1
|
Gala K, Ghusn W, Fansa S, Anazco D, Storm AC, Abu Dayyeh BK, Acosta A. Impact of Leptin-Melanocortin Pathway Genetic Variants on Weight Loss Outcomes After Endoscopic Transoral Outlet Reduction. Obes Surg 2024:10.1007/s11695-024-07547-0. [PMID: 39419959 DOI: 10.1007/s11695-024-07547-0] [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: 04/29/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 10/19/2024]
Abstract
PURPOSE Variants in the leptin-melanocortin pathway (LMP) are associated with severe obesity. We evaluated weight loss of patients with or without heterozygous LMP variants, with weight recurrence after Roux-en-Y gastric bypass, who underwent endoscopic transoral outlet reduction (TORe). MATERIALS AND METHODS We retrospectively reviewed patients genotyped for an LMP who had undergone TORe, classified as "carriers" or "non-carriers" of genetic variants. RESULTS We included 54 patients (22 carriers, 32 non-carriers). We identified 34 genetic variants in 21 different genes in 22 patients. Total body weight loss (%TBWL) after TORe was significantly different at 9 and 12 months (12 months: 0.68 ± 7.5% vs. 9.6 ± 8.2%, p < 0.01). This difference in weight loss was present even when analyzed in subgroups of patients who had undergone tubular TORe technique, and TORe plus APC. At 3, 6, and 12 months, the percentage of carriers achieving ≥ 5% and ≥ 10% TBWL was lower than non-carriers. CONCLUSIONS Patients with LMP variant who underwent RYGB had decreased weight loss 1 year after undergoing TORe.
Collapse
Affiliation(s)
- Khushboo Gala
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Wissam Ghusn
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Sima Fansa
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Diego Anazco
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Andrew C Storm
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Barham K Abu Dayyeh
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Andres Acosta
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
2
|
Abstract
Obesity is a multi-factorial disease that is influenced by genetic, epigenetic, and environmental factors. Precision medicine is a practice wherein prevention and treatment strategies take individual variability into account. It involves using a variety of factors including deep phenotyping using clinical, physiologic, and behavioral characteristics, 'omics assays (eg, genomics, epigenomics, transcriptomics, and microbiomics among others), and environmental factors to devise practices that are individualized to subsets of patients. Personalizing the therapeutic modality to the individual can lead to enhanced effectiveness and tolerability. The authors review advances in precision medicine made in the field of bariatrics and discuss future avenues and challenges.
Collapse
Affiliation(s)
- Khushboo Gala
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55902, USA. https://twitter.com/KhushbooSGala
| | - Wissam Ghusn
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55902, USA; Department of Internal Medicine, Boston University Medical Center, Harrison Avenue, Boston, MA 02111, USA. https://twitter.com/Wissam_Ghusn
| | - Andres Acosta
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55902, USA.
| |
Collapse
|
3
|
Wen J, Tian YE, Skampardoni I, Yang Z, Cui Y, Anagnostakis F, Mamourian E, Zhao B, Toga AW, Zalesky A, Davatzikos C. The genetic architecture of biological age in nine human organ systems. NATURE AGING 2024; 4:1290-1307. [PMID: 38942983 PMCID: PMC11446180 DOI: 10.1038/s43587-024-00662-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 05/30/2024] [Indexed: 06/30/2024]
Abstract
Investigating the genetic underpinnings of human aging is essential for unraveling the etiology of and developing actionable therapies for chronic diseases. Here, we characterize the genetic architecture of the biological age gap (BAG; the difference between machine learning-predicted age and chronological age) across nine human organ systems in 377,028 participants of European ancestry from the UK Biobank. The BAGs were computed using cross-validated support vector machines, incorporating imaging, physical traits and physiological measures. We identify 393 genomic loci-BAG pairs (P < 5 × 10-8) linked to the brain, eye, cardiovascular, hepatic, immune, metabolic, musculoskeletal, pulmonary and renal systems. Genetic variants associated with the nine BAGs are predominantly specific to the respective organ system (organ specificity) while exerting pleiotropic links with other organ systems (interorgan cross-talk). We find that genetic correlation between the nine BAGs mirrors their phenotypic correlation. Further, a multiorgan causal network established from two-sample Mendelian randomization and latent causal variance models revealed potential causality between chronic diseases (for example, Alzheimer's disease and diabetes), modifiable lifestyle factors (for example, sleep duration and body weight) and multiple BAGs. Our results illustrate the potential for improving human organ health via a multiorgan network, including lifestyle interventions and drug repurposing strategies.
