1
|
Chen M, Xiao Z, Wang Y, Ou W, Hou C, Huang HZ. New insights on underlying shared genetic architectures and causality of underweight and depression in East Asian populations. J Affect Disord 2025; 380:226-229. [PMID: 40122258 DOI: 10.1016/j.jad.2025.03.126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Extensive evidence links a lower body mass index (BMI) to higher odds of depression in individuals of East Asian ancestry, differing from patterns observed in European populations. However, the shared genetic etiology underlying underweight and depression remains unclear in East Asian populations. METHOD Utilizing large-scale genome-wide association study (GWAS) data, we investigated the shared genetics between BMI (N = 323,298) and depression traits (N = 286,052) through linkage disequilibrium score regression, cross-trait meta-analysis and colocalization analysis. Additionally, we evaluated causal associations using bidirectional Mendelian randomization (MR) analysis. RESULTS We found a significantly negative genetic correlation between BMI and depression (rg = -0.19, P = 0.002). The cross-trait analysis identified 26 shared risk SNPs, including FTO and more. Moreover, the risk gene AGBL4 showed evidence of colocalization. Using the MR method, lower BMI was associated with higher odds of depression in individuals of East Asian ancestry (OR: 1.14, 95 % CI: 1.02 to 1.28, P = 0.021) but no reverse causal effect was observed. CONCLUSIONS Our study indicates a genetic correlation, shared risk genes, and causality between underweight and depression in East Asian populations. These findings provide insights into the potential mechanisms behind their comorbidity and inform the future development of therapeutics for East Asian populations.
Collapse
Affiliation(s)
- Ming Chen
- Guangdong Mental Health Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510100, China
| | - Zhen Xiao
- Guangdong Mental Health Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510100, China
| | - Yueya Wang
- Guangdong Mental Health Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510100, China
| | - Wanqi Ou
- Guangdong Mental Health Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510100, China
| | - Cailan Hou
- Guangdong Mental Health Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510100, China.
| | - Hao-Zhang Huang
- Department of Cardiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510100, China; Department of Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510100, China.
| |
Collapse
|
2
|
Martin SS, Aday AW, Allen NB, Almarzooq ZI, Anderson CAM, Arora P, Avery CL, Baker-Smith CM, Bansal N, Beaton AZ, Commodore-Mensah Y, Currie ME, Elkind MSV, Fan W, Generoso G, Gibbs BB, Heard DG, Hiremath S, Johansen MC, Kazi DS, Ko D, Leppert MH, Magnani JW, Michos ED, Mussolino ME, Parikh NI, Perman SM, Rezk-Hanna M, Roth GA, Shah NS, Springer MV, St-Onge MP, Thacker EL, Urbut SM, Van Spall HGC, Voeks JH, Whelton SP, Wong ND, Wong SS, Yaffe K, Palaniappan LP. 2025 Heart Disease and Stroke Statistics: A Report of US and Global Data From the American Heart Association. Circulation 2025; 151:e41-e660. [PMID: 39866113 DOI: 10.1161/cir.0000000000001303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
BACKGROUND The American Heart Association (AHA), in conjunction with the National Institutes of Health, annually reports the most up-to-date statistics related to heart disease, stroke, and cardiovascular risk factors, including core health behaviors (smoking, physical activity, nutrition, sleep, and obesity) and health factors (cholesterol, blood pressure, glucose control, and metabolic syndrome) that contribute to cardiovascular health. The AHA Heart Disease and Stroke Statistical Update presents the latest data on a range of major clinical heart and circulatory disease conditions (including stroke, brain health, complications of pregnancy, kidney disease, congenital heart disease, rhythm disorders, sudden cardiac arrest, subclinical atherosclerosis, coronary heart disease, cardiomyopathy, heart failure, valvular disease, venous thromboembolism, and peripheral artery disease) and the associated outcomes (including quality of care, procedures, and economic costs). METHODS The AHA, through its Epidemiology and Prevention Statistics Committee, continuously monitors and evaluates sources of data on heart disease and stroke in the United States and globally to provide the most current information available in the annual Statistical Update with review of published literature through the year before writing. The 2025 AHA Statistical Update is the product of a full year's worth of effort in 2024 by dedicated volunteer clinicians and scientists, committed government professionals, and AHA staff members. This year's edition includes a continued focus on health equity across several key domains and enhanced global data that reflect improved methods and incorporation of ≈3000 new data sources since last year's Statistical Update. RESULTS Each of the chapters in the Statistical Update focuses on a different topic related to heart disease and stroke statistics. CONCLUSIONS The Statistical Update represents a critical resource for the lay public, policymakers, media professionals, clinicians, health care administrators, researchers, health advocates, and others seeking the best available data on these factors and conditions.
Collapse
|
3
|
Zhang T, Park S. Energy Intake-Dependent Genetic Associations with Obesity Risk: BDNF Val66Met Polymorphism and Interactions with Dietary Bioactive Compounds. Antioxidants (Basel) 2025; 14:170. [PMID: 40002356 PMCID: PMC11851519 DOI: 10.3390/antiox14020170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 01/25/2025] [Accepted: 01/28/2025] [Indexed: 02/27/2025] Open
Abstract
Obesity represents a complex interplay between genetics, nutrition, and lifestyle. This study aimed to elucidate the intricate relationship between genetic variants, energy intake, and bioactive compounds in influencing obesity risk, particularly in low energy intake, to reveal how dietary intake modulates molecular-level interactions. We analyzed 53,117 participants stratified by obesity status and energy intake levels. Genome-wide association studies explored the genetic variants associated with obesity risk in low-energy- and high-energy-intake subgroups. Advanced computational approaches, including molecular docking, k-means clustering, and uniform manifold approximation and projection (UMAP), were employed to analyze interactions between missense variants and natural compounds. Ten genetic variants were significantly associated with obesity, particularly in participants with low energy intake. The most prominent variants included brain-derived neurotrophic factor (BDNF) Val66Met polymorphism (rs6265). Molecular docking identified 152 bioactive compounds with strong binding affinity to BDNF Val66Met, including 107 compounds binding to both wild and mutant types. Citrus fruits and green vegetables showed selective binding to the mutant type. Antioxidant nutrient intake (anthocyanins, isoflavonoids, vitamins C and E, selenium) was higher in lean versus obese individuals in the high-energy-intake group. Alcohol consumption and selenium intake modulated polygenic risk scores' influence on obesity risk in high-energy-intake individuals. Notably, citrus fruit intake correlated with lower BMI across all BDNF rs6265 genotypes. In conclusion, energy intake-specific genetic associations with obesity and identifies potential bioactive compounds for targeted interventions. The findings suggest that antioxidant nutrient intake, particularly from citrus fruits, may help manage obesity risk, especially in individuals with specific genetic variants.
Collapse
Affiliation(s)
- Ting Zhang
- Department of Bioconvergence, Hoseo University, Asan 31499, Republic of Korea;
| | - Sunmin Park
- Department of Bioconvergence, Hoseo University, Asan 31499, Republic of Korea;
- Department of Food and Nutrition, Obesity/Diabetes Research Center, Hoseo University, Asan 31499, Republic of Korea
| |
Collapse
|
4
|
Wang RQ, Deng ZM, Chen GT, Dai FF, Xia LB. Obesity and recurrent spontaneous abortion: the crucial role of weight management in pregnancy. Reprod Biol Endocrinol 2025; 23:10. [PMID: 39844265 PMCID: PMC11752768 DOI: 10.1186/s12958-024-01326-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 11/29/2024] [Indexed: 01/24/2025] Open
Abstract
Recurrent spontaneous abortion (RSA), characterized by the loss of two or more pregnancies, impacts approximately 1-2% of couples and poses a significant challenge for individuals of childbearing age. The precise mechanisms underlying RSA remain incompletely understood. Concurrently, the global prevalence of obesity is on the rise, with obesity being closely associated with female reproductive disorders and infertility. This study initially examines the pathways through which obesity contributes to RSA, encompassing factors such as embryonic euploid miscarriage, endometrial development, immune function, among others. Furthermore, adipokines and the fat mass and obesity-related (FTO) are identified as potential contributors to RSA. The study also explores the enhancement of pregnancy outcomes through various weight management strategies, with a particular focus on the roles of dietary interventions, physical activity, and weight control during pregnancy. Obesity is closely related to RSA in multiple aspects. Additional clinical prospective and experimental studies are required to explore its precise pathogenesis. Through this review, we aim to provide strategies for improvement and treatment approaches for RSA related to obesity. Through this review, we suggest potential clinical management strategies and research avenues aimed at offering enhancements and therapeutic insights for miscarriages linked to obesity and its associated risk factors.
Collapse
Affiliation(s)
- Rui-Qi Wang
- Department of Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, 430060, China
| | - Zhi-Min Deng
- Department of Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, 430060, China
| | - Gan-Tao Chen
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China.
| | - Fang-Fang Dai
- Department of Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, 430060, China.
| | - Liang-Bin Xia
- Department of Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, 430060, China.
| |
Collapse
|
5
|
de Soysa AKH, Langaas M, Grill V, Martins C, Løvold Mostad I. Exploring associations between the FTO rs9939609 genotype and plasma concentrations of appetite-related hormones in adults with obesity. PLoS One 2025; 20:e0312815. [PMID: 39792913 PMCID: PMC11723609 DOI: 10.1371/journal.pone.0312815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 10/14/2024] [Indexed: 01/12/2025] Open
Abstract
Associations between variants in the FTO locus and plasma concentrations of appetite related hormones are inconsistent, and might not work in a dose dependent fashion in people with obesity. Moreover, it is relevant to report meal related plasma concentrations of these hormones in persons with obesity given the growing interest in their pharmacological potential in obesity therapy. We find it clinically relevant to examine associations between the SNP rs9939609 genotypes and homeostatic appetite regulation in individuals with BMI ≥35 kg/m2. This study explored associations of the rs9939609 genotypes to plasma concentrations of acylated ghrelin, active glucagon-like peptide 1 (GLP-1), and total peptide YY (PYY), and moderating effects of fat mass (FM), in 96 adults (69% female) with BMI ≥35 kg/m2, using a cross sectional observation study designed to have 1/3 of participants each with genotypes TT, AT and AA, respectively. Participants were median (25th, 75th percentile) 42.5 (32, 50) years of age, weighed 120.9 (109.6, 142.4) kg, and had a BMI of 42.8 (39.5, 46.4) kg/m2. Acylated ghrelin, active GLP-1, and total PYY were measured in the fasted state and half-hourly for 2.5h after a standardized meal. We evaluated associations between genotype and appetite hormones in regression analysis controlling for FM and sex. Genotype did not associate with fasting or postprandial (area under curve, AUC) GLP-1 or PYY. Genotype did not associate with fasting acylated ghrelin, but in females with genotype AA, increased FM was associated with higher fasting and postprandial (AUC) acylated ghrelin concentrations relative to genotypes TT (fasting p = 0.025; AUC p = 0.004) and AT (fasting p = 0.002; AUC p < 0.001). This novel finding warrants further investigation.
Collapse
Affiliation(s)
- Ann Kristin Hjelle de Soysa
- Outpatient Obesity Clinic, Clinic of Surgery, St. Olavs hospital–Trondheim University Hospital, Trondheim, Norway
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Mette Langaas
- Department of Mathematical Sciences, Faculty of Information Technology and Electrical Engineering, Norwegian University of Science and Technology, Trondheim, Norway
| | - Valdemar Grill
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Catia Martins
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Center for Obesity Research and Innovation (ObeCe), Clinic of Surgery, St. Olavs Hospital–Trondheim University Hospital, Trondheim, Norway
| | - Ingrid Løvold Mostad
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Nutrition and Speech-Language Therapy, Clinic of Rehabilitation, St. Olavs hospital–Trondheim University Hospital, Trondheim, Norway
| |
Collapse
|
6
|
García-Pastor T, Muñoz-Puente I, Pérez-Pelayo M, Púa I, Roberts JD, Del Coso J. Maximal Fat Oxidation During Exercise in Healthy Individuals: Lack of Genetic Association with the FTO rs9939609 Polymorphism. Genes (Basel) 2024; 16:4. [PMID: 39858551 PMCID: PMC11764838 DOI: 10.3390/genes16010004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 12/22/2024] [Accepted: 12/23/2024] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: Previous studies suggest that there is a genetically determined component of fat oxidation at rest and during exercise. To date, the FTO gene has been proposed as a candidate gene to affect fat oxidation during exercise because of the association of the "at-risk" A allele with different obesity-related factors such as increased body fat, higher appetite and elevated insulin and triglyceride levels. The A allele of the FTO gene may also be linked to obesity through a reduced capacity for fat oxidation during exercise, a topic that remains largely underexplored in the current literature. The aim of this study was to analyze the association between the FTO rs9939609 polymorphism with the rate of fat oxidation during exercise and metabolic syndrome criteria in healthy participants. Methods: A total of 80 healthy participants (41 men and 39 women) underwent comprehensive assessments, including measurements of anthropometric variables, blood pressure and blood measures of fasting glucose, triglycerides, low- and high-density lipoprotein cholesterol (LDL-c and HDL-c), insulin, interleukin-6 (IL-6) and C-reactive protein (CRP) concentrations. Additionally, the Homeostatic Model Assessment (HOMA-IR) was used to evaluate insulin resistance. Peak oxygen uptake (VO2peak) and maximal fat oxidation rate (MFO) were also measured during an incremental cycling test. FTO rs9939609 genotyping (TT, AT, AA) was performed using genomic DNA samples obtained from a buccal swab and measured with PCR. Results: There were 32 participants (40.0%) with the TT genotype; 31 (38.8%) with the AT genotype; and 17 (21.2%) with the AA genotype. Age, body characteristics, VO2peak, blood pressure and blood variables were similar across all three genotypes. However, serum insulin concentration and HOMA-IR were associated with the FTO rs9939609 genotype with higher values in AA with respect to AT and TT participants (p < 0.050). Still, MFO was similar in TT, AT and AA participants (0.35 ± 0.13, 0.37 ± 0.11, 0.33 ± 0.11 g/min, p = 0.702). In the dominant model, there was no statistical difference between TT and A allele carriers. However, the recessive model revealed that AA participants had higher values of body mass, body mass index, blood insulin concentration and HOMA-IR than T allele carriers (p < 0.050), with no differences in MFO. Conclusions: In our sample of healthy individuals, the FTO rs9939609 polymorphism was associated with several phenotypes associated with obesity and insulin resistance, particularly under the AA vs. T allele/recessive model. However, the FTO rs9939609 polymorphism was not associated with MFO during exercise as fat oxidation was similar across genotypes. This suggests that reduced fat oxidation during exercise is unlikely to be a cause of the obesogenic influence of the FTO AA genotype. Clinically, these findings suggest that the obesogenic effects of the FTO AA genotype are unlikely driven by impaired fat oxidation during exercise. Instead, attention should focus on mechanisms like appetite regulation and energy intake. Moreover, exercise interventions may still effectively mitigate obesity risk, as AA individuals retain normal fat oxidation capacity during exercise.
Collapse
Affiliation(s)
- Teresa García-Pastor
- Exercise Physiology Laboratory (GIDECS), Facultad HM de Ciencias de la Salud, Universidad Camilo José Cela, 28692 Villanueva de la Cañada, Madrid, Spain;
- Instituto de Investigación Sanitaria HM Hospitales, 28692 Madrid, Spain
| | - Iván Muñoz-Puente
- Exercise Physiology Laboratory (GIDECS), Facultad HM de Ciencias de la Salud, Universidad Camilo José Cela, 28692 Villanueva de la Cañada, Madrid, Spain;
- Instituto de Investigación Sanitaria HM Hospitales, 28692 Madrid, Spain
| | | | - Isabel Púa
- Severo Ochoa Hospital, 28914 Leganés, Madrid, Spain; (M.P.-P.); (I.P.)
| | - Justin D. Roberts
- Cambridge Centre for Sport and Exercise Sciences, Faculty of Science and Engineering, Anglia Ruskin University, Cambridge CB1 1PT, UK;
| | - Juan Del Coso
- Sport Sciences Research Centre, Rey Juan Carlos University, 28943 Fuenlabrada, Madrid, Spain
| |
Collapse
|
7
|
Bombassaro B, Araujo EP, Velloso LA. The hypothalamus as the central regulator of energy balance and its impact on current and future obesity treatments. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2024; 68:e240082. [PMID: 39876968 PMCID: PMC11771753 DOI: 10.20945/2359-4292-2024-0082] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 06/10/2024] [Indexed: 01/31/2025]
Abstract
The hypothalamus is a master regulator of energy balance in the body. First-order hypothalamic neurons localized in the arcuate nucleus sense systemic signals that indicate the energy stores in the body. Through distinct projections, arcuate nucleus neurons communicate with second-order neurons, which are mostly localized in the paraventricular nucleus and in the lateral hypothalamus. The signals then proceed to third- and fourth-order neurons that activate complex responses aimed at maintaining whole-body energy homeostasis. During the last 30 years, since the identification of leptin in 1994, there has been a great advance in the unveiling of the hypothalamic and extra-hypothalamic neuronal networks that control energy balance. This has contributed to the characterization of the mechanisms by which glucagon-like peptide-1 receptor agonists promote body mass reduction and has opened new windows of opportunity for the development of drugs to treat obesity. This review presents an overview of the mechanisms involved in the hypothalamic regulation of energy balance and discusses how advancements in this field are contributing to the development of new pharmacological strategies to treat obesity.
