulated and 438 down regulated genes. Functional enrichment analysis was performed. These DEGs were shown to be significantly enriched in different obesity related pathways and GO functions. Then protein-protein interaction network, target genes - miRNA regulatory network and target genes - TF regulatory network were constructed and analyzed. The module analysis was performed based on the whole PPI network. We finally filtered out STAT3, CORO1C, SERPINH1, MVP, ITGB5, PCM1, SIRT1, EEF1G, PTEN and RPS2 hub genes. Hub genes were validated by ICH analysis, receiver operating curve (ROC) analysis and RT-PCR. Finally a molecular docking study was performed to find small drug molecules. The robust DEGs linked with the development of obesity were screened through the expression profile, and integrated bioinformatics analysis was conducted. Our study provides reliable molecular biomarkers for screening and diagnosis, prognosis as well as novel therapeutic targets for obesity.


Subject(s)
Computational Biology , Gene Regulatory Networks , Molecular Docking Simulation , Obesity/genetics , Signal Transduction/genetics , Down-Regulation/genetics , Gene Ontology , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Protein Interaction Maps/genetics , ROC Curve , Reproducibility of Results , Thinness/genetics , Transcription Factors/metabolism , Transcriptome , Up-Regulation/genetics
16.
Physiol Rep ; 9(11): e14886, 2021 06.
Article in English | MEDLINE | ID: mdl-34086412

ABSTRACT

Cystic fibrosis (CF) is a life-limiting autosomal recessive genetic disease caused by variants in the CFTR gene, most commonly by the [F508del] variant. Although CF is a classical Mendelian disease, genetic variants in several modifier genes have been associated with variation of the clinical phenotype for pulmonary and gastrointestinal function and urogenital development. We hypothesized that whole genome sequencing of a well-phenotyped CF populations might identify novel variants in known, or hitherto unknown, modifier genes. Whole genome sequencing was performed on the Illumina HiSeq X platform for 98 clinically diagnosed cystic fibrosis patient samples from the Adult CF Clinic at the University of California San Diego (UCSD). We compared protein-coding, non-silent variants genome wide between CFTR [F508del] homozygotes vs CFTR compound heterozygotes. Based on a single variant score test, we found 3 SNPs in common variants (MAF >5%) that occurred at significantly different rates between homozygous [F508del]CFTR and compound heterozygous [F508del]CFTR patients. The 3 SNPs were all located in one gene on chromosome 2: Tensin 1 (TNS1: rs3796028; rs2571445: and rs918949). We observed significantly lower BMIs in homozygous [F508del]CFTR patients who were also homozygous for Tensin 1 rs918949 (T/T) (p = 0.023) or rs2571445 (G/G) (p = 0.02) variants. The Tensin 1 gene is thus a potential modifier gene for low BMI in CF patients homozygous for the [F508del]CFTR variant.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Tensins/physiology , Thinness/genetics , Adult , Body Mass Index , Cystic Fibrosis/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Female , Heterozygote , Homozygote , Humans , Male , Polymorphism, Single Nucleotide/genetics , Tensins/genetics , Whole Genome Sequencing
17.
Obesity (Silver Spring) ; 29 Suppl 1: S39-S50, 2021 04.
Article in English | MEDLINE | ID: mdl-33759393

ABSTRACT

Most individuals with obesity or overweight have difficulty maintaining weight loss. The weight-reduced state induces changes in many physiological processes that appear to drive weight regain. Here, we review the use of cell biology, genetics, and imaging techniques that are being used to begin understanding why weight regain is the normal response to dieting. As with obesity itself, weight regain has both genetic and environmental drivers. Genetic drivers for "thinness" and "obesity" largely overlap, but there is evidence for specific genetic loci that are different for each of these weight states. There is only limited information regarding the genetics of weight regain. Currently, most genetic loci related to weight point to the central nervous system as the organ responsible for determining the weight set point. Neuroimaging tools have proved useful in studying the contribution of the central nervous system to the weight-reduced state in humans. Neuroimaging technologies fall into three broad categories: functional, connectivity, and structural neuroimaging. Connectivity and structural imaging techniques offer unique opportunities for testing mechanistic hypotheses about changes in brain function or tissue structure in the weight-reduced state.


Subject(s)
Brain Mapping , Brain/diagnostic imaging , Genetic Testing , Weight Loss , Animals , Body Weight/physiology , Brain/physiopathology , Brain Mapping/methods , Genetic Predisposition to Disease , Humans , Neuroimaging/methods , Obesity/diagnosis , Obesity/genetics , Obesity/metabolism , Obesity/therapy , Overweight/diagnosis , Overweight/genetics , Overweight/metabolism , Overweight/therapy , Thinness/diagnosis , Thinness/genetics , Thinness/metabolism , Thinness/therapy , Weight Gain/genetics , Weight Gain/physiology , Weight Loss/genetics , Weight Loss/physiology
18.
Biochim Biophys Acta Mol Basis Dis ; 1867(6): 166107, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33621651