Collapse
Affiliation(s)
- Junhao Wen
- Laboratory of AI and Biomedical Science (LABS), University of Southern California, Los Angeles, CA, USA.
| | - Ye Ella Tian
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, Melbourne Medical School, The University of Melbourne, Melbourne, Victoria, Australia
| | - Ioanna Skampardoni
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhijian Yang
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yuhan Cui
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Elizabeth Mamourian
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Bingxin Zhao
- Department of Statistics and Data Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Arthur W Toga
- Laboratory of Neuro Imaging (LONI), Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Andrew Zalesky
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, Melbourne Medical School, The University of Melbourne, Melbourne, Victoria, Australia
| | - Christos Davatzikos
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
4
|
Jiang Y, Xun Y, Zhang Z. Central regulation of feeding and body weight by ciliary GPR75. J Clin Invest 2024; 134:e182121. [PMID: 39137039 PMCID: PMC11444156 DOI: 10.1172/jci182121] [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: 04/16/2024] [Accepted: 08/07/2024] [Indexed: 08/15/2024] Open
Abstract
Variants of the G protein-coupled receptor 75 (GPR75) are associated with a lower BMI in large-scale human exome-sequencing studies. However, how GPR75 regulates body weight remains poorly understood. Using random germline mutagenesis in mice, we identified a missense allele (Thinner) of Gpr75 that resulted in a lean phenotype and verified the decreased body weight and fat weight in Gpr75-knockout (Gpr75-/-) mice. Gpr75-/- mice displayed reduced food intake under high-fat diet (HFD) feeding, and pair-feeding normalized their body weight. The endogenous GPR75 protein was exclusively expressed in the brains of 3xFlag-tagged Gpr75-knockin (3xFlag-Gpr75) mice, with consistent expression across different brain regions. GPR75 interacted with Gαq to activate various signaling pathways after HFD feeding. Additionally, GPR75 was localized in the primary cilia of hypothalamic cells, whereas the Thinner mutation (L144P) and human GPR75 variants in individuals with a lower BMI failed to localize in the cilia. Loss of GPR75 selectively inhibited weight gain in HFD-fed mice but failed to suppress the development of obesity in leptin ob-mutant (Lepob-mutant) mice and adenylate cyclase 3-mutant (Adcy3-mutant) mice on a chow diet. Our data reveal that GPR75 is a ciliary protein expressed in the brain and plays an important role in regulating food intake.
Collapse
Affiliation(s)
- Yiao Jiang
- Center for the Genetics of Host Defense and
- Division of Endocrinology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Yu Xun
- Center for the Genetics of Host Defense and
- Division of Endocrinology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Zhao Zhang
- Center for the Genetics of Host Defense and
- Division of Endocrinology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
5
|
Wen J, Tian YE, Skampardoni I, Yang Z, Cui Y, Anagnostakis F, Mamourian E, Zhao B, Toga AW, Zaleskey A, Davatzikos C. The Genetic Architecture of Biological Age in Nine Human Organ Systems. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.06.08.23291168. [PMID: 37398441 PMCID: PMC10312870 DOI: 10.1101/2023.06.08.23291168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Understanding the genetic basis of biological aging in multi-organ systems is vital for elucidating age-related disease mechanisms and identifying therapeutic interventions. This study characterized the genetic architecture of the biological age gap (BAG) across nine human organ systems in 377,028 individuals of European ancestry from the UK Biobank. We discovered 393 genomic loci-BAG pairs (P-value<5×10-8) linked to the brain, eye, cardiovascular, hepatic, immune, metabolic, musculoskeletal, pulmonary, and renal systems. We observed BAG-organ specificity and inter-organ connections. Genetic variants associated with the nine BAGs are predominantly specific to the respective organ system while exerting pleiotropic effects on traits linked to multiple organ systems. A gene-drug-disease network confirmed the involvement of the metabolic BAG-associated genes in drugs targeting various metabolic disorders. Genetic correlation analyses supported Cheverud's Conjecture1 - the genetic correlation between BAGs mirrors their phenotypic correlation. A causal network revealed potential causal effects linking chronic diseases (e.g., Alzheimer's disease), body weight, and sleep duration to the BAG of multiple organ systems. Our findings shed light on promising therapeutic interventions to enhance human organ health within a complex multi-organ network, including lifestyle modifications and potential drug repositioning strategies for treating chronic diseases. All results are publicly available at https://labs-laboratory.com/medicine.