Collapse
Affiliation(s)
- Bruna Bombassaro
- Universidade de Campinas Centro de Pesquisa em Obesidade e Comorbidades CampinasSP Brasil Centro de Pesquisa em Obesidade e Comorbidades, Universidade de Campinas, Campinas, SP, Brasil
| | - Eliana P Araujo
- Universidade de Campinas Centro de Pesquisa em Obesidade e Comorbidades CampinasSP Brasil Centro de Pesquisa em Obesidade e Comorbidades, Universidade de Campinas, Campinas, SP, Brasil
| | - Licio A Velloso
- Universidade de Campinas Centro de Pesquisa em Obesidade e Comorbidades CampinasSP Brasil Centro de Pesquisa em Obesidade e Comorbidades, Universidade de Campinas, Campinas, SP, Brasil
| |
Collapse
|
8
|
Poosri S, Boonyuen U, Chupeerach C, Soonthornworasiri N, Kwanbunjan K, Prangthip P. Association of FTO variants rs9939609 and rs1421085 with elevated sugar and fat consumption in adult obesity. Sci Rep 2024; 14:25618. [PMID: 39463443 PMCID: PMC11514288 DOI: 10.1038/s41598-024-77004-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 10/18/2024] [Indexed: 10/29/2024] Open
Abstract
This cross-sectional study explores the impact of FTO gene single nucleotide polymorphisms (SNPs) rs9939609 and rs1421085 on dietary habits contributing to obesity risk in Thai adults. The study enrolled 384 participants from Bangkok, categorized as non-obese (BMI < 25 kg/m2) or obese (BMI ≥ 25 kg/m2) based on WHO Asia Pacific Guidelines. Genotyping for FTO variants was performed using DNA from blood samples. While both SNPs adhered to Hardy-Weinberg equilibrium, the association between risk alleles and anthropometric measurements was not statistically significant. However, risk allele carriers showed significantly higher intakes of sugar and saturated fat compared to homozygous dominant individuals. In the obese group, the odds ratio for high-sugar intake was 2.22 (95% CI 1.13-4.37, p = 0.021) for rs9939609 risk allele carriers. For high-saturated fat intake, the odds ratio was 1.86 (95% CI 1.02-3.40, p = 0.041). Similar associations were observed for rs1421085. Risk allele carriers also exhibited significantly higher leptin levels (p < 0.043) and a positive correlation with myeloperoxidase levels (p < 0.038). These findings highlight the complex relationship between FTO risk alleles, increased consumption of sugar and saturated fat, and obesity-related parameters. The insights emphasize the importance of considering both genetic and dietary factors in obesity prevention strategies.
Collapse
Affiliation(s)
- Sakawrut Poosri
- Department of Tropical Nutrition and Food Science, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Usa Boonyuen
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Chaowanee Chupeerach
- Food and Nutrition Academic and Research Cluster, Institute of Nutrition, Mahidol University, Nakhon Pathom, Thailand
| | | | - Karunee Kwanbunjan
- Department of Tropical Nutrition and Food Science, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Pattaneeya Prangthip
- Department of Tropical Nutrition and Food Science, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
9
|
Ning M, Xu A, Zeng R, Xue J, Wang B, Liu X. The effect of overweight/obesity and FTO gene polymorphism on liver function-related parameters in Chinese adolescents. THE NATIONAL MEDICAL JOURNAL OF INDIA 2024; 37:248-252. [PMID: 39953885 DOI: 10.25259/nmji_264_2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2025]
Abstract
Background Overweight/obesity is an important risk factor for liver disease, affecting changes in liver function-related parameters. The fat mass and obesity associated (FTO) gene has been reported to have a link between overweight/obesity and liver fat metabolism. We studied the association of FTO rs9939609 variants with liver function-related parameters and overweight/obesity in Chinese adolescents aged 16 to 26 as freshmen. Methods We examined rs9939609 polymorphisms in 198 control and 173 overweight/obese people, and the genotypes of the samples were analyzed by Sanger sequencing. We investigated the effects of FTO gene polymorphism on overweight/obesity and liver function-related parameters. Results The values of aspartate transaminase (AST), alanine transaminase (ALT), gamma-glutamyl transferase (GGT) and alkaline phosphatase (ALP) in overweight/obesity group were significantly higher than those in control group. The rs9939609 AA genotype increased the risk of overweight/obesity by 3.45 times independent of body mass index (BMI) compared with TT genotype, the rs9939609 (AA) genotype was significantly associated with AST and ALT. Conclusion FTO rs9939609 variants were associated with overweight/obesity and overweight/obesity has a significant influence on the increased liver function-related parameters. The rs9939609 (AA) positively correlated with AST and ALT levels. Overweight/obese patients should pay more attention to liver function-related parameters.
Collapse
Affiliation(s)
- Meiwei Ning
- Department of Laboratory Medicine, Heilongjiang University of Chinese Medicine Jiamusi College, Jiamusi, 154007, China
| | - Aohong Xu
- Department of Laboratory Medicine, Center for Medical Experiments (CME), Guangming District People's Hospital, Shenzhen, 518106, China
| | - Rong Zeng
- Department of Laboratory Medicine, Hubei University of Chinese Medicine, Hongshan District, Wuhan, Hubei, China
| | - Jinruo Xue
- Department of Laboratory Medicine, Hubei University of Chinese Medicine, Hongshan District, Wuhan, Hubei, China
| | - Boda Wang
- Clinical Laboratory, Huangjiahu Hospital of Hubei University of Chinese Medicine, Wuhan, China
| | - Xiang Liu
- Department of Laboratory Medicine, Hubei University of Chinese Medicine, Hongshan District, Wuhan, Hubei, China
| |
Collapse
|
10
|
Cheraghi S, Taheri G, Safari S, Bakhshandeh H, Malek M, Moghimian B, Mottaghi A. Survey on Interaction Between Nutrient Status and Selected Polymorphisms in Association with Weight Loss of Patients with Severe Obesity Underwent Bariatric Surgery. Obes Surg 2024; 34:2854-2861. [PMID: 38972938 DOI: 10.1007/s11695-024-07305-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 05/18/2024] [Accepted: 05/23/2024] [Indexed: 07/09/2024]
Abstract
BACKGROUND There is little information about the effect of single nucleotide polymorphisms (SNP) and nutritional status and weight loss after bariatric surgery. This study investigated the interactive effect of eight obesity-related SNPs and nutritional status on weight loss after Roux-en-Y gastric bypass (RYGB). METHOD This is a case-control study. After 1-year follow-up, the patients who underwent RYGB were dividing into two groups. The case group consisted of patients who lost more than 50% of their excess body weight (EBW%) 1 year after the surgery. The control group included patients who lost < 50% of EBW at same time frame. Then, the relationship between eight SNPs related to UCP2, FTO, LEPR, GHRL, and NPY genes with weight loss were checked. RESULTS In this study, 160 patients were recruited. The median of age for case and control group were 43 and 42 respectively. The presence of mutant variant NPYrs16147 had a significant relationship in terms of weight loss between the two groups (P > 0.05). In dominant model, two SNPs, UCP2 rs659366 and UCP2 rs660339, showed protective effect of the vitamin D deficiency. CONCLUSION In conclusion, the presence mutant variant of NPYrs16147 is directly related to the incidence of weight loss greater than 50% of EBW. However, it is apparent individual behavioral, dietary, and other factors may have more influence on weight loss among patients underwent RYGB.
Collapse
Affiliation(s)
- Sara Cheraghi
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran
| | - Golmehr Taheri
- Department of Endocrinology and Metabolism, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Saeed Safari
- Firoozgar General Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Hooman Bakhshandeh
- Heart Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mojtaba Malek
- Research Center for Prevention of Cardiovascular Diseases, Institute of Endocrinology Metabolism, Iran University of Medical Sciences, Endocrinology & Metabolism, Tehran, Iran
| | - Bahar Moghimian
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran
| | - Azadeh Mottaghi
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran.
- Research Center for Prevention of Cardiovascular Diseases, Institute of Endocrinology Metabolism, Iran University of Medical Sciences, Endocrinology & Metabolism, Tehran, Iran.
| |
Collapse
|
11
|
Eghbali M, Mottaghi A, Taghizadeh S, Cheraghi S. Genetic Variants in the Fat Mass and Obesity-Associated Gene and Risk of Obesity/Overweight in Children and Adolescents: A Systematic Review and Meta-Analysis. Endocrinol Diabetes Metab 2024; 7:e00510. [PMID: 38973101 PMCID: PMC11227992 DOI: 10.1002/edm2.510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/16/2024] [Accepted: 06/23/2024] [Indexed: 07/09/2024] Open
Abstract
OBJECTIVE The variations in the single-nucleotide polymorphisms (SNPs) of the fat mass and obesity (FTO)-associated gene have been linked to being overweight or obese in children. In this research a thorough examination was performed to elucidate the connection between various FTO gene SNPs and overweight or obesity in children and adolescents. METHOD We searched PubMed, Google scholar, Web of Science and Scopus until January 2024 to find studies that investigate the association between different SNPs of FTO gene and the risk of overweight/obesity in children and adolescents. After filtering the relevant studies, meta-analysis was used to quantify the association of FTO gene SNPs within different genetic inheritance models. RESULTS We have identified 32 eligible studies with 14,930 obese/overweight cases and 24,765 healthy controls. Our recessive model showed a significant association with rs9939609 (OR: 1.56, 95% CI: 1.20; 2.02, p < 0.01) and rs1421085 (OR: 1.77, 95% CI: 1.14; 2.75, p < 0.01). Besides, in the homozygote model, rs1421085 showed the highest association (OR: 2.32, 95% CI: 1.38; 3.89, p < 0.01) with the risk of obesity in a population of children and adolescents. Moreover, there are other SNPs of FTO genes, such as rs9921255, rs9928094 and rs9930333, which showed a positive association with obesity and overweight. However, their effects were evaluated in very few numbers of studies. CONCLUSION In this study, we have found that the FTO rs9939609 and rs1421085 are associated to an increased risk of obesity among children and adolescents. Besides, the findings of this study further reaffirmed the established link between rs9939609 and obesity in children and adolescents.
Collapse
Affiliation(s)
- Maryam Eghbali
- Endocrine Research Center, Institute of Endocrinology and MetabolismIran University of Medical SciencesTehranIran
| | - Azadeh Mottaghi
- Research Center for Prevention of Cardiovascular Diseases, Endocrinology & Metabolism, Institute of Endocrinology MetabolismIran University of Medical SciencesTehranIran
| | - Sara Taghizadeh
- Translational Ophthalmology Research CenterTehran University of Medical SciencesTehranIran
| | - Sara Cheraghi
- Endocrine Research Center, Institute of Endocrinology and MetabolismIran University of Medical SciencesTehranIran
| |
Collapse
|
12
|
Wang X, Gan M, Wang Y, Wang S, Lei Y, Wang K, Zhang X, Chen L, Zhao Y, Niu L, Zhang S, Zhu L, Shen L. Comprehensive review on lipid metabolism and RNA methylation: Biological mechanisms, perspectives and challenges. Int J Biol Macromol 2024; 270:132057. [PMID: 38710243 DOI: 10.1016/j.ijbiomac.2024.132057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 04/26/2024] [Accepted: 05/01/2024] [Indexed: 05/08/2024]
Abstract
Adipose tissue plays a crucial role in maintaining energy balance, regulating hormones, and promoting metabolic health. To address disorders related to obesity and develop effective therapies, it is essential to have a deep understanding of adipose tissue biology. In recent years, RNA methylation has emerged as a significant epigenetic modification involved in various cellular functions and metabolic pathways. Particularly in the realm of adipogenesis and lipid metabolism, extensive research is ongoing to uncover the mechanisms and functional importance of RNA methylation. Increasing evidence suggests that RNA methylation plays a regulatory role in adipocyte development, metabolism, and lipid utilization across different organs. This comprehensive review aims to provide an overview of common RNA methylation modifications, their occurrences, and regulatory mechanisms, focusing specifically on their intricate connections to fat metabolism. Additionally, we discuss the research methodologies used in studying RNA methylation and highlight relevant databases that can aid researchers in this rapidly advancing field.
Collapse
Affiliation(s)
- Xingyu Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Mailin Gan
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Yan Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Saihao Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Yuhang Lei
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Kai Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Xin Zhang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Lei Chen
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Ye Zhao
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Lili Niu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Shunhua Zhang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Li Zhu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China.
| | - Linyuan Shen
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China.
| |
Collapse
|
13
|
Prunell-Castañé A, Beyer F, Witte V, Sánchez Garre C, Hernán I, Caldú X, Jurado MÁ, Garolera M. From the reward network to whole-brain metrics: structural connectivity in adolescents and young adults according to body mass index and genetic risk of obesity. Int J Obes (Lond) 2024; 48:567-574. [PMID: 38145996 DOI: 10.1038/s41366-023-01451-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 12/05/2023] [Accepted: 12/12/2023] [Indexed: 12/27/2023]
Abstract
BACKGROUND Obesity is a multifactorial condition. Genetic variants, such as the fat mass and obesity related gene (FTO) polymorphism, may increase the vulnerability of developing obesity by disrupting dopamine signaling within the reward network. Yet, the association of obesity, genetic risk of obesity, and structural connectivity of the reward network in adolescents and young adults remains unexplored. We investigate, in adolescents and young adults, the structural connectivity differences in the reward network and at the whole-brain level according to body mass index (BMI) and the FTO rs9939609 polymorphism. METHODS One hundred thirty-two adolescents and young adults (age range: [10, 21] years, BMI z-score range: [-1.76, 2.69]) were included. Genetic risk of obesity was determined by the presence of the FTO A allele. Whole-brain and reward network structural connectivity were analyzed using graph metrics. Hierarchical linear regression was applied to test the association between BMI-z, genetic risk of obesity, and structural connectivity. RESULTS Higher BMI-z was associated with higher (B = 0.76, 95% CI = [0.30, 1.21], P = 0.0015) and lower (B = -0.003, 95% CI = [-0.006, -0.00005], P = 0.048) connectivity strength for fractional anisotropy at the whole-brain level and of the reward network, respectively. The FTO polymorphism was not associated with structural connectivity nor with BMI-z. CONCLUSIONS We provide evidence that, in healthy adolescents and young adults, higher BMI-z is associated with higher connectivity at the whole-brain level and lower connectivity of the reward network. We did not find the FTO polymorphism to correlate with structural connectivity. Future longitudinal studies with larger sample sizes are needed to assess how genetic determinants of obesity change brain structural connectivity and behavior.