ABSTRACT

The melanocortin-3 receptor (MC3R) and melanocortin-4 receptor (MC4R), known as neural melanocortin receptors, have been implicated to be critical components of the hypothalamic leptin-melanocortin pathway and related to obesity pathogenesis. In contrast to extensive evidence from physiologic, biological, genetic studies demonstrating that MC4R is a critical regulator in obesity, whether MC3R mutation causes obesity is still controversial. In the present study, we screened for coding variants in the MC3R gene of 176 obese individuals (mean BMI 34.84 ± 0.19 kg/m2) and 170 lean controls (mean BMI 20.70 ± 0.08 kg/m2) to assess the prevalence of MC3R mutations in a Chinese cohort. Two novel mutations, A33D (c.C98 > A) and A259T (c.G775 > A), were identified in two subjects with morbid obesity, respectively. A259T was also identified in the carrier's sibling. In vitro functional studies showed that A33D was defective in the cAMP signaling pathway, whereas A259T MC3R had defective maximal binding and cAMP generation in response to NDP- and α-MSH, likely due to decreased cell surface expression. In addition, we showed that A33D and A259T were biased receptors and defect in constitutive activation of ERK1/2 signaling through MC3R might be a cause for morbid obesity. Our sequencing and co-segregation studies combined with comprehensive functional analysis demonstrated that A259T might be predisposing to obesity. Further investigations in larger cohorts will be needed in order to define this association and the specific phenotypic characteristics resulting from these mutations.


Subject(s)
Asian People/genetics , Mutation , Obesity/epidemiology , Receptor, Melanocortin, Type 3/genetics , Thinness/epidemiology , Adult , Case-Control Studies , China/epidemiology , Cohort Studies , Female , Humans , Male , Obesity/genetics , Obesity/pathology , Signal Transduction , Thinness/genetics , Thinness/pathology
19.
Eur J Sport Sci ; 21(7): 1073-1082, 2021 Jul.
Article in English | MEDLINE | ID: mdl-32713261

ABSTRACT

We evaluated the impact of maximal exercise on oxidative stress and DNA damage in peripheral blood mononuclear cells (PBMC) from sedentary and exercised lean and obese men. PBMC were collected before, immediately and 1-h after exercise and exposed to hydrogen peroxide (H2O2; 25 and 50 µM, 4 h). A leukocytosis was induced by maximal exercise immediately and 1-h after exercise in all groups. However, a lymphopenia was observed 1-h after exercise in the Sedentary obese group. In the control condition, low DNA damage index concomitant to increases in intracellular glutathione content (GSH) was identified immediately after exercise in all groups. However, higher DNA damage index and lipid peroxidation occurred 1-h after the bout in Sedentary and Exercised Obese groups. PBMC exposed to both H2O2 25 and 50 µM experienced higher DNA damage and lipid peroxidation index immediately after exercise in all groups. Both lipid peroxidation and DNA damage index remained higher in PBMC of Sedentary Lean, Sedentary Obese, and Exercised obese groups obtained 1-h after exercise in both H2O2 25 and 50 µM, with the highest values identified in PBMC from Sedentary Obese group. However, increases in GSH content were identified in treated PBMC from sedentary and exercised lean groups as well as exercised obese group 1-h after exercise. Habitual exercise confers increased resistance of PBMC to DNA damage induced by oxidative stress, reducing the detrimental effects of obesity.Keywords: Exercise, physical activity, DNA damage, obesity, mutagenesis, oxidative stress.


Subject(s)
DNA Damage , Exercise/physiology , Leukocytes, Mononuclear/metabolism , Obesity/genetics , Thinness/genetics , Adult , Glutathione/metabolism , Humans , Lipid Peroxidation , Male , Mutagenesis , Obesity/metabolism , Oxidative Stress , Thinness/metabolism , Young Adult
20.
Am J Physiol Endocrinol Metab ; 320(2): E244-E258, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33103450

ABSTRACT

During nutritional overload and obesity, hepatocyte function is grossly altered, and a subset of hepatocytes begins to accumulate fat droplets, leading to nonalcoholic fatty liver disease (NAFLD). Recent single-cell studies revealed how nonparenchymal cells, such as macrophages, hepatic stellate cells, and endothelial cells, heterogeneously respond to NAFLD. However, it remains to be characterized how hepatocytes, the major constituents of the liver, respond to nutritional overload in NAFLD. Here, using droplet-based, single-cell RNA sequencing (Drop-seq), we characterized how the transcriptomic landscape of individual hepatocytes is altered in response to high-fat diet (HFD) and NAFLD. We showed that the entire hepatocyte population undergoes substantial transcriptome changes upon HFD, although the patterns of alteration were highly heterogeneous, with zonation-dependent and -independent effects. Periportal (zone 1) hepatocytes downregulated many zone 1-specific marker genes, whereas a small number of genes mediating gluconeogenesis were upregulated. Pericentral (zone 3) hepatocytes also downregulated many zone 3-specific genes; however, they upregulated several genes that promote HFD-induced fat droplet formation, consistent with findings that zone 3 hepatocytes accumulate more lipid droplets. Zone 3 hepatocytes also upregulated ketogenic pathways as an adaptive mechanism to HFD. Interestingly, many of the top HFD-induced genes, which encode proteins regulating lipid metabolism, were strongly co-expressed with each other in a subset of hepatocytes, producing a variegated pattern of spatial co-localization that is independent of metabolic zonation. In conclusion, our data set provides a useful resource for understanding hepatocellular alteration during NAFLD at single cell level.


Subject(s)
Diet, High-Fat , Dietary Fats/pharmacology , Hepatocytes , Transcriptome/drug effects , Animals , Cells, Cultured , Gene Expression Profiling/methods , Gene Expression Regulation/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Male , Mice , Mice, Inbred C57BL , Obesity/genetics , Obesity/metabolism , Obesity/pathology , Single-Cell Analysis/methods , Thinness/genetics , Thinness/metabolism , Thinness/pathology
SELECTION OF CITATIONS
SEARCH DETAIL