Collapse
Affiliation(s)
- Junhao Wen
- Laboratory of AI and Biomedical Science (LABS), Keck School of Medicine of USC, University of Southern California, Los Angeles, California, USA
| | - Ye Ella Tian
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, Melbourne Medical School, The University of Melbourne, Melbourne, Victoria, Australia
| | - Ioanna Skampardoni
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Zhijian Yang
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Yuhan Cui
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Filippos Anagnostakis
- Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy
| | - Elizabeth Mamourian
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Bingxin Zhao
- Department of Statistics and Data Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Arthur W. Toga
- Laboratory of Neuro Imaging (LONI), Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, California, USA
| | - Andrew Zaleskey
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, Melbourne Medical School, The University of Melbourne, Melbourne, Victoria, Australia
| | - Christos Davatzikos
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| |
Collapse
|
6
|
Leung AKC, Wong AHC, Hon KL. Childhood Obesity: An Updated Review. Curr Pediatr Rev 2024; 20:2-26. [PMID: 35927921 DOI: 10.2174/1573396318666220801093225] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 04/05/2022] [Accepted: 05/19/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Childhood obesity is an important and serious public health problem worldwide. OBJECTIVE This article aims to familiarize physicians with the evaluation, management, and prevention of childhood. METHODS A PubMed search was conducted in May, 2021, in Clinical Queries using the key terms "obesity" OR "obese". The search included clinical trials, randomized controlled trials, case-control studies, cohort studies, meta-analyses, observational studies, clinical guidelines, case reports, case series, and reviews. The search was restricted to English literature and children. The information retrieved from the above search was used in the compilation of the present article. RESULTS Most obese children have exogenous obesity characterized by a growth rate for height above the 50th percentile, normal intelligence, normal genitalia, and lack of historical or physical evidence of an endocrine abnormality or a congenital syndrome. Obese children are at risk for dyslipidemia, hypertension, diabetes mellitus, non-alcoholic fatty liver disease, obstructive sleep apnea, psychosocial disturbances, impaired quality of life, and shorter life expectancy. The multitude of serious comorbidities necessitates effective treatment modalities. Dietary modification, therapeutic exercise, and behavioral modification are the fundamentals of treatment. Pharmacotherapy and/or bariatric surgery should be considered for obese individuals who do not respond to the above measures and suffer from a serious comorbid condition. CONCLUSION Childhood obesity, once established, is often refractory to treatment. Most treatment programs lead to a brief period of weight loss, followed by rapid re-accumulation of the lost weight after the termination of therapy. As such, preventive activity is the key to solving the problem of childhood obesity. Childhood obesity can be prevented by promoting a healthy diet, regular physical activity, and lifestyle modification. Parents should be encouraged to get involved in school and community programs that improve their children's nutritional status and physical activity.