Collapse
Affiliation(s)
- Anna Prunell-Castañé
- Departament de Psicologia Clínica i Psicobiologia, Facultat de Psicologia, Universitat de Barcelona, Passeig de la Vall d'Hebron, 171, 08035, Barcelona, Spain
- Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
- Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain
| | - Frauke Beyer
- Clinic for Cognitive Neurology, University of Leipzig Medical Center, Leipzig, Germany
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Veronica Witte
- Clinic for Cognitive Neurology, University of Leipzig Medical Center, Leipzig, Germany
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Consuelo Sánchez Garre
- Pediatric Endocrinology Unit, Hospital de Terrassa, Consorci Sanitari de Terrassa, Terrassa, Barcelona, Spain
| | - Imma Hernán
- Molecular Genetics Unit, Consorci Sanitari de Terrassa, Terrassa, Spain
| | - Xavier Caldú
- Departament de Psicologia Clínica i Psicobiologia, Facultat de Psicologia, Universitat de Barcelona, Passeig de la Vall d'Hebron, 171, 08035, Barcelona, Spain
- Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
- Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain
| | - María Ángeles Jurado
- Departament de Psicologia Clínica i Psicobiologia, Facultat de Psicologia, Universitat de Barcelona, Passeig de la Vall d'Hebron, 171, 08035, Barcelona, Spain.
- Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.
- Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain.
| | - Maite Garolera
- Brain, Cognition and Behavior: Clinical Research, Consorci Sanitari de Terrassa, Terrassa, Barcelona, Spain
- Neuropsychology Unit, Hospital de Terrassa, Consorci Sanitari de Terrassa, Terrassa, Barcelona, Spain
| |
Collapse
|
14
|
Martin SS, Aday AW, Almarzooq ZI, Anderson CAM, Arora P, Avery CL, Baker-Smith CM, Barone Gibbs B, Beaton AZ, Boehme AK, Commodore-Mensah Y, Currie ME, Elkind MSV, Evenson KR, Generoso G, Heard DG, Hiremath S, Johansen MC, Kalani R, Kazi DS, Ko D, Liu J, Magnani JW, Michos ED, Mussolino ME, Navaneethan SD, Parikh NI, Perman SM, Poudel R, Rezk-Hanna M, Roth GA, Shah NS, St-Onge MP, Thacker EL, Tsao CW, Urbut SM, Van Spall HGC, Voeks JH, Wang NY, Wong ND, Wong SS, Yaffe K, Palaniappan LP, American Heart Association Council on Epidemiology and Prevention Statistics Committee and Stroke Statistics Subcommittee. 2024 Heart Disease and Stroke Statistics: A Report of US and Global Data From the American Heart Association. Circulation 2024; 149:e347-e913. [PMID: 38264914 PMCID: PMC12146881 DOI: 10.1161/cir.0000000000001209] [Citation(s) in RCA: 845] [Impact Index Per Article: 845.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2024]
Abstract
BACKGROUND The American Heart Association (AHA), in conjunction with the National Institutes of Health, annually reports the most up-to-date statistics related to heart disease, stroke, and cardiovascular risk factors, including core health behaviors (smoking, physical activity, nutrition, sleep, and obesity) and health factors (cholesterol, blood pressure, glucose control, and metabolic syndrome) that contribute to cardiovascular health. The AHA Heart Disease and Stroke Statistical Update presents the latest data on a range of major clinical heart and circulatory disease conditions (including stroke, brain health, complications of pregnancy, kidney disease, congenital heart disease, rhythm disorders, sudden cardiac arrest, subclinical atherosclerosis, coronary heart disease, cardiomyopathy, heart failure, valvular disease, venous thromboembolism, and peripheral artery disease) and the associated outcomes (including quality of care, procedures, and economic costs). METHODS The AHA, through its Epidemiology and Prevention Statistics Committee, continuously monitors and evaluates sources of data on heart disease and stroke in the United States and globally to provide the most current information available in the annual Statistical Update with review of published literature through the year before writing. The 2024 AHA Statistical Update is the product of a full year's worth of effort in 2023 by dedicated volunteer clinicians and scientists, committed government professionals, and AHA staff members. The AHA strives to further understand and help heal health problems inflicted by structural racism, a public health crisis that can significantly damage physical and mental health and perpetuate disparities in access to health care, education, income, housing, and several other factors vital to healthy lives. This year's edition includes additional global data, as well as data on the monitoring and benefits of cardiovascular health in the population, with an enhanced focus on health equity across several key domains. RESULTS Each of the chapters in the Statistical Update focuses on a different topic related to heart disease and stroke statistics. CONCLUSIONS The Statistical Update represents a critical resource for the lay public, policymakers, media professionals, clinicians, health care administrators, researchers, health advocates, and others seeking the best available data on these factors and conditions.
Collapse
|
15
|
Karmazyn M, Gan XT. Molecular and Cellular Mechanisms Underlying the Cardiac Hypertrophic and Pro-Remodelling Effects of Leptin. Int J Mol Sci 2024; 25:1137. [PMID: 38256208 PMCID: PMC10816997 DOI: 10.3390/ijms25021137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Since its initial discovery in 1994, the adipokine leptin has received extensive interest as an important satiety factor and regulator of energy expenditure. Although produced primarily by white adipocytes, leptin can be synthesized by numerous tissues including those comprising the cardiovascular system. Cardiovascular function can thus be affected by locally produced leptin via an autocrine or paracrine manner but also by circulating leptin. Leptin exerts its effects by binding to and activating specific receptors, termed ObRs or LepRs, belonging to the Class I cytokine family of receptors of which six isoforms have been identified. Although all ObRs have identical intracellular domains, they differ substantially in length in terms of their extracellular domains, which determine their ability to activate cell signalling pathways. The most important of these receptors in terms of biological effects of leptin is the so-called long form (ObRb), which possesses the complete intracellular domain linked to full cell signalling processes. The heart has been shown to express ObRb as well as to produce leptin. Leptin exerts numerous cardiac effects including the development of hypertrophy likely through a number of cell signaling processes as well as mitochondrial dynamics, thus demonstrating substantial complex underlying mechanisms. Here, we discuss mechanisms that potentially mediate leptin-induced cardiac pathological hypertrophy, which may contribute to the development of heart failure.
Collapse
|
16
|
Yu H, Armstrong N, Pavela G, Kaiser K. Sex and Race Differences in Obesity-Related Genetic Susceptibility and Risk of Cardiometabolic Disease in Older US Adults. JAMA Netw Open 2023; 6:e2347171. [PMID: 38064210 PMCID: PMC10709778 DOI: 10.1001/jamanetworkopen.2023.47171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 10/29/2023] [Indexed: 12/18/2023] Open
Abstract
Importance The fat mass and obesity-associated gene (FTO) is associated with obesity phenotypes, but the association is inconsistent across populations. Within-population differences may explain some of the variability observed. Objective To investigate sex differences in the association between FTO single-nucleotide variants (SNVs) and obesity traits among self-identified non-Hispanic Black and non-Hispanic White US adults, to examine whether the SNVs were associated with cardiometabolic diseases, and to evaluate whether obesity mediated the association between FTO SNVs and cardiometabolic diseases. Design, Setting, and Participants This cross-sectional study used data from the Reasons for Geographic and Racial Differences in Stroke (REGARDS) study, a US population-based cohort study with available genetic data (assayed in 2018) and phenotypic data at baseline (enrolled 2003-2007). Participants were aged 45 to 98 years at baseline. Data were analyzed from October 2021 to October 2022. Exposures Eleven SNVs in the FTO gene present among both Black and White participants. Main Outcomes and Measures Objectively measured obesity indicators (body mass index and waist-to-height ratio), objectively measured and/or self-reported cardiometabolic diseases (hypertension, stroke history, heart disease, and diabetes), and self-reported social-economic and psychosocial status. Results A total of 10 447 participants (mean [SD] age, 64.4 [9.7] years; 5276 [55.8%] women; 8743 [83.7%] Black and 1704 [16.3%] White) were included. In the White group, 11 FTO SNVs were significantly associated with obesity, hypertension, and diabetes using linear models (eg, body mass index: β = 0.536; 95% CI, 0.197-0.875), but none of the FTO SNVs were associated with obesity traits in the Black group. White males had a higher risk of obesity while White females had a higher risk of hypertension and diabetes. However, 1 FTO SNV (rs1121980) was associated with a direct increase in the risk of heart disease in Black participants not mediated by obesity (c' = 0.145 [SE, 0.0517]; P = .01). Conclusions and Relevance In this cross-sectional study of obesity phenotypes and their association with cardiometabolic diseases, the tested FTO SNVs reflected sex differences in White participants. Different patterns of associations were observed among self-identified Black participants. Therefore, these results could inform future work discovering risk alleles or risk scores unique to Black individuals or further investigating genetic risk in all US residents.
Collapse
Affiliation(s)
- Hairui Yu
- Department of Health Behavior, School of Public Health, University of Alabama at Birmingham
- Department of Family and Community Medicine, School of Medicine, University of Alabama at Birmingham
| | - Nicole Armstrong
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham
| | - Greg Pavela
- Department of Health Behavior, School of Public Health, University of Alabama at Birmingham
| | - Kathryn Kaiser
- Department of Health Behavior, School of Public Health, University of Alabama at Birmingham
| |
Collapse
|
17
|
Huang C, Chen W, Wang X. Studies on the fat mass and obesity-associated (FTO) gene and its impact on obesity-associated diseases. Genes Dis 2023; 10:2351-2365. [PMID: 37554175 PMCID: PMC10404889 DOI: 10.1016/j.gendis.2022.04.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 03/29/2022] [Accepted: 04/01/2022] [Indexed: 10/18/2022] Open
Abstract
Obesity has become a major health crisis in the past ∼50 years. The fat mass and obesity-associated (FTO) gene, identified by genome-wide association studies (GWAS), was first reported to be positively associated with obesity in humans. Mice with more copies of the FTO gene were observed to be obese, while loss of the gene in mice was found to protect from obesity. Later, FTO was found to encode an m6A RNA demethylase and has a profound effect on many biological and metabolic processes. In this review, we first summarize recent studies that demonstrate the critical roles and regulatory mechanisms of FTO in obesity and metabolic disease. Second, we discuss the ongoing debates concerning the association between FTO polymorphisms and obesity. Third, since several small molecule drugs and micronutrients have been found to regulate metabolic homeostasis through controlling the expression or activity of FTO, we highlight the broad potential of targeting FTO for obesity treatment. Improving our understanding of FTO and the underlying mechanisms may provide new approaches for treating obesity and metabolic diseases.
Collapse
Affiliation(s)
- Chaoqun Huang
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Ministry of Education, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, Zhejiang 310058, China
| | - Wei Chen
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Ministry of Education, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, Zhejiang 310058, China
| | - Xinxia Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Ministry of Education, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, Zhejiang 310058, China
| |
Collapse
|
18
|
Qiu L, Jing Q, Li Y, Han J. RNA modification: mechanisms and therapeutic targets. MOLECULAR BIOMEDICINE 2023; 4:25. [PMID: 37612540 PMCID: PMC10447785 DOI: 10.1186/s43556-023-00139-x] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 07/28/2023] [Indexed: 08/25/2023] Open
Abstract
RNA modifications are dynamic and reversible chemical modifications on substrate RNA that are regulated by specific modifying enzymes. They play important roles in the regulation of many biological processes in various diseases, such as the development of cancer and other diseases. With the help of advanced sequencing technologies, the role of RNA modifications has caught increasing attention in human diseases in scientific research. In this review, we briefly summarized the basic mechanisms of several common RNA modifications, including m6A, m5C, m1A, m7G, Ψ, A-to-I editing and ac4C. Importantly, we discussed their potential functions in human diseases, including cancer, neurological disorders, cardiovascular diseases, metabolic diseases, genetic and developmental diseases, as well as immune disorders. Through the "writing-erasing-reading" mechanisms, RNA modifications regulate the stability, translation, and localization of pivotal disease-related mRNAs to manipulate disease development. Moreover, we also highlighted in this review all currently available RNA-modifier-targeting small molecular inhibitors or activators, most of which are designed against m6A-related enzymes, such as METTL3, FTO and ALKBH5. This review provides clues for potential clinical therapy as well as future study directions in the RNA modification field. More in-depth studies on RNA modifications, their roles in human diseases and further development of their inhibitors or activators are needed for a thorough understanding of epitranscriptomics as well as diagnosis, treatment, and prognosis of human diseases.
Collapse
Affiliation(s)
- Lei Qiu
- State Key Laboratory of Biotherapy and Cancer Center, Research Laboratory of Tumor Epigenetics and Genomics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, P.R. China
| | - Qian Jing
- State Key Laboratory of Biotherapy and Cancer Center, Research Laboratory of Tumor Epigenetics and Genomics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, P.R. China
| | - Yanbo Li
- State Key Laboratory of Biotherapy and Cancer Center, Research Laboratory of Tumor Epigenetics and Genomics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, P.R. China
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Junhong Han
- State Key Laboratory of Biotherapy and Cancer Center, Research Laboratory of Tumor Epigenetics and Genomics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, P.R. China.
| |
Collapse
|
19
|
Szcześniewska P, Bryl E, Dutkiewicz A, Borkowska AR, Bilska K, Paszyńska E, Słopień A, Dmitrzak-Węglarz M, Hanć T. Cool executive functions and their association with body mass & fatness and the FTO gene in school-aged children. Sci Rep 2023; 13:12148. [PMID: 37500688 PMCID: PMC10374888 DOI: 10.1038/s41598-023-38808-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 07/14/2023] [Indexed: 07/29/2023] Open
Abstract
The FTO gene rs9936909 polymorphism is one of the well-documented single nucleotide polymorphisms in the context of increased risk of obesity, including in children. Few studies have tested the association of the FTO gene with cognitive functions. Deficits of "cool" executive functions (EFs) are considered a potential risk factor for excessive weight. The aims of our study were to investigate whether cool EFs are associated with the Body Mass Index, the Fat Mass Index and the risk of excess body mass and overfatness in neurotypically school-aged children, and whether the FTO gene polymorphism is involved in development of this possible association. The sample consisted of 553 children aged 6-12 years old. A body composition analysis, a neuropsychological assessment of EFs, and FTO polymorphism genotyping were performed in the children studied. The study found a significant association of an interference effect in theStroop Color-Word Interference Task and the risk of excessive body fatness, but not excessive body mass. There were no explicit associations between the FTO genotype and EFs deficits. Environmental factors, and particularly low maternal education, appeared to be the strongest contributors to the increased risk of obesity.
Collapse
Affiliation(s)
- Paula Szcześniewska
- Institute of Biology and Human Evolution, Faculty of Biology, Adam Mickiewicz University, 61-614, Poznan, Poland.
| | - Ewa Bryl
- Institute of Biology and Human Evolution, Faculty of Biology, Adam Mickiewicz University, 61-614, Poznan, Poland
| | - Agata Dutkiewicz
- Department of Child and Adolescent Psychiatry, Poznan University of Medical Sciences, 60-572, Poznan, Poland
| | - Aneta R Borkowska
- Faculty of Education and Psychology, Maria Curie-Sklodowska University, 20-400, Lublin, Poland
| | - Karolina Bilska
- Department of Psychiatric Genetics, Department of Psychiatry, Poznan University of Medical Sciences, 61-701, Poznan, Poland
| | - Elżbieta Paszyńska
- Department of Integrated Dentistry, Poznan University of Medical Sciences, 60-812, Poznan, Poland
| | - Agnieszka Słopień
- Department of Child and Adolescent Psychiatry, Poznan University of Medical Sciences, 60-572, Poznan, Poland
| | - Monika Dmitrzak-Węglarz
- Department of Psychiatric Genetics, Department of Psychiatry, Poznan University of Medical Sciences, 61-701, Poznan, Poland
| | - Tomasz Hanć
- Institute of Biology and Human Evolution, Faculty of Biology, Adam Mickiewicz University, 61-614, Poznan, Poland.
| |
Collapse
|
20
|
Hu Q, Gao Y, Xie Y, Li D, An T, Chen L, Ji W, Jin Y, Long J, Yang H, Duan G, Chen S. Mechanism of RNA m 6 A methylation modification regulating NLRP3 inflammasome activation for hand, foot, and mouth disease progression. J Med Virol 2023; 95:e28939. [PMID: 37409616 DOI: 10.1002/jmv.28939] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/20/2023] [Accepted: 06/22/2023] [Indexed: 07/07/2023]
Abstract
Some children infected with hand, foot, and mouth disease (HFMD) caused by enterovirus 71 (EV71) progressed to severe disease with various neurological complications in the short term, with a poor prognosis and high mortality. Studies had revealed that RNA N6 -methyladenosine (m6 A) modification had a significant impact on EV71 replication, but it was unknown how m6 A modification regulated the host cell's innate immune response brought on by EV71 infection. We used MeRIP-seq (methylation RNA immunoprecipitation sequencing), RNA-seq (RNA sequencing), cell transfection, and other techniques. MeRIP-seq and RNA-seq results showed the m6 A methylation modification map of control and EV71-infected groups of RD cells. And multilevel validation indicated that decreased expression of demethylase FTO (fat mass and obesity-associated protein) was responsible for the elevated total m6 A modification levels in EV71-infected RD cells and that thioredoxin interacting protein (TXNIP) may be a target gene for demethylase FTO action. Further functional experiments showed that demethylase knockdown of FTO promoted TXNIP expression, activation of NLRP3 inflammasome and promoted the release of proinflammatory factors in vitro, and the opposite result occurred with demethylase FTO overexpression. And further tested in an animal model of EV71 infection in vitro, with results consistent with in vitro. Our findings elucidated that depletion of the demethylase FTO during EV71 infection increased the m6 A modification level of TXNIP mRNA 3' untranslated region (UTR), enhancing mRNA stability, and promoting TXNIP expression. Consequently, the NLRP3 inflammasome was stimulated, leading to the release of proinflammatory factors and facilitating HFMD progression.