Collapse
Affiliation(s)
- Alexander K C Leung
- Department of Pediatrics, The University of Calgary, The Alberta Children's Hospital, Calgary, Alberta, Canada
| | - Alex H C Wong
- Department of Family Medicine, The University of Calgary, Calgary, Alberta, Canada
| | - Kam Lun Hon
- Department of Paediatrics, The Chinese University of Hong Kong, Hong Kong, China
- Department of Paediatrics and Adolescent Medicine, The Hong Kong Children's Hospital, Hong Kong, China
| |
Collapse
|
7
|
van der Meer R, Mohamed SA, Monpellier VM, Liem RSL, Hazebroek EJ, Franks PW, Frayling TM, Janssen IMC, Serlie MJ. Genetic variants associated with weight loss and metabolic outcomes after bariatric surgery: A systematic review. Obes Rev 2023; 24:e13626. [PMID: 37632325 DOI: 10.1111/obr.13626] [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: 02/06/2023] [Revised: 07/15/2023] [Accepted: 07/17/2023] [Indexed: 08/27/2023]
Abstract
The extent to which genetic variations contribute to interindividual differences in weight loss and metabolic outcomes after bariatric surgery is unknown. Identifying genetic variants that impact surgery outcomes may contribute to clinical decision making. This review evaluates current evidence addressing the association of genetic variants with weight loss and changes in metabolic parameters after bariatric surgery. A search was conducted using Medline, Embase, Scopus, Web of Science, and Cochrane Library. Fifty-two eligible studies were identified. Single nucleotide polymorphisms (SNPs) at ADIPOQ (rs226729, rs1501299, rs3774261, and rs17300539) showed a positive association with postoperative change in measures of glucose homeostasis and lipid profiles (n = 4), but not with weight loss after surgery (n = 6). SNPs at FTO (rs11075986, rs16952482, rs8050136, rs9939609, rs9930506, and rs16945088) (n = 10) and MC4R (rs11152213, rs476828, rs2229616, rs9947255, rs17773430, rs5282087, and rs17782313) (n = 9) were inconsistently associated with weight loss and metabolic improvement. Four studies examining the UCP2 SNP rs660339 reported associations with postsurgical weight loss. In summary, there is limited evidence supporting a role for specific genetic variants in surgical outcomes after bariatric surgery. Most studies have adopted a candidate gene approach, limiting the scope for discovery, suggesting that the absence of compelling evidence is not evidence of absence.
Collapse
Affiliation(s)
- Rieneke van der Meer
- Nederlandse Obesitas Kliniek, Huis ter Heide, The Netherlands
- Department of Endocrinology & Metabolism, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Siham A Mohamed
- Genetic and Molecular Epidemiology Unit, Department of Clinical Sciences, Lund University, Lund, Sweden
| | | | - Ronald S L Liem
- Department of Surgery, Groene Hart Hospital, Gouda, The Netherlands
- Nederlandse Obesitas Kliniek, The Hague and Gouda, The Netherlands
| | - Eric J Hazebroek
- Department of Surgery, Rijnstate Hospital/Vitalys Clinics, Arnhem, The Netherlands
| | - Paul W Franks
- Genetic and Molecular Epidemiology Unit, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Timothy M Frayling
- Genetics of Complex Traits, University of Exeter Medical School, University of Exeter, Exeter, UK
| | | | - Mireille J Serlie
- Department of Endocrinology & Metabolism, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Department of Endocrinology & Metabolism, Yale University, New Haven, CT, USA
| |
Collapse
|
8
|
Xu Y, Yang D, Wang L, Król E, Mazidi M, Li L, Huang Y, Niu C, Liu X, Lam SM, Shui G, Douglas A, Speakman JR. Maternal High Fat Diet in Lactation Impacts Hypothalamic Neurogenesis and Neurotrophic Development, Leading to Later Life Susceptibility to Obesity in Male but Not Female Mice. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2305472. [PMID: 37867217 PMCID: PMC10724448 DOI: 10.1002/advs.202305472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Indexed: 10/24/2023]
Abstract
Early life nutrition can reprogram development and exert long-term consequences on body weight regulation. In mice, maternal high-fat diet (HFD) during lactation predisposed male but not female offspring to diet-induced obesity when adult. Molecular and cellular changes in the hypothalamus at important time points are examined in the early postnatal life in relation to maternal diet and demonstrated sex-differential hypothalamic reprogramming. Maternal HFD in lactation decreased the neurotropic development of neurons formed at the embryo stage (e12.5) and impaired early postnatal neurogenesis in the hypothalamic regions of both males and females. Males show a larger increased ratio of Neuropeptide Y (NPY) to Pro-opiomelanocortin (POMC) neurons in early postnatal neurogenesis, in response to maternal HFD, setting an obese tone for male offspring. These data provide insights into the mechanisms by which hypothalamic reprograming by early life overnutrition contributes to the sex-dependent susceptibility to obesity in adult life in mice.