Collapse
Affiliation(s)
- Quanman Hu
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Yanlei Gao
- Zhengzhou Center for Disease Control and Prevention, Zhengzhou, Henan, China
| | - Yaqi Xie
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Dong Li
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Tongyan An
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Long Chen
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Wangquan Ji
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Yuefei Jin
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Jinzhao Long
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Haiyan Yang
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Guangcai Duan
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Shuaiyin Chen
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
21
|
Rader MA, Jaime OG, Abarca VO, Young KA. Photoperiod alters testicular methyltransferase complex mRNA expression in Siberian hamsters. Gen Comp Endocrinol 2023; 333:114186. [PMID: 36521516 PMCID: PMC10575611 DOI: 10.1016/j.ygcen.2022.114186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 11/06/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022]
Abstract
Exposure to long photoperiods stimulates, whereas exposure to short photoperiods transiently inhibit testicular function in Siberian hamsters via well-described neuroendocrine mechanisms. However, less is known about the intra-testicular regulation of these photoperiod-mediated changes. N6-methyladenosine (m6A) is one of the most common mRNA modifications in eukaryotes, with alterations in m6A mRNA methylation affecting testis function and fertility. We hypothesized that genes controlling m6A methylation such as methyltransferase-like-3 (Mettl3) and -14 (Mettl14) and Wilms' tumor-1 associated protein (Wtap), part of an mRNA methylating methyl-transferase complex, or the fat-mass-and-obesity-associated (Fto) and the α-ketoglutarate-dependent dioxygenase alkB homolog-5 (Alkbh5) genes responsible for m6A demethylation, may be differentially regulated by photoperiod in the testis. Male hamsters were exposed to long (LD, control) photoperiod for 14-weeks, short (SD) photoperiod for 2, 5, 8, 11 and 14-weeks to induce regression, or SD for 14-weeks followed by transfer to LD for 1, 2, 4 or 8-weeks to induce recrudescence (post-transfer, PT). SD exposure significantly reduced body, testis, and epididymal masses compared to all other groups. Spermatogenic index, seminiferous tubule diameters and testosterone concentrations significantly decreased in SD as compared to LD, returning to levels no different than LD in post-transfer groups. SD exposure significantly decreased Wtap, Fto, Alkbh5, but increased Mettl14 mRNA expression as compared to LD, with values in PT groups restored to LD levels. Mettl3 mRNA expression did not change. These results suggest that testicular recovery induced by stimulatory photoperiod is relatively rapid, and that the methyltransferase complex may play a role during photostimulated testicular recrudescence.
Collapse
Affiliation(s)
- Melanie A Rader
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA 90840, USA
| | - Olga G Jaime
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA 90840, USA
| | - Victor O Abarca
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA 90840, USA
| | - Kelly A Young
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA 90840, USA.
| |
Collapse
|
22
|
Bila WC, Romano MCC, Dos Santos LL, da Silva VR, Capanema FD, Pfrimer K, Ferriolli E, Alves NMC, Campos CG, Carlos FM, Dos Santos MESM, Lamounier JA. Body fat, cardiovascular risk factors and polymorphism in the FTO gene: randomized clinical trial and different physical exercise for adolescents. J Pediatr (Rio J) 2023; 99:139-146. [PMID: 36030815 PMCID: PMC10031311 DOI: 10.1016/j.jped.2022.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 07/09/2022] [Accepted: 07/11/2022] [Indexed: 10/15/2022] Open
Abstract
OBJECTIVE To investigate the effects of different physical exercise programs and polymorphisms of the FTO (fat mass and obesity-associated gene) on body composition and cardiovascular risk factors in adolescents with overweight and obesity. METHODS A randomized, parallel, double-blind clinical trial consisting of the adolescent overweight from the state public network, in a simple representative random sample, who participated in an aerobic exercise or weight training intervention for 10 weeks. Anthropometry, body composition, biochemical markers, sexual maturation, and rs9939609 polymorphism in the FTO gene were assessed. 347 adolescents had their characterization of nutritional status. 72 individuals with overweight and obesity were invited to participate. 39 remained for the start of the program and were randomly allocated to both types of intervention. In the end, 26 subjects participated in the intervention programs, with 12 and 14 in the aerobic and weight training programs, respectively. RESULTS Heterozygous and homozygous bearers of risk allele A participating in the aerobic program showed improvements in glycemia (p = 0.002) and total cholesterol (p = 0.023) and a reduction in body fat mass (p = 0.041). The weight training program reduced glycemia in patients with the risk allele A (p = 0.027). Cameron's stage four sexual maturation participants were 2.1 times more likely to improve their body fat (CI = 1.31-3.39). CONCLUSION Aerobic exercises produced exclusively a significant decrease in fat mass and total cholesterol in patients with risk allele A. Distinct physical exercise programs may cause diverse changes in risk variables related to the health of adolescents.
Collapse
Affiliation(s)
- Wendell C Bila
- Universidade Federal de São João del Rei, Programa de Pós-Graduação em Ciências da Saúde, Divinópolis, MG, Brazil.
| | - Márcia C C Romano
- Universidade Federal de São João del Rei, Programa de Pós-Graduação em Enfermagem, Divinópolis, MG, Brazil
| | - Luciana L Dos Santos
- Universidade Federal de São João del Rei, Programa de Pós-Graduação em Ciências da Saúde, Divinópolis, MG, Brazil
| | - Valmin R da Silva
- Faculdade de Ciências da Santa Casa de Misericórdia de Vitória, Programa de Mestrado em Políticas Públicas e Desenvolvimento Local, Vitória, ES, Brazil
| | - Flávio D Capanema
- Fundação Hospitalar do Estado de Minas Gerais, Centro de Inovação Tecnológica e Proteção do Conhecimento, Belo Horizonte, MG, Brazil
| | - Karina Pfrimer
- Universidade de São Paulo, Faculdade de Medicina de Ribeirão Preto, Ribeirão Preto, SP, Brazil
| | - Eduardo Ferriolli
- Universidade de São Paulo, Faculdade de Medicina de Ribeirão Preto, Ribeirão Preto, SP, Brazil
| | - Natália M C Alves
- Universidade de São Paulo, Faculdade de Medicina de Ribeirão Preto, Ribeirão Preto, SP, Brazil
| | - Cezenário G Campos
- Universidade Federal de São João del Rei, Programa de Pós-Graduação em Ciências da Saúde, Divinópolis, MG, Brazil
| | - Fabiângelo M Carlos
- Universidade Federal de São João del Rei, Programa de Pós-Graduação em Enfermagem, Divinópolis, MG, Brazil
| | - Maria E S M Dos Santos
- Universidade Federal do Triângulo Mineiro, Departamento de Bioquímica, Farmacologia e Fisiologia/ICBN, Uberaba, MG, Brazil
| | - Joel A Lamounier
- Universidade Federal de São João del Rei, Departamento de Medicina, São João del Rei, MG, Brazil
| |
Collapse
|
23
|
Tsao CW, Aday AW, Almarzooq ZI, Anderson CAM, Arora P, Avery CL, Baker-Smith CM, Beaton AZ, Boehme AK, Buxton AE, Commodore-Mensah Y, Elkind MSV, Evenson KR, Eze-Nliam C, Fugar S, Generoso G, Heard DG, Hiremath S, Ho JE, Kalani R, Kazi DS, Ko D, Levine DA, Liu J, Ma J, Magnani JW, Michos ED, Mussolino ME, Navaneethan SD, Parikh NI, Poudel R, Rezk-Hanna M, Roth GA, Shah NS, St-Onge MP, Thacker EL, Virani SS, Voeks JH, Wang NY, Wong ND, Wong SS, Yaffe K, Martin SS. Heart Disease and Stroke Statistics-2023 Update: A Report From the American Heart Association. Circulation 2023; 147:e93-e621. [PMID: 36695182 DOI: 10.1161/cir.0000000000001123] [Citation(s) in RCA: 2306] [Impact Index Per Article: 1153.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND The American Heart Association, in conjunction with the National Institutes of Health, annually reports the most up-to-date statistics related to heart disease, stroke, and cardiovascular risk factors, including core health behaviors (smoking, physical activity, diet, and weight) and health factors (cholesterol, blood pressure, and glucose control) that contribute to cardiovascular health. The Statistical Update presents the latest data on a range of major clinical heart and circulatory disease conditions (including stroke, congenital heart disease, rhythm disorders, subclinical atherosclerosis, coronary heart disease, heart failure, valvular disease, venous disease, and peripheral artery disease) and the associated outcomes (including quality of care, procedures, and economic costs). METHODS The American Heart Association, through its Epidemiology and Prevention Statistics Committee, continuously monitors and evaluates sources of data on heart disease and stroke in the United States to provide the most current information available in the annual Statistical Update with review of published literature through the year before writing. The 2023 Statistical Update is the product of a full year's worth of effort in 2022 by dedicated volunteer clinicians and scientists, committed government professionals, and American Heart Association staff members. The American Heart Association strives to further understand and help heal health problems inflicted by structural racism, a public health crisis that can significantly damage physical and mental health and perpetuate disparities in access to health care, education, income, housing, and several other factors vital to healthy lives. This year's edition includes additional COVID-19 (coronavirus disease 2019) publications, as well as data on the monitoring and benefits of cardiovascular health in the population, with an enhanced focus on health equity across several key domains. RESULTS Each of the chapters in the Statistical Update focuses on a different topic related to heart disease and stroke statistics. CONCLUSIONS The Statistical Update represents a critical resource for the lay public, policymakers, media professionals, clinicians, health care administrators, researchers, health advocates, and others seeking the best available data on these factors and conditions.
Collapse
|
24
|
de Soysa AKH, Martins C, Langaas M, Grill V, Mostad IL. Exploring Dietary Intake in Adults with Severe Obesity and Associations with the FTO rs9939609 Genotypes. Curr Dev Nutr 2023; 7:100032. [PMID: 37180087 PMCID: PMC10111606 DOI: 10.1016/j.cdnut.2023.100032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 01/18/2023] [Accepted: 01/20/2023] [Indexed: 01/31/2023] Open
Abstract
Background Few have studied the associations between rs9939609 genotypes in the obesity candidate locus FTO and energy and nutrient intakes and meal frequencies in adults with severe obesity. We are unaware of studies that have assessed adherence to key dietary recommendations in this population, at least in Norway. Increased knowledge of genotype associations with dietary factors could improve personalized obesity therapy. Objectives The present study aimed to explore how the rs9939609 genotypes associate with dietary variables and adherence to key dietary recommendations in a sample of adults with severe obesity. Methods A cross-sectional observation study designed to have similar numbers of participants with genotypes TT, AT, and AA included 100 patients (70% women) with median (25th, 75th percentile) age 42 (32, 50) y and BMI 42.8 (39.5, 46.4) kg/m2. We assessed intakes of food groups, energy, and macro- and micronutrients from three 24-h dietary recalls and meal frequencies. Genotype associations were analyzed using regression analyses. Reported intakes were evaluated against national diet recommendations. Results Using a significance level of 0.01, we found no genotype associations with energy intake, energy density, adherence to recommendations, or meal frequency but tendencies of associations with energy adjusted protein intake (AA > AT, P = 0.037; AT > TT, P = 0.064), food groups milk and cream (AT > TT, P = 0.029), and Mixed dishes (AA > TT, P = 0.039). Few participants complied with recommendations for intakes of whole grains (21%), fruits and vegetables (11%), and fish (37%); however, 67% followed the recommendation to limit added sugar. Less than 20% had recommended intakes of vitamin D and folate. Conclusions In our patients with severe obesity, we found tendencies of associations between the FTO rs9939609 genotypes and diet but no significant associations at the 0.01 level and below. Few met key food-based diet recommendations, suggesting that the food habits in this population pose an increased risk of nutrient deficiencies. Curr Dev Nutr 2023;xx:xx.
Collapse
Affiliation(s)
- Ann Kristin H. de Soysa
- Department of Clinical Nutrition and Speech-Language Therapy, Clinic of Clinical Services, St. Olavs hospital - Trondheim University Hospital, Trondheim, Norway
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Catia Martins
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Center for Obesity Research and Innovation (ObeCe), Clinic of Surgery, St. Olavs Hospital–Trondheim University Hospital, Trondheim, Norway
- Department of Nutrition Sciences, the University of Alabama at Birmingham, Birmingham, AL, USA
| | - Mette Langaas
- Department of Mathematical Sciences, Faculty of Information Technology and Electrical Engineering, Norwegian University of Science and Technology, Trondheim, Norway
| | - Valdemar Grill
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Ingrid Løvold Mostad
- Department of Clinical Nutrition and Speech-Language Therapy, Clinic of Clinical Services, St. Olavs hospital - Trondheim University Hospital, Trondheim, Norway
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
25
|
Identification and Association of Single Nucleotide Polymorphisms of the FTO Gene with Indicators of Overweight and Obesity in a Young Mexican Population. Genes (Basel) 2023; 14:genes14010159. [PMID: 36672899 PMCID: PMC9858641 DOI: 10.3390/genes14010159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/29/2022] [Accepted: 01/04/2023] [Indexed: 01/10/2023] Open
Abstract
(1) Background: obesity is a global public health problem; various factors have been associated with this disease, and genetic factors play a very important role. Previous studies in multiple populations have associated a gene with fat mass and obesity (FTO). Thus, the present work aims to identify and determine associations between genetic variants of FTO with indicators of overweight and obesity in the Mexican population. (2) Methods: a total of 638 subjects were evaluated to compile data on body mass index (BMI), the percentage of body fat (%BF), the waist circumference (WC), the serum levels of triglycerides (TG), and food consumption. A total of 175 genetic variants in the FTO gene were sampled by a microarray in the evaluated population, followed by association statistical analyses and comparisons of means. (3) Results: a total of 34 genetic variants were associated with any of the 6 indicators of overweight and obesity, but only 15 showed mean differences using the recessive model after the Bonferroni correction. The present study shows a wide evaluation of FTO genetic variants associated with a classic indicator of overweight and obesity, which highlights the importance of genetic analyses in the study of obesity.
Collapse
|
26
|
Bourbour F, Pourtaheri A, Abbasi K, Hasanpour Ardekanizadeh N, Gholamalizadeh M, Hajipour A, Abdollahi S, Bagheri SE, Ahmadzadeh M, Doaei S, Haghighian A. Interactions dietary components with expression level of breast cancer-related genes. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2022. [DOI: 10.1186/s43042-022-00375-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Abstract
Background
Dietary components can influence the effects of genetic background in breast cancer (BC). This review study aimed to investigate the effect of dietary components on the expression level of BC-related genes.
Methods
In this narrative review, Embase, PubMed, PsycInfo, and the Cochrane databases were used to collect the related papers with interactions of BC, genetics, and dietary intake. Appropriate keywords such as BC, gene expression, mutation, nutrient, and diet (alone and together) were applied for data collection.
Results
The association of BC with some genes including the BC1 gene (BRCA1), the human epidermal growth factor receptor 2 (HER2), and the fat mass and obesity-associated (FTO) gene can be affected by dietary components. Moderate B12 supplementation may be protective against BC in people with the inherited mutation of BRCA. The olive oil may have a protective effect against BC through several mechanisms such as suppressing HER-2 expression. Furthermore, high glycemic index foods may increase the risk of BC by the activation of the phosphoinositide 3-kinase/protein kinase B (PI3K/AKT) pathway and the up-regulation of FTO gene expression.
Conclusion
There are interactions between BC, BC-related genes, and dietary intake. Dietary components such as macronutrients, micronutrients, and phytochemicals may regulate the expression level of BC-related genes. Further longitudinal studies are needed to confirm the associations between BC-related genes and diet and to discover the underlying mechanisms.
Collapse
|
27
|
Liu T, Xu Y, Yi CX, Tong Q, Cai D. The hypothalamus for whole-body physiology: from metabolism to aging. Protein Cell 2022; 13:394-421. [PMID: 33826123 PMCID: PMC9095790 DOI: 10.1007/s13238-021-00834-x] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 03/01/2021] [Indexed: 01/05/2023] Open
Abstract
Obesity and aging are two important epidemic factors for metabolic syndrome and many other health issues, which contribute to devastating diseases such as cardiovascular diseases, stroke and cancers. The brain plays a central role in controlling metabolic physiology in that it integrates information from other metabolic organs, sends regulatory projections and orchestrates the whole-body function. Emerging studies suggest that brain dysfunction in sensing various internal cues or processing external cues may have profound effects on metabolic and other physiological functions. This review highlights brain dysfunction linked to genetic mutations, sex, brain inflammation, microbiota, stress as causes for whole-body pathophysiology, arguing brain dysfunction as a root cause for the epidemic of aging and obesity-related disorders. We also speculate key issues that need to be addressed on how to reveal relevant brain dysfunction that underlines the development of these disorders and diseases in order to develop new treatment strategies against these health problems.
Collapse
Affiliation(s)
- Tiemin Liu
- grid.8547.e0000 0001 0125 2443State Key Laboratory of Genetic Engineering, Department of Endocrinology and Metabolism, Institute of Metabolism and Integrative Biology, Human Phenome Institute, and Collaborative Innovation Center for Genetics and Development, Zhongshan Hospital, School of Life Sciences, Fudan University, Shanghai, 200438 China
| | - Yong Xu
- grid.39382.330000 0001 2160 926XChildren’s Nutrition Research Center, Department of Pediatrics, Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
| | - Chun-Xia Yi
- grid.7177.60000000084992262Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, Amsterdam Gastroenterology Endocrinology Metabolism, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, Netherlands
| | - Qingchun Tong
- grid.453726.10000 0004 5906 7293Brown Foundation Institute of Molecular Medicine, Department of Neurobiology and Anatomy, University of Texas McGovern Medical School, Graduate Program in Neuroscience of MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030 USA
| | - Dongsheng Cai
- grid.251993.50000000121791997Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, NY 10461 USA
| |
Collapse
|
28
|
Chen LS, Zhang M, Chen P, Xiong XF, Liu PQ, Wang HB, Wang JJ, Shen J. The m 6A demethylase FTO promotes the osteogenesis of mesenchymal stem cells by downregulating PPARG. Acta Pharmacol Sin 2022; 43:1311-1323. [PMID: 34462564 PMCID: PMC9061799 DOI: 10.1038/s41401-021-00756-8] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 08/02/2021] [Indexed: 12/15/2022]
Abstract
N6-methyladenosine (m6A) is the most abundant posttranscriptional methylation modification that occurs in mRNA and modulates the fine-tuning of various biological processes in mammalian development and human diseases. In this study we investigated the role of m6A modification in the osteogenesis of mesenchymal stem cells (MSCs), and the possible mechanisms by which m6A modification regulated the processes of osteoporosis and bone necrosis. We performed systematic analysis of the differential gene signatures in patients with osteoporosis and bone necrosis and conducted m6A-RNA immunoprecipitation (m6A-RIP) sequencing to identify the potential regulatory genes involved in osteogenesis. We showed that fat mass and obesity (FTO), a primary m6A demethylase, was significantly downregulated in patients with osteoporosis and osteonecrosis. During the differentiation of human MSCs into osteoblasts, FTO was markedly upregulated. Both depletion of FTO and application of the FTO inhibitor FB23 or FB23-2 impaired osteogenic differentiation of human MSCs. Knockout of FTO in mice resulted in decreased bone mineral density and impaired bone formation. PPARG, a biomarker for osteoporosis, was identified as a critical downstream target of FTO. We further revealed that FTO mediated m6A demethylation in the 3'UTR of PPARG mRNA, and reduced PPARG mRNA stability in an YTHDF1-dependent manner. Overexpression of PPARG alleviated FTO-mediated osteogenic differentiation of MSCs, whereas knockdown of PPARG promoted FTO-induced expression of the osteoblast biomarkers ALPL and OPN during osteogenic differentiation. Taken together, this study demonstrates the functional significance of the FTO-PPARG axis in promoting the osteogenesis of human MSCs and sheds light on the role of m6A modification in mediating osteoporosis and osteonecrosis.
Collapse
Affiliation(s)
- Liu-shan Chen
- grid.411847.f0000 0004 1804 4300Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, 510006 China
| | - Meng Zhang
- grid.414011.10000 0004 1808 090XDepartment of Orthopedics, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, People’s Hospital of Henan University, Zhengzhou, 450003 China
| | - Peng Chen
- grid.411866.c0000 0000 8848 7685The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405 China
| | - Xiao-feng Xiong
- grid.12981.330000 0001 2360 039XDepartment of Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006 China
| | - Pei-qing Liu
- grid.12981.330000 0001 2360 039XDepartment of Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006 China
| | - Hai-bin Wang
- grid.411866.c0000 0000 8848 7685The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405 China
| | - Jun-jian Wang
- grid.12981.330000 0001 2360 039XDepartment of Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006 China
| | - Juan Shen
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| |
Collapse
|
29
|
Hall KD, Farooqi IS, Friedman JM, Klein S, Loos RJF, Mangelsdorf DJ, O'Rahilly S, Ravussin E, Redman LM, Ryan DH, Speakman JR, Tobias DK. The energy balance model of obesity: beyond calories in, calories out. Am J Clin Nutr 2022; 115:1243-1254. [PMID: 35134825 PMCID: PMC9071483 DOI: 10.1093/ajcn/nqac031] [Citation(s) in RCA: 154] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 02/02/2022] [Indexed: 02/06/2023] Open
Abstract
A recent Perspective article described the "carbohydrate-insulin model (CIM)" of obesity, asserting that it "better reflects knowledge on the biology of weight control" as compared with what was described as the "dominant energy balance model (EBM)," which fails to consider "biological mechanisms that promote weight gain." Unfortunately, the Perspective conflated and confused the principle of energy balance, a law of physics that is agnostic as to obesity mechanisms, with the EBM as a theoretical model of obesity that is firmly based on biology. In doing so, the authors presented a false choice between the CIM and a caricature of the EBM that does not reflect modern obesity science. Here, we present a more accurate description of the EBM where the brain is the primary organ responsible for body weight regulation operating mainly below our conscious awareness via complex endocrine, metabolic, and nervous system signals to control food intake in response to the body's dynamic energy needs as well as environmental influences. We also describe the recent history of the CIM and show how the latest "most comprehensive formulation" abandons a formerly central feature that required fat accumulation in adipose tissue to be the primary driver of positive energy balance. As such, the new CIM can be considered a special case of the more comprehensive EBM but with a narrower focus on diets high in glycemic load as the primary factor responsible for common obesity. We review data from a wide variety of studies that address the validity of each model and demonstrate that the EBM is a more robust theory of obesity than the CIM.
Collapse
Affiliation(s)
- Kevin D Hall
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health
| | - I Sadaf Farooqi
- Wellcome-MRC Institute of Metabolic Science, University of Cambridge
| | | | - Samuel Klein
- Washington University School of Medicine in St Louis
| | - Ruth J F Loos
- Washington University School of Medicine in St Louis.,Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen
| | | | - Stephen O'Rahilly
- Wellcome-MRC Institute of Metabolic Science, University of Cambridge
| | | | | | | | - John R Speakman
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzen, China, and the University of Aberdeen, Aberdeen, United Kingdom
| | | |
Collapse
|
30
|
Ortega-Contreras B, Armella A, Appel J, Mennickent D, Araya J, González M, Castro E, Obregón AM, Lamperti L, Gutiérrez J, Guzmán-Gutiérrez E. Pathophysiological Role of Genetic Factors Associated With Gestational Diabetes Mellitus. Front Physiol 2022; 13:769924. [PMID: 35450164 PMCID: PMC9016477 DOI: 10.3389/fphys.2022.769924] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 03/15/2022] [Indexed: 11/13/2022] Open
Abstract
Gestational Diabetes Mellitus (GDM) is a highly prevalent maternal pathology characterized by maternal glucose intolerance during pregnancy that is, associated with severe complications for both mother and offspring. Several risk factors have been related to GDM; one of the most important among them is genetic predisposition. Numerous single nucleotide polymorphisms (SNPs) in genes that act at different levels on various tissues, could cause changes in the expression levels and activity of proteins, which result in glucose and insulin metabolism dysfunction. In this review, we describe various SNPs; which according to literature, increase the risk of developing GDM. These SNPs include: (1) those associated with transcription factors that regulate insulin production and excretion, such as rs7903146 (TCF7L2) and rs5015480 (HHEX); (2) others that cause a decrease in protective hormones against insulin resistance such as rs2241766 (ADIPOQ) and rs6257 (SHBG); (3) SNPs that cause modifications in membrane proteins, generating dysfunction in insulin signaling or cell transport in the case of rs5443 (GNB3) and rs2237892 (KCNQ1); (4) those associated with enzymes such as rs225014 (DIO2) and rs9939609 (FTO) which cause an impaired metabolism, resulting in an insulin resistance state; and (5) other polymorphisms, those are associated with growth factors such as rs2146323 (VEGFA) and rs755622 (MIF) which could cause changes in the expression levels of these proteins, producing endothelial dysfunction and an increase of pro-inflammatory cytokines, characteristic on GDM. While the pathophysiological mechanism is unclear, this review describes various potential effects of these polymorphisms on the predisposition to develop GDM.
Collapse
Affiliation(s)
- B. Ortega-Contreras
- Pregnancy Diseases Laboratory, Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, Universidad de Concepción, Concepción, Chile
| | - A. Armella
- Pregnancy Diseases Laboratory, Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, Universidad de Concepción, Concepción, Chile
| | - J. Appel
- Pregnancy Diseases Laboratory, Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, Universidad de Concepción, Concepción, Chile
| | - D. Mennickent
- Pregnancy Diseases Laboratory, Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, Universidad de Concepción, Concepción, Chile
- Department of Instrumental Analysis, Faculty of Pharmacy, Universidad de Concepción, Concepción, Chile
| | - J. Araya
- Department of Instrumental Analysis, Faculty of Pharmacy, Universidad de Concepción, Concepción, Chile
| | - M. González
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universidad de Concepción, Concepción, Chile
| | - E. Castro
- Departamento de Obstetricia y Puericultura, Facultad de Ciencias de la Salud, Universidad de Atacama, Copiapó, Chile
| | - A. M. Obregón
- Faculty of Health Care, Universidad San Sebastián, Concepción, Chile
| | - L. Lamperti
- Pregnancy Diseases Laboratory, Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, Universidad de Concepción, Concepción, Chile
| | - J. Gutiérrez
- Faculty of Health Sciences, Universidad San Sebastián, Santiago,Chile
| | - E. Guzmán-Gutiérrez
- Pregnancy Diseases Laboratory, Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, Universidad de Concepción, Concepción, Chile
- *Correspondence: E. Guzmán-Gutiérrez,
| |
Collapse
|
31
|
Novais PFS, Crisp AH, Leandro-Merhi VA, Verlengia R, Rasera I, Oliveira MRMD. Lack of Association Between 11 Gene Polymorphisms on Weight Loss One Year After Roux-en-Y Gastric Bypass Surgery in Women. J Hum Nutr Diet 2022; 35:731-738. [PMID: 35239993 DOI: 10.1111/jhn.13000] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 02/28/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Although effective, the impact of bariatric surgery on weight loss is variable, and little is known about the influence of single nucleotide polymorphisms (SNPs). This study investigated the association of eleven SNPs related to obesity with weight loss one year after Roux-Y gastric bypass (RYGB) surgery in female patients. METHODS This prospective study included 351 women with obesity. The genotypes for eleven SNPs (GHRL - rs26802; GHSR - rs572169; LEP - rs7799039; LEPR - rs1137101; 5HT2C - rs3813929; UCP2 - rs659366; UCP3 - rs1800849; SH2B1 - rs7498665; TAS1R2 - rs35874116; TAS1R2 - rs9701796; FTO - rs9939609) were determined using real-time polymerase chain reaction (PCR) and TaqMan assays. Anthropometric measurements were performed before and one year after RYGB surgery. To evaluate the factors that influenced the proportion of weight loss 1 year after surgery, beta regression analysis was used. The models were estimated using the SAS software GLIMMIX procedure. The level of significance adopted for the statistical tests was 5%. RESULTS The average percentage of total body weight loss in one year was 64.4 ± 5.8% and the median was 65.0%. In assessing the proportion of weight loss in one year after surgery, using univariate analysis (beta regression), no single nucleotide polymorphisms (SNPs) influenced weight loss. And in the multiple analysis, with stepwise process of variable selection, no variable was significant to compose the multiple model. CONCLUSION The 11 SNPs investigated did not influence weight loss one year after RYGB surgery in female patients. This result indicates that individual behaviors and other factors might better contribute to the magnitude of loss weight loss in a short period after bariatric surgery. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
| | - Alex Harley Crisp
- Human Performance Laboratory, Universidade Metodista de Piracicaba, Piracicaba-SP, Brazil
| | | | - Rozangela Verlengia
- Faculty of Health Sciences, Universidade Metodista de Piracicaba, Piracicaba-SP, Brazil
| | - Irineu Rasera
- Clínica Bariátrica de Piracicaba, Piracicaba-SP, Brazil
| | | |
Collapse
|
32
|
Tsao CW, Aday AW, Almarzooq ZI, Alonso A, Beaton AZ, Bittencourt MS, Boehme AK, Buxton AE, Carson AP, Commodore-Mensah Y, Elkind MSV, Evenson KR, Eze-Nliam C, Ferguson JF, Generoso G, Ho JE, Kalani R, Khan SS, Kissela BM, Knutson KL, Levine DA, Lewis TT, Liu J, Loop MS, Ma J, Mussolino ME, Navaneethan SD, Perak AM, Poudel R, Rezk-Hanna M, Roth GA, Schroeder EB, Shah SH, Thacker EL, VanWagner LB, Virani SS, Voecks JH, Wang NY, Yaffe K, Martin SS. Heart Disease and Stroke Statistics-2022 Update: A Report From the American Heart Association. Circulation 2022; 145:e153-e639. [PMID: 35078371 DOI: 10.1161/cir.0000000000001052] [Citation(s) in RCA: 3181] [Impact Index Per Article: 1060.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND The American Heart Association, in conjunction with the National Institutes of Health, annually reports the most up-to-date statistics related to heart disease, stroke, and cardiovascular risk factors, including core health behaviors (smoking, physical activity, diet, and weight) and health factors (cholesterol, blood pressure, and glucose control) that contribute to cardiovascular health. The Statistical Update presents the latest data on a range of major clinical heart and circulatory disease conditions (including stroke, congenital heart disease, rhythm disorders, subclinical atherosclerosis, coronary heart disease, heart failure, valvular disease, venous disease, and peripheral artery disease) and the associated outcomes (including quality of care, procedures, and economic costs). METHODS The American Heart Association, through its Statistics Committee, continuously monitors and evaluates sources of data on heart disease and stroke in the United States to provide the most current information available in the annual Statistical Update. The 2022 Statistical Update is the product of a full year's worth of effort by dedicated volunteer clinicians and scientists, committed government professionals, and American Heart Association staff members. This year's edition includes data on the monitoring and benefits of cardiovascular health in the population and an enhanced focus on social determinants of health, adverse pregnancy outcomes, vascular contributions to brain health, and the global burden of cardiovascular disease and healthy life expectancy. RESULTS Each of the chapters in the Statistical Update focuses on a different topic related to heart disease and stroke statistics. CONCLUSIONS The Statistical Update represents a critical resource for the lay public, policymakers, media professionals, clinicians, health care administrators, researchers, health advocates, and others seeking the best available data on these factors and conditions.