Collapse
Affiliation(s)
- Yanchao Xu
- Shenzhen key laboratory for metabolic healthCenter for Energy Metabolism and ReproductionShenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhen518055P. R. China
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental BiologyChinese Academy of SciencesBeijing100101P. R. China
| | - Dengbao Yang
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental BiologyChinese Academy of SciencesBeijing100101P. R. China
| | - Lu Wang
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental BiologyChinese Academy of SciencesBeijing100101P. R. China
- Institute of Biological and Environmental SciencesUniversity of AberdeenAberdeenScotlandAB24 2TZUK
- University of Chinese Academy of SciencesShijingshanBeijing100049P. R. China
- School of PharmacyKey Laboratory of Molecular Pharmacology and Drug EvaluationMinistry of EducationYantai UniversityYantai264005P. R. China
| | - Elżbieta Król
- Institute of Biological and Environmental SciencesUniversity of AberdeenAberdeenScotlandAB24 2TZUK
| | - Mohsen Mazidi
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental BiologyChinese Academy of SciencesBeijing100101P. R. China
- University of Chinese Academy of SciencesShijingshanBeijing100049P. R. China
| | - Li Li
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental BiologyChinese Academy of SciencesBeijing100101P. R. China
- University of Chinese Academy of SciencesShijingshanBeijing100049P. R. China
| | - Yi Huang
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental BiologyChinese Academy of SciencesBeijing100101P. R. China
| | - Chaoqun Niu
- Shenzhen key laboratory for metabolic healthCenter for Energy Metabolism and ReproductionShenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhen518055P. R. China
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental BiologyChinese Academy of SciencesBeijing100101P. R. China
| | - Xue Liu
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental BiologyChinese Academy of SciencesBeijing100101P. R. China
| | - Sin Man Lam
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental BiologyChinese Academy of SciencesBeijing100101P. R. China
| | - Guanghou Shui
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental BiologyChinese Academy of SciencesBeijing100101P. R. China
| | - Alex Douglas
- Institute of Biological and Environmental SciencesUniversity of AberdeenAberdeenScotlandAB24 2TZUK
| | - John R. Speakman
- Shenzhen key laboratory for metabolic healthCenter for Energy Metabolism and ReproductionShenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhen518055P. R. China
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental BiologyChinese Academy of SciencesBeijing100101P. R. China
- Institute of Biological and Environmental SciencesUniversity of AberdeenAberdeenScotlandAB24 2TZUK
- China medical universityShenyang110000P. R. China
| |
Collapse
|
9
|
Tapanee P, Reeder N, Christensen R, Tolar-Peterson T. Sugar, non-nutritive sweetener intake and obesity risk in college students. JOURNAL OF AMERICAN COLLEGE HEALTH : J OF ACH 2023; 71:2093-2098. [PMID: 34468279 DOI: 10.1080/07448481.2021.1960844] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 05/27/2021] [Accepted: 07/22/2021] [Indexed: 06/13/2023]
Abstract
OBJECTIVE The purpose of this study was to investigate the association between sugar and NNS consumption with body fat percentage in young adults. METHODS 524 college students were asked to complete the Web-based National Institutes of Health (NIH) Diet History Questionnaire II and had their body fat percentage measured using a bioelectrical impedance analysis scale. RESULT One-fourth of the participants were classified as overfat/obese and of those overfat or