Collapse
|
33
|
Kanders SH, Nilsson KW, Åslund C. Breastfeeding moderates the relationship between fat mass and obesity-associated gene rs9939609 and body mass index among adolescents. Obes Sci Pract 2022; 8:66-76. [PMID: 35127123 PMCID: PMC8804930 DOI: 10.1002/osp4.546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 07/02/2021] [Accepted: 07/22/2021] [Indexed: 11/29/2022] Open
Abstract
OBJECTIVE Breastfeeding, which is important for early growth, is a possible moderator of genetic influence, such as the effect of the fat mass and obesity-associated gene (FTO) on body mass index (BMI). The aim of this study was to assess the moderating effect of breastfeeding duration on the relationship between FTO rs9939609 and BMI in a Caucasian sample. METHODS Adolescents born in 1997 and in 1999, who were living in the Swedish county Västmanland in 2012, were invited to participate in the Survey of Adolescent Life in Västmanland. The adolescents and their parents completed self-reported questionnaires in 2012, 2015, and 2018. Genotyping of rs9939609 T > A polymorphism was conducted from saliva DNA samples. Interaction effects of parental reported breastfeeding duration in months, including regions of significance, on the relationship between rs9939609 and BMI plus overweight were assessed. RESULTS Considering physical activity levels, parental reported breastfeeding duration was a moderator of the relationship between rs9939609 and BMI for the younger (regions of significance = <1.6 and >28.1 months) and older adolescents (region of significance = >19.9 months), but not for the young adults. Plots of the association between breastfeeding duration and BMI showed higher BMI for AA with short breastfeeding, but lower BMI with longer breastfeeding than AT and TT. Longer breastfeeding lowered the odds for overweight among the younger adolescents, especially among AA individuals. CONCLUSION Rs9939609 AA individuals were more susceptible than AT and TT individuals to both short and long breastfeeding durations, which is consistent with the differential susceptibility hypothesis. FTO rs9939609 AA might be a plasticity variant with differential susceptibility to environmental influences. Breastfeeding duration may be one of many factors that affect the relationship between rs9939609 and BMI.
Collapse
Affiliation(s)
| | - Kent W. Nilsson
- Centre for Clinical ResearchRegion VästmanlandUppsala UniversityCounty HospitalVästeråsSweden
- School of Health, Care and Social WelfareMälardalen UniversityVästeråsSweden
| | - Cecilia Åslund
- Centre for Clinical ResearchRegion VästmanlandUppsala UniversityCounty HospitalVästeråsSweden
- Department of Public Health and Caring SciencesUppsala UniversityUppsalaSweden
| |
Collapse
|
34
|
Yang Z, Yu GL, Zhu X, Peng TH, Lv YC. Critical roles of FTO-mediated mRNA m6A demethylation in regulating adipogenesis and lipid metabolism: Implications in lipid metabolic disorders. Genes Dis 2022; 9:51-61. [PMID: 35005107 PMCID: PMC8720706 DOI: 10.1016/j.gendis.2021.01.005] [Citation(s) in RCA: 98] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 12/19/2020] [Accepted: 01/20/2021] [Indexed: 02/06/2023] Open
Abstract
The goal this review is to clarify the effects of the fat mass and obesity-associated protein (FTO) in lipid metabolism regulation and related underlying mechanisms through the FTO-mediated demethylation of m6A modification. FTO catalyzes the demethylation of m6A to alter the processing, maturation and translation of the mRNAs of lipid-related genes. FTO overexpression in the liver promotes lipogenesis and lipid droplet (LD) enlargement and suppresses CPT-1–mediated fatty acid oxidation via the SREBP1c pathway, promoting excessive lipid storage and nonalcoholic fatty liver diseases (NAFLD). FTO enhances preadipocyte differentiation through the C/EBPβ pathway, and facilitates adipogenesis and fat deposition by altering the alternative splicing of RUNX1T1, the expression of PPARγ and ANGPTL4, and the phosphorylation of PLIN1, whereas it inhibits lipolysis by inhibiting IRX3 expression and the leptin pathway, causing the occurrence and development of obesity. Suppression of the PPARβ/δ and AMPK pathways by FTO-mediated m6A demethylation damages lipid utilization in skeletal muscles, leading to the occurrence of diabetic hyperlipidemia. m6A demethylation by FTO inhibits macrophage lipid influx by downregulating PPARγ protein expression and accelerates cholesterol efflux by phosphorylating AMPK, thereby impeding foam cell formation and atherosclerosis development. In summary, FTO-mediated m6A demethylation modulates the expression of lipid-related genes to regulate lipid metabolism and lipid disorder diseases.
Collapse
Affiliation(s)
- Zhou Yang
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, PR China
| | - Guang-Li Yu
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, PR China
| | - Xiao Zhu
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi 541199, PR China
| | - Tian-Hong Peng
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, PR China
| | - Yun-Cheng Lv
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, PR China.,Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi 541199, PR China
| |
Collapse
|
35
|
Association of ADIPOQ-rs2241766 and FTO-rs9939609 genetic variants with body mass index trajectory in women of reproductive age over 6 years of follow-up: the PREDI study. Eur J Clin Nutr 2022; 76:159-172. [PMID: 33850313 DOI: 10.1038/s41430-021-00911-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 03/12/2021] [Accepted: 03/26/2021] [Indexed: 02/02/2023]
Abstract
BACKGROUND Interindividual variations in body mass index (BMI) can be partially explained by genetic differences. We aimed to examine the association of the ADIPOQ-rs2241766, LEP-rs7799039 and FTO-rs9939609 genetic variants with BMI trajectory in women of reproductive age over 6 years of follow-up. METHODS This was a prospective study that used data from 435 women of the PREDI Study conducted in Brazil. Socioeconomic, biological and anthropometric data were collected at four time points: 2012 (baseline) in the maternity hospital, and 2013-14, 2016-17 and 2018 (1st, 2nd and 3rd follow-ups) at the participant's home. Genotyping was performed by PCR-RFLP. Linear mixed-effect and Poisson regression models were used to address the association of ADIPOQ, LEP and FTO genotypes with BMI and overweight/obesity status. RESULTS Women carrying the risk allele (TA or AA) of the FTO-rs9939609 genetic variant had a 1.16 kg/m2 higher BMI over the follow-up period than those carrying the wild-type genotype (TT), even when adjusted for potential confounders (95% CI: 0.23-2.10, p = 0.015). The risk of obesity associated with the FTO-TA or AA genotype decreased over the years, demonstrating an influence of time on its trajectory (IRR = 0.99, 95% CI: 0.98-0.99, p = 0.016). There was no variation in BMI trajectories for the ADIPOQ-rs2241766, LEP-rs7799039 or FTO-rs9939609 genetic variant. CONCLUSIONS The results of this study suggest that monitoring women of reproductive age with ADIPOQ-rs2241766 TG/GG or FTO-rs9939609 TA/AA genotypes may be an important strategy to reduce maternal excess body weight and, consequently, the long-term public health burden of obesity.
Collapse
|
36
|
FTO and PLAG1 Genes Expression and FTO Methylation Predict Changes in Circulating Levels of Adipokines and Gastrointestinal Peptides in Children. Nutrients 2021; 13:nu13103585. [PMID: 34684585 PMCID: PMC8538237 DOI: 10.3390/nu13103585] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/03/2021] [Accepted: 10/06/2021] [Indexed: 12/19/2022] Open
Abstract
Adipokines and gastrointestinal tract hormones are important metabolic parameters, and both epigenetic factors and differential gene expression patterns may be associated with the alterations in their concentrations in children. The function of the FTO gene (FTO alpha-ketoglutarate dependent dioxygenase) in the regulation of the global metabolic rate is well described, whereas the influence of protooncogene PLAG1 (PLAG1 zinc finger) is still not fully understood. A cross-sectional study on a group of 26 children with various BMI values (15.3–41.7; median 28) was carried out. The aim was to evaluate the dependencies between the level of methylation and expression of aforementioned genes with the concentration of selected gastrointestinal tract hormones and adipokines in children. Expression and methylation were measured in peripheral blood mononuclear DNA by a microarray technique and a restriction enzyme method, respectively. All peptide concentrations were determined using the enzyme immunoassay method. The expression level of both FTO and PLAG1 genes was statistically significantly related to the concentration of adipokines: negatively for apelin and leptin receptor, and positively for leptin. Furthermore, both FTO methylation and expression negatively correlated with the concentration of resistin and visfatin. Cholecystokinin was negatively correlated, whereas fibroblast growth factor 21 positively correlated with methylation and expression of the FTO gene, while FTO and PLAG1 expression was negatively associated with the level of cholecystokinin and glucagon-like peptide-1. The PLAG1 gene expression predicts an increase in leptin and decrease in ghrelin levels. Our results indicate that the FTO gene correlates with the concentration of hormones produced by the adipose tissue and gastrointestinal tract, and PLAG1 gene may be involved in adiposity pathogenesis. However, the exact molecular mechanisms still need to be clarified.
Collapse
|
37
|
Wu PF, Han QQ, Chen FF, Shen TT, Li YH, Cao Y, Chen JG, Wang F. Erasing m 6A-dependent transcription signature of stress-sensitive genes triggers antidepressant actions. Neurobiol Stress 2021; 15:100390. [PMID: 34527794 PMCID: PMC8430387 DOI: 10.1016/j.ynstr.2021.100390] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 08/08/2021] [Accepted: 09/03/2021] [Indexed: 11/29/2022] Open
Abstract
Emerging evidence has shown that stress responsivity and psychiatric diseases are associated with alterations in N6-methyladenosine (m6A) mRNA epigenetic modifications. Fat mass and obesity-associated protein (FTO) is an m6A demethylase that has been linked to increased body mass and obesity. Here, we show that tricyclic antidepressants (TCAs) with weight-gain side effects, such as imipramine and amitriptyline, directly increased FTO expression and activated its epigenetic function in the ventral tegmental area (VTA). VTA-specific genetic disruption of FTO increased stress vulnerability and abolished the antidepressant activity of TCAs, whereas erasing m6A modification in the VTA by FTO overexpression or cycloleucine led to significant antidepressant activity. Mechanistically, both transcriptome sequencing and quantitative PCR revealed that overexpression of FTO in the VTA decreased the transcription of stress-related neuropeptides, such as cocaine- and amphetamine-regulated transcript peptide and urocortin, in the social defeat model, which was mimicked by imipramine, suggesting an m6A-dependent transcription mechanism of stress-related neuropeptides may underlie the responses to antidepressant. Collectively, our results demonstrate that inhibiting m6A-dependent transcription of stress-related genes may work as a novel antidepressant strategy and highlight a previously unrecognized activator of FTO-dependent epigenetic function that may be used for the treatment of other neurological diseases. TCAs erase m6A epigenetic modification by activating FTO. FTO mediates the antidepressant activity of TCAs. FTO in the VTA confers stress resistance. FTO in the VTA limits m6A-dependent transcription of stress-sensitive genes.
Collapse
Affiliation(s)
- Peng-Fei Wu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan City, Hubei, 430030, China.,The Research Center for Depression, Tongji Medical College, Huazhong University of Science, 430030, Wuhan, China.,Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan City, Hubei, 430030, China.,The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan City, Hubei, 430030, China.,Laboratory of Neuropsychiatric Diseases, The Institute of Brain Research, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qian-Qian Han
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan City, Hubei, 430030, China.,The Research Center for Depression, Tongji Medical College, Huazhong University of Science, 430030, Wuhan, China
| | - Fu-Feng Chen
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan City, Hubei, 430030, China.,The Research Center for Depression, Tongji Medical College, Huazhong University of Science, 430030, Wuhan, China
| | - Tian-Tian Shen
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan City, Hubei, 430030, China.,The Research Center for Depression, Tongji Medical College, Huazhong University of Science, 430030, Wuhan, China
| | - Yi-Heng Li
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan City, Hubei, 430030, China.,The Research Center for Depression, Tongji Medical College, Huazhong University of Science, 430030, Wuhan, China
| | - Yu Cao
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan City, Hubei, 430030, China.,The Research Center for Depression, Tongji Medical College, Huazhong University of Science, 430030, Wuhan, China
| | - Jian-Guo Chen
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan City, Hubei, 430030, China.,The Research Center for Depression, Tongji Medical College, Huazhong University of Science, 430030, Wuhan, China.,Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan City, Hubei, 430030, China.,The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan City, Hubei, 430030, China.,Laboratory of Neuropsychiatric Diseases, The Institute of Brain Research, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Fang Wang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan City, Hubei, 430030, China.,The Research Center for Depression, Tongji Medical College, Huazhong University of Science, 430030, Wuhan, China.,Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan City, Hubei, 430030, China.,The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan City, Hubei, 430030, China.,Laboratory of Neuropsychiatric Diseases, The Institute of Brain Research, Huazhong University of Science and Technology, Wuhan, 430030, China
| |
Collapse
|
38
|
Common and Unique Genetic Background between Attention-Deficit/Hyperactivity Disorder and Excessive Body Weight. Genes (Basel) 2021; 12:genes12091407. [PMID: 34573389 PMCID: PMC8464917 DOI: 10.3390/genes12091407] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/08/2021] [Accepted: 09/09/2021] [Indexed: 02/07/2023] Open
Abstract
Comorbidity studies show that children with ADHD have a higher risk of being overweight and obese than healthy children. This study aimed to assess the genetic alternations that differ between and are shared by ADHD and excessive body weight (EBW). The sample consisted of 743 Polish children aged between 6 and 17 years. We analyzed a unique set of genes and polymorphisms selected for ADHD and/or obesity based on gene prioritization tools. Polymorphisms in the KCNIP1, SLC1A3, MTHFR, ADRA2A, and SLC6A2 genes proved to be associated with the risk of ADHD in the studied population. The COMT gene polymorphism was one that specifically increased the risk of EBW in the ADHD group. Using the whole-exome sequencing technique, we have shown that the ADHD group contains rare and protein-truncating variants in the FBXL17, DBH, MTHFR, PCDH7, RSPH3, SPTBN1, and TNRC6C genes. In turn, variants in the ADRA2A, DYNC1H1, MAP1A, SEMA6D, and ZNF536 genes were specific for ADHD with EBW. In this way, we confirmed, at the molecular level, the existence of genes specifically predisposing to EBW in ADHD patients, which are associated with the biological pathways involved in the regulation of the reward system, intestinal microbiome, and muscle metabolism.
Collapse
|
39
|
|
40
|
Gao H, Cheng X, Chen J, Ji C, Guo H, Qu W, Dong X, Chen Y, Ma L, Shu Q, Li X. Fto-modulated lipid niche regulates adult neurogenesis through modulating adenosine metabolism. Hum Mol Genet 2021; 29:2775-2787. [PMID: 32766784 DOI: 10.1093/hmg/ddaa171] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/01/2020] [Accepted: 07/01/2020] [Indexed: 12/16/2022] Open
Abstract
Adult neurogenesis is regulated by diverse factors including the local environment, i.e. the neurogenic niche. However, whether the lipid in the brain regulates adult neurogenesis and related mechanisms remains largely unknown. In the present study, we found that lipid accumulates in the brain during postnatal neuronal development. Conditional knockout of Fto (cKO) in lipid not only reduced the level of lipid in the brain but also impaired the learning and memory of mice. In addition, Fto deficiency in lipid did not affect the proliferation of adult neural stem cells (aNSCs), but it did inhibit adult neurogenesis by inducing cell apoptosis. Mechanistically, specific deleting Fto in lipid altered gene expression and increased adenosine secretion of adipocytes. The treatment of adenosine promoted the apoptosis of newborn neurons. As a whole, these results reveal the important function of the lipid niche and its associated mechanism in regulating adult neurogenesis.
Collapse
Affiliation(s)
- Hui Gao
- The Children's Hospital, School of Medicine, Zhejiang University, Hangzhou 310052, China.,The Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou 310029, China.,National Clinical Research Center for Child Health, Hangzhou 310052, China
| | - Xuejun Cheng
- The Children's Hospital, School of Medicine, Zhejiang University, Hangzhou 310052, China.,National Clinical Research Center for Child Health, Hangzhou 310052, China
| | - Junchen Chen
- The Children's Hospital, School of Medicine, Zhejiang University, Hangzhou 310052, China.,The Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou 310029, China
| | - Chai Ji
- The Children's Hospital, School of Medicine, Zhejiang University, Hangzhou 310052, China.,National Clinical Research Center for Child Health, Hangzhou 310052, China
| | - Hongfeng Guo
- The Children's Hospital, School of Medicine, Zhejiang University, Hangzhou 310052, China.,The Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou 310029, China
| | - Wenzheng Qu
- The Children's Hospital, School of Medicine, Zhejiang University, Hangzhou 310052, China.,National Clinical Research Center for Child Health, Hangzhou 310052, China
| | - Xiaoxue Dong
- The Children's Hospital, School of Medicine, Zhejiang University, Hangzhou 310052, China.,National Clinical Research Center for Child Health, Hangzhou 310052, China
| | - Yingyan Chen
- The Children's Hospital, School of Medicine, Zhejiang University, Hangzhou 310052, China.,The Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou 310029, China
| | - Linghan Ma
- The Children's Hospital, School of Medicine, Zhejiang University, Hangzhou 310052, China.,The Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou 310029, China
| | - Qiang Shu
- The Children's Hospital, School of Medicine, Zhejiang University, Hangzhou 310052, China.,National Clinical Research Center for Child Health, Hangzhou 310052, China
| | - Xuekun Li
- The Children's Hospital, School of Medicine, Zhejiang University, Hangzhou 310052, China.,The Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou 310029, China.,National Clinical Research Center for Child Health, Hangzhou 310052, China
| |
Collapse
|
41
|
Gholamalizadeh M, Doaei S, Mokhtari Z, Jalili V, Bourbour F, Omidi S, Ebrahimi K, Kalantari N, Abdi S, Azizi Tabesh G, Naimi Joubani M, Roohbakhsh E, Mosavi Jarrahi SA. Association of serum 25-OH-vitamin D level with FTO and IRX3 genes expression in obese and overweight boys with different FTO rs9930506 genotypes. J Transl Med 2021; 19:350. [PMID: 34399781 PMCID: PMC8365950 DOI: 10.1186/s12967-021-03029-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 08/07/2021] [Indexed: 11/10/2022] Open
Abstract
Background The roles of FTO gene and the level of serum 25-OH-vitamin D in obesity are frequently reported. This study aimed to investigate the interactions of serum 25-OH-vitamin D level, FTO and IRX3 genes expression, and FTO genotype in obese and overweight boys. Methods This study was carried out on the 120 male adolescents with overweight in Tehran, Iran. Blood samples were collected from the participants in order to evaluate the serum level of 25-OH-vitamin D, the expression level of FTO and IRX3 genes, and FTO genotype for rs9930506 at baseline and after 18 weeks of the study. Results In general, no significant association was found between serum 25-OH-vitamin D level and IRX3 and FTO genes expression. The results of linear regression on the relationship between 25-OH-vitamin D serum level and FTO and IRX3 genes expression based on FTO genotypes for rs9930506 indicated that in AA/AG genotype carriers, serum 25-OH-vitamin D level was positively associated with FTO gene expression (B = 0.07, p = 0.02) and inversely associated with IRX3 gene expression (B = − 0.07, p = 0.03). In GG carriers, serum 25-OH-vitamin D level was not associated with expression of IRX3 and FTO genes. Conclusion There are significant interactions between 25-OH-vitamin D and the expression of FTO and IRX3 genes in the subset of obese patients with specific genotypes for FTO rs9930506. There was no association between serum 25-OH-vitamin D levels and the expression of FTO and IRX genes in individuals with a homozygous genotype for the risk allele of the FTO gene polymorphism.
Collapse
Affiliation(s)
- Maryam Gholamalizadeh
- Student Research Committee, Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeid Doaei
- Research center of Health and Enviroment, School of Health, Guilan university of Medical Sciences, rasht, Iran
| | - Zohreh Mokhtari
- Department of Clinical Biochemistry, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Vahideh Jalili
- Faculty of Medicine, Urmia University of Medical sciences, Urmia, Iran
| | - Fatemeh Bourbour
- Department of Clinical Nutrition and Dietetics, Research Institute Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Saeed Omidi
- Research center of Health and Enviroment, School of Health, Guilan university of Medical Sciences, rasht, Iran
| | - Kamal Ebrahimi
- Msc Student of Psychiatric Nursing, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Naser Kalantari
- Department of community Nutrition and Dietetics, Research Institute Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Sheyda Abdi
- Department of Biochemistry and Dietetics, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ghasem Azizi Tabesh
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Naimi Joubani
- Research center of Health and Enviroment, School of Health, Guilan university of Medical Sciences, rasht, Iran
| | - Esmaeil Roohbakhsh
- Research center of Health and Enviroment, School of Health, Guilan university of Medical Sciences, rasht, Iran
| | | |
Collapse
|
42
|
Appel M, Zentgraf K, Krüger K, Alack K. Effects of Genetic Variation on Endurance Performance, Muscle Strength, and Injury Susceptibility in Sports: A Systematic Review. Front Physiol 2021; 12:694411. [PMID: 34366884 PMCID: PMC8334364 DOI: 10.3389/fphys.2021.694411] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 06/07/2021] [Indexed: 11/25/2022] Open
Abstract
The aim of this systematic review was to assess the effects of genetic variations and polymorphisms on endurance performance, muscle strength and injury susceptibility in competitive sports. The electronic databases PubMed and Web of Science were searched for eligible studies. The study quality was assessed using the RoBANS tool. Studies were included if they met the following criteria: (1) human study in English or German; (2) published in the period 2015–2019; (3) investigation of an association between genetic variants and endurance performance and/or muscle strength and/or endurance/strength training status as well as ligament, tendon, or muscle injuries; (4) participants aged 18–60 years and national or international competition participation; (5) comparison with a control group. Nineteen studies and one replication study were identified. Results revealed that the IGF-1R 275124 A>C rs1464430 polymorphism was overrepresented in endurance trained athletes. Further, genotypes of PPARGC1A polymorphism correlated with performance in endurance exercise capacity tests in athletes. Moreover, the RR genotype of ACTN3 R577X polymorphism, the C allele of IGF-1R polymorphism and the gene variant FTO T>A rs9939609 and/or their AA genotype were linked to muscle strength. In addition, gene variants of MCT1 (T1470A rs1049434) and ACVR1B (rs2854464) were also positively associated with strength athletes. Among others, the gene variants of the MMP group (rs591058 and rs679620) as well as the polymorphism COL5A1 rs13946 were associated with susceptibility to injuries of competitive athletes. Based on the identified gene variants, individualized training programs for injury prevention and optimization of athletic performance could be created for competitive athletes using gene profiling techniques.
Collapse
Affiliation(s)
- Milena Appel
- Department of Exercise Physiology and Sports Therapy, Institute of Sports Science, Justus-Liebig-University Giessen, Giessen, Germany
| | - Karen Zentgraf
- Department of Exercise and Movement Science, Institute of Sports Sciences, Goethe-University Frankfurt, Frankfurt, Germany
| | - Karsten Krüger
- Department of Exercise Physiology and Sports Therapy, Institute of Sports Science, Justus-Liebig-University Giessen, Giessen, Germany
| | - Katharina Alack
- Department of Exercise Physiology and Sports Therapy, Institute of Sports Science, Justus-Liebig-University Giessen, Giessen, Germany
| |
Collapse
|
43
|
Takeda Y, Chijimatsu R, Vecchione A, Arai T, Kitagawa T, Ofusa K, Yabumoto M, Hirotsu T, Eguchi H, Doki Y, Ishii H. Impact of One-Carbon Metabolism-Driving Epitranscriptome as a Therapeutic Target for Gastrointestinal Cancer. Int J Mol Sci 2021; 22:ijms22147278. [PMID: 34298902 PMCID: PMC8306097 DOI: 10.3390/ijms22147278] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 07/01/2021] [Accepted: 07/02/2021] [Indexed: 12/22/2022] Open
Abstract
One-carbon (1C) metabolism plays a key role in biological functions linked to the folate cycle. These include nucleotide synthesis; the methylation of DNA, RNA, and proteins in the methionine cycle; and transsulfuration to maintain the redox condition of cancer stem cells in the tumor microenvironment. Recent studies have indicated that small therapeutic compounds affect the mitochondrial folate cycle, epitranscriptome (RNA methylation), and reactive oxygen species reactions in cancer cells. The epitranscriptome controls cellular biochemical reactions, but is also a platform for cell-to-cell interaction and cell transformation. We present an update of recent advances in the study of 1C metabolism related to cancer and demonstrate the areas where further research is needed. We also discuss approaches to therapeutic drug discovery using animal models and propose further steps toward developing precision cancer medicine.
Collapse
Affiliation(s)
- Yu Takeda
- Center of Medical Innovation and Translational Research, Department of Medical Data Science, Osaka University Graduate School of Medicine, Suita, Yamadaoka 2-2, Osaka 565-0871, Japan; (Y.T.); (R.C.); (T.A.); (T.K.); (K.O.); (M.Y.); (T.H.)
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; (H.E.); (Y.D.)
| | - Ryota Chijimatsu
- Center of Medical Innovation and Translational Research, Department of Medical Data Science, Osaka University Graduate School of Medicine, Suita, Yamadaoka 2-2, Osaka 565-0871, Japan; (Y.T.); (R.C.); (T.A.); (T.K.); (K.O.); (M.Y.); (T.H.)
| | - Andrea Vecchione
- Department of Clinical and Molecular Medicine, University of Rome “Sapienza”, Santo Andrea Hospital, Via di Grottarossa, 1035-00189 Rome, Italy;
| | - Takahiro Arai
- Center of Medical Innovation and Translational Research, Department of Medical Data Science, Osaka University Graduate School of Medicine, Suita, Yamadaoka 2-2, Osaka 565-0871, Japan; (Y.T.); (R.C.); (T.A.); (T.K.); (K.O.); (M.Y.); (T.H.)
- Unitech Co., Ltd., Kashiwa 277-0005, Japan
| | - Toru Kitagawa
- Center of Medical Innovation and Translational Research, Department of Medical Data Science, Osaka University Graduate School of Medicine, Suita, Yamadaoka 2-2, Osaka 565-0871, Japan; (Y.T.); (R.C.); (T.A.); (T.K.); (K.O.); (M.Y.); (T.H.)
- Kyowa-kai Medical Corporation, Osaka 540-0008, Japan
| | - Ken Ofusa
- Center of Medical Innovation and Translational Research, Department of Medical Data Science, Osaka University Graduate School of Medicine, Suita, Yamadaoka 2-2, Osaka 565-0871, Japan; (Y.T.); (R.C.); (T.A.); (T.K.); (K.O.); (M.Y.); (T.H.)
- Food and Life-Science Laboratory, Prophoenix Division, Idea Consultants, Inc., Osaka 559-8519, Japan
| | - Masami Yabumoto
- Center of Medical Innovation and Translational Research, Department of Medical Data Science, Osaka University Graduate School of Medicine, Suita, Yamadaoka 2-2, Osaka 565-0871, Japan; (Y.T.); (R.C.); (T.A.); (T.K.); (K.O.); (M.Y.); (T.H.)
- Kinshu-kai Medical Corporation, Osaka 558-0041, Japan
| | - Takaaki Hirotsu
- Center of Medical Innovation and Translational Research, Department of Medical Data Science, Osaka University Graduate School of Medicine, Suita, Yamadaoka 2-2, Osaka 565-0871, Japan; (Y.T.); (R.C.); (T.A.); (T.K.); (K.O.); (M.Y.); (T.H.)
- Hirotsu Bio Science Inc., Tokyo 107-0062, Japan
| | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; (H.E.); (Y.D.)
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; (H.E.); (Y.D.)
| | - Hideshi Ishii
- Center of Medical Innovation and Translational Research, Department of Medical Data Science, Osaka University Graduate School of Medicine, Suita, Yamadaoka 2-2, Osaka 565-0871, Japan; (Y.T.); (R.C.); (T.A.); (T.K.); (K.O.); (M.Y.); (T.H.)
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; (H.E.); (Y.D.)
- Correspondence: ; Tel.: +81-(0)6-6210-8406 (ext. 8405); Fax: +81-(0)6-6210-8407
| |
Collapse
|
44
|
Rana S, Sultana A, Bhatti AA. Effect of interaction between obesity-promoting genetic variants and behavioral factors on the risk of obese phenotypes. Mol Genet Genomics 2021; 296:919-938. [PMID: 33966103 DOI: 10.1007/s00438-021-01793-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 04/22/2021] [Indexed: 01/28/2023]
Abstract
The studies investigating gene-gene and gene-environment (or gene-behavior) interactions provide valuable insight into the pathomechanisms underlying obese phenotypes. The Pakistani population due to its unique characteristics offers numerous advantages for conducting such studies. In this view, the current study was undertaken to examine the effects of gene-gene and gene-environment/behavior interactions on the risk of obesity in a sample of Pakistani population. A total of 578 adult participants including 290 overweight/obese cases and 288 normal-weight controls were involved. The five key obesity-associated genetic variants namely MC4R rs17782313, BDNF rs6265, FTO rs1421085, TMEM18 rs7561317, and NEGR1 rs2815752 were genotyped using the TaqMan allelic discrimination assays. The data related to behavioral factors, such as eating pattern, diet consciousness, the tendency toward fat-dense food (TFDF), sleep duration, sleep-wake cycle (SWC), shift work (SW), and physical activity levels were collected via a questionnaire. Gene-gene and gene-behavior interactions were analyzed by multifactor dimensionality reduction and linear regression, respectively. In our study, only TMEM18 rs7561317 was found to be significantly associated with anthropometric traits with no significant effect of gene-gene interactions were observed on obesity-related phenotypes. However, the genetic variants were found to interact with the behavioral factors to significantly influence various obesity-related anthropometric traits including BMI, waist circumference, hip circumference, waist-to-hip ratio, waist-to-height ratio, and percentage of body fat. In conclusion, the interaction between genetic architecture and behavior/environment determines the outcome of obesity-related anthropometric phenotypes. Thus, gene-environment/behavior interaction studies should be promoted to explore the risk of complex and multifactorial disorders, such as obesity.
Collapse
Affiliation(s)
- Sobia Rana
- Molecular Biology and Human Genetics Laboratory, Dr. Panjwani Center for Molecular Medicine and Drug Research (PCMD), International Center for Chemical and Biological Sciences (ICCBS), University of Karachi, Karachi, 75270, Pakistan.
| | - Ayesha Sultana
- Molecular Biology and Human Genetics Laboratory, Dr. Panjwani Center for Molecular Medicine and Drug Research (PCMD), International Center for Chemical and Biological Sciences (ICCBS), University of Karachi, Karachi, 75270, Pakistan
| | - Adil Anwar Bhatti
- Molecular Biology and Human Genetics Laboratory, Dr. Panjwani Center for Molecular Medicine and Drug Research (PCMD), International Center for Chemical and Biological Sciences (ICCBS), University of Karachi, Karachi, 75270, Pakistan
| |
Collapse
|
45
|
Naja F, Itani L, Hammoudeh S, Manzoor S, Abbas N, Radwan H, Saber-Ayad M. Dietary Patterns and Their Associations With the FTO and FGF21 Gene Variants Among Emirati Adults. Front Nutr 2021; 8:668901. [PMID: 34095191 PMCID: PMC8171665 DOI: 10.3389/fnut.2021.668901] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 04/12/2021] [Indexed: 01/03/2023] Open
Abstract
Purpose: To examine the dietary patterns and their associations with the FTO and FGF21 gene variants among Emirati adults. Methods: Using a cross-sectional design, healthy adult male and female Emiratis (n = 194) were recruited from primary health care centers in Sharjah, UAE. Participants completed a 61-item semi-quantitative food frequency questionnaire. In addition, a saliva sample was obtained for the genetic analysis. Genotyping was performed for FTOrs9939609(A>T), FTOrs9930506(A>G), FGF21 rs838133 (A > G), and FGF21 rs838145 (A > G). Dietary patterns were derived using the principal component analysis. Logistic regression analyses were used to examine the association of dietary patterns with genetic variants. Results: Three dietary patterns were identified: "Western": consisting of fast food, sweets, and processed meat; "Traditional Emirati" rich in vegetables, traditional Emirati-mixed-dishes and whole dairy; while whole grains, low-fat dairy, and bulgur were components of the "Prudent" pattern. Subjects carrying the A allele of the FTO rs9939609 were 2.41 times more likely to adhere to the Western pattern compared to subjects with genotype TT (OR:2.41; 95%CI:1.05-5.50). Compared with subjects with A/A, those carrying the G allele of the FTO rs9930506 were more likely to follow a Western diet (OR: 2.19; 95%CI: 1.00-4.97). Participants carrying the risk allele (A) of the FGF21 rs838133 were twice more likely to adhere to the Traditional pattern as compared to subjects with genotype GG (OR: 1.9, 95%CI: 1.01-3.57). Conclusions: The findings of this study suggested associations among specific FTO and FGF21 gene variants with dietary patterns among Emirati adults. These findings could be used to inform evidence-based targeted nutrition preventive recommendations, especially those aiming to limit intake of western type foods.
Collapse
Affiliation(s)
- Farah Naja
- Clinical Nutrition and Dietetics Department, College of Health Sciences, University of Sharjah, Sharjah, United Arab Emirates.,Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates.,Nutrition and Food Sciences Department, American University of Beirut, Beirut, Lebanon
| | - Leila Itani
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Beirut Arab University, Beirut, Lebanon
| | - Sarah Hammoudeh
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Shaista Manzoor
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Nada Abbas
- Nutrition and Food Sciences Department, American University of Beirut, Beirut, Lebanon
| | - Hadia Radwan
- Clinical Nutrition and Dietetics Department, College of Health Sciences, University of Sharjah, Sharjah, United Arab Emirates.,Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Maha Saber-Ayad
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates.,Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
46
|
Holzapfel C, Sag S, Graf-Schindler J, Fischer M, Drabsch T, Illig T, Grallert H, Stecher L, Strack C, Caterson ID, Jebb SA, Hauner H, Baessler A. Association between Single Nucleotide Polymorphisms and Weight Reduction in Behavioural Interventions-A Pooled Analysis. Nutrients 2021; 13:nu13030819. [PMID: 33801339 PMCID: PMC7998423 DOI: 10.3390/nu13030819] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 02/23/2021] [Accepted: 02/25/2021] [Indexed: 11/24/2022] Open
Abstract
Knowledge of the association between single nucleotide polymorphisms (SNPs) and weight loss is limited. The aim was to analyse whether selected obesity-associated SNPs within the fat mass and obesity-associated (FTO), transmembrane protein 18 (TMEM18), melanocortin-4 receptor (MC4R), SEC16 homolog B (SEC16B), and brain-derived neurotrophic factor (BDNF) gene are associated with anthropometric changes during behavioural intervention for weight loss. genetic and anthropometric data from 576 individuals with overweight and obesity from four lifestyle interventions were obtained. A genetic predisposition score (GPS) was calculated. Our results show that study participants had a mean age of 48.2 ± 12.6 years and a mean baseline body mass index of 33.9 ± 6.4 kg/m2. Mean weight reduction after 12 months was −7.7 ± 10.9 kg. After 12 months of intervention, the MC4R SNPs rs571312 and rs17782313 were significantly associated with a greater decrease in body weight and BMI (p = 0.012, p = 0.011, respectively). The investigated SNPs within the other four genetic loci showed no statistically significant association with changes in anthropometric parameters. The GPS showed no statistically significant association with weight reduction. In conclusion there was no consistent evidence for statistically significant associations of SNPs with anthropometric changes during a behavioural intervention. It seems that other factors play a more significant in weight management than the investigated SNPs.
Collapse
Affiliation(s)
- Christina Holzapfel
- Institute for Nutritional Medicine, School of Medicine, Technical University of Munich, 80992 Munich, Germany; (J.G.-S.); (T.D.); (L.S.); (H.H.)
- Correspondence: ; Tel.: +49-89-28924923; Fax: +49-89-28924922
| | - Sabine Sag
- Clinic of Internal Medicine II, University Hospital of Regensburg, 93053 Regensburg, Germany; (S.S.); (M.F.); (C.S.); (A.B.)
| | - Johanna Graf-Schindler
- Institute for Nutritional Medicine, School of Medicine, Technical University of Munich, 80992 Munich, Germany; (J.G.-S.); (T.D.); (L.S.); (H.H.)
| | - Marcus Fischer
- Clinic of Internal Medicine II, University Hospital of Regensburg, 93053 Regensburg, Germany; (S.S.); (M.F.); (C.S.); (A.B.)
| | - Theresa Drabsch
- Institute for Nutritional Medicine, School of Medicine, Technical University of Munich, 80992 Munich, Germany; (J.G.-S.); (T.D.); (L.S.); (H.H.)
| | - Thomas Illig
- Hannover Unified Biobank, Hannover Medical School, 30625 Hannover, Germany;
| | - Harald Grallert
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany;
| | - Lynne Stecher
- Institute for Nutritional Medicine, School of Medicine, Technical University of Munich, 80992 Munich, Germany; (J.G.-S.); (T.D.); (L.S.); (H.H.)
| | - Christina Strack
- Clinic of Internal Medicine II, University Hospital of Regensburg, 93053 Regensburg, Germany; (S.S.); (M.F.); (C.S.); (A.B.)
| | - Ian D. Caterson
- Boden Collaboration, Charles Perkins Centre, University of Sydney, Sydney, NSW 2006, Australia;
| | - Susan A. Jebb
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford OX2 6GG, UK;
| | - Hans Hauner
- Institute for Nutritional Medicine, School of Medicine, Technical University of Munich, 80992 Munich, Germany; (J.G.-S.); (T.D.); (L.S.); (H.H.)
- ZIEL Institute for Food and Health, Else Kröner-Fresenius-Center of Nutritional Medicine, School of Life Sciences, Technical University of Munich, 85354 Freising-Weihenstephan, Germany
| | - Andrea Baessler
- Clinic of Internal Medicine II, University Hospital of Regensburg, 93053 Regensburg, Germany; (S.S.); (M.F.); (C.S.); (A.B.)
| |
Collapse
|
47
|
Virani SS, Alonso A, Aparicio HJ, Benjamin EJ, Bittencourt MS, Callaway CW, Carson AP, Chamberlain AM, Cheng S, Delling FN, Elkind MSV, Evenson KR, Ferguson JF, Gupta DK, Khan SS, Kissela BM, Knutson KL, Lee CD, Lewis TT, Liu J, Loop MS, Lutsey PL, Ma J, Mackey J, Martin SS, Matchar DB, Mussolino ME, Navaneethan SD, Perak AM, Roth GA, Samad Z, Satou GM, Schroeder EB, Shah SH, Shay CM, Stokes A, VanWagner LB, Wang NY, Tsao CW. Heart Disease and Stroke Statistics-2021 Update: A Report From the American Heart Association. Circulation 2021; 143:e254-e743. [PMID: 33501848 DOI: 10.1161/cir.0000000000000950] [Citation(s) in RCA: 3544] [Impact Index Per Article: 886.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND The American Heart Association, in conjunction with the National Institutes of Health, annually reports the most up-to-date statistics related to heart disease, stroke, and cardiovascular risk factors, including core health behaviors (smoking, physical activity, diet, and weight) and health factors (cholesterol, blood pressure, and glucose control) that contribute to cardiovascular health. The Statistical Update presents the latest data on a range of major clinical heart and circulatory disease conditions (including stroke, congenital heart disease, rhythm disorders, subclinical atherosclerosis, coronary heart disease, heart failure, valvular disease, venous disease, and peripheral artery disease) and the associated outcomes (including quality of care, procedures, and economic costs). METHODS The American Heart Association, through its Statistics Committee, continuously monitors and evaluates sources of data on heart disease and stroke in the United States to provide the most current information available in the annual Statistical Update. The 2021 Statistical Update is the product of a full year's worth of effort by dedicated volunteer clinicians and scientists, committed government professionals, and American Heart Association staff members. This year's edition includes data on the monitoring and benefits of cardiovascular health in the population, an enhanced focus on social determinants of health, adverse pregnancy outcomes, vascular contributions to brain health, the global burden of cardiovascular disease, and further evidence-based approaches to changing behaviors related to cardiovascular disease. RESULTS Each of the 27 chapters in the Statistical Update focuses on a different topic related to heart disease and stroke statistics. CONCLUSIONS The Statistical Update represents a critical resource for the lay public, policy makers, media professionals, clinicians, health care administrators, researchers, health advocates, and others seeking the best available data on these factors and conditions.
Collapse
|
48
|
Hoang T, Song D, Lee J, Lee EK, Hwangbo Y, Kim J. Association among Body Mass Index, Genetic Variants of FTO, and Thyroid Cancer Risk: A Hospital-Based Case-Control Study of the Cancer Screenee Cohort in Korea. Cancer Res Treat 2020; 53:857-873. [PMID: 33285050 PMCID: PMC8291195 DOI: 10.4143/crt.2020.720] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 12/04/2020] [Indexed: 12/24/2022] Open
Abstract
Purpose Obesity has been determined to be associated with fat mass and obesity-associated (FTO) gene and thyroid cancer risk. However, the effect of combined interactions between obesity and the FTO gene on thyroid cancer needs further investigation. This study aimed to examine whether interactions between body mass index (BMI) and the FTO gene are associated with an increased risk of thyroid cancer. Materials and Methods A total of 705 thyroid cancer cases and 705 sex- and age-matched normal controls were selected from the Cancer Screenee Cohort in National Cancer Center, Korea. A conditional logistic regression model was used to calculate the odds ratios (ORs) and 95% confidence intervals (CIs) for the measure of associations and the combined effect of BMI and FTO gene on thyroid cancer. Results BMI was associated with an increased risk of thyroid cancer in subclasses of overweight (23–24.9 kg/m2; adjusted OR, 1.50; 95% CI, 1.12 to 2.00) and obese (≥ 25 kg/m2) (adjusted OR, 1.62; 95% CI, 1.23 to 2.14). There were positive associations between the FTO genetic variants rs8047395 and rs8044769 and an increased risk of thyroid cancer. Additionally, the combination of BMI subclasses and FTO gene variants was significantly associated with thyroid cancer risk in the codominant (rs17817288), dominant (rs9937053, rs12149832, rs1861867, and rs7195539), and recessive (rs17817288 and rs8044769) models. Conclusion Findings from this study identified the effects of BMI on thyroid cancer risk among individuals carrying rs17817288, rs9937053, rs12149832, rs1861867, rs7195539, and rs8044769, whereas the effects of BMI may be modified according to individual characteristics of other FTO variants.
Collapse
Affiliation(s)
- Tung Hoang
- Department of Cancer Biomedical Science, National Cancer Center Graduate School of Cancer Science and Policy, Goyang, Korea
| | - Dayoung Song
- Department of Cancer Control and Population Health, National Cancer Center Graduate School of Cancer Science and Policy, Goyang, Korea
| | - Jeonghee Lee
- Department of Cancer Biomedical Science, National Cancer Center Graduate School of Cancer Science and Policy, Goyang, Korea
| | - Eun Kyung Lee
- Center for Thyroid Cancer, National Cancer Center, Goyang, Korea
| | - Yul Hwangbo
- Center for Thyroid Cancer, National Cancer Center, Goyang, Korea
| | - Jeongseon Kim
- Department of Cancer Biomedical Science, National Cancer Center Graduate School of Cancer Science and Policy, Goyang, Korea
| |
Collapse
|
49
|
Mehrdad M, Doaei S, Gholamalizadeh M, Fardaei M, Fararouei M, Eftekhari MH. Association of FTO rs9939609 polymorphism with serum leptin, insulin, adiponectin, and lipid profile in overweight adults. Adipocyte 2020; 9:51-56. [PMID: 31996075 PMCID: PMC6999843 DOI: 10.1080/21623945.2020.1722550] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
FTO gene polymorphisms are associated with obesity and food intake. This study aimed to investigate the association of FTO rs9939609 polymorphism genotypes with serum glucose, lipid profile and serum hormones level. This cross-sectional study was carried out on 196 randomly selected overweight adults. Anthropometric measurements including weight, height, body mass index (BMI), fat mass, and fat-free mass were assessed. Serum TGs, total cholesterol, HDL cholesterol, LDL cholesterol, glucose and insulin levels were measured. The FTO gene was Genotyped for rs9939609 polymorphism. Dietary intake was assessed by avalid 168-item semi-quantitative food frequency questionnaire (FFQ). The homozygotes for the FTO rs9939609 risk allele (A) had higher serum leptin (p = 0.005, F: 5.131) and lower HDL (p = 0.001, F: 7.687) level than TT genotype. The differences between TT and AT genotypes were not significant. The association remained significant for HDL level after adjustments for age and sex, calorie intake, physical activity, and BMI. The association between rs9939609 polymorphism genotypes and leptin was disappeared after adjustments for calorie intake and physical activity. In conclusion, rs9939609 risk allele was associated with higher serum leptin and lower HDL levels in overweight people. Further studies are warranted.
Collapse
Affiliation(s)
- Mahsa Mehrdad
- Department of Clinical Nutrition, School of Nutrition & Food Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saeid Doaei
- Student Research Committee, Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Research Center of Health and Environment, Guilan University of Medical Sciences, Rasht, Iran
| | - Maryam Gholamalizadeh
- Student Research Committee, Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Majid Fardaei
- Department of Medical Genetics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Fararouei
- HIV/AIDs Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Hassan Eftekhari
- Department of Clinical Nutrition, School of Nutrition & Food Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
50
|
Svoboda LK, Neier K, Wang K, Cavalcante RG, Rygiel CA, Tsai Z, Jones TR, Liu S, Goodrich JM, Lalancette C, Colacino JA, Sartor MA, Dolinoy DC. Tissue and sex-specific programming of DNA methylation by perinatal lead exposure: implications for environmental epigenetics studies. Epigenetics 2020; 16:1102-1122. [PMID: 33164632 DOI: 10.1080/15592294.2020.1841872] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Early developmental environment can influence long-term health through reprogramming of the epigenome. Human environmental epigenetics studies rely on surrogate tissues, such as blood, to assess the effects of environment on disease-relevant but inaccessible target tissues. However, the extent to which environment-induced epigenetic changes are conserved between these tissues is unclear. A better understanding of this conservation is imperative for effective design and interpretation of human environmental epigenetics studies. The Toxicant Exposures and Responses by Genomic and Epigenomic Regulators of Transcription (TaRGET II) consortium was established by the National Institute of Environmental Health Sciences to address the utility of surrogate tissues as proxies for toxicant-induced epigenetic changes in target tissues. We and others have recently reported that perinatal exposure to lead (Pb) is associated with adverse metabolic outcomes. Here, we investigated the sex-specific effects of perinatal exposure to a human environmentally relevant level of Pb on DNA methylation in paired liver and blood samples from adult mice using enhanced reduced-representation bisulphite sequencing. Although Pb exposure ceased at 3 weeks of age, we observed thousands of sex-specific differentially methylated cytosines in the blood and liver of Pb-exposed animals at 5 months of age, including 44 genomically imprinted loci. We observed significant tissue overlap in the genes mapping to differentially methylated cytosines. A small but significant subset of Pb-altered genes exhibit basal sex differences in gene expression in the mouse liver. Collectively, these data identify potential molecular targets for Pb-induced metabolic diseases, and inform the design of more robust human environmental epigenomics studies.
Collapse
Affiliation(s)
- Laurie K Svoboda
- Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Kari Neier
- Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Kai Wang
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School Palmer Commons, Ann Arbor, MI, USA
| | | | - Christine A Rygiel
- Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Zing Tsai
- Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA.,Department of Computational Medicine and Bioinformatics, University of Michigan Medical School Palmer Commons, Ann Arbor, MI, USA
| | - Tamara R Jones
- Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Siyu Liu
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School Palmer Commons, Ann Arbor, MI, USA
| | - Jaclyn M Goodrich
- Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Claudia Lalancette
- Epigenomics Core, University of Michigan, Medical School, Ann Arbor, MI, USA
| | - Justin A Colacino
- Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA.,Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Maureen A Sartor
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School Palmer Commons, Ann Arbor, MI, USA.,Department of Biostatistics, University of Michigan, School of Public Health, Ann Arbor, MI, USA
| | - Dana C Dolinoy
- Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA.,Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| |
Collapse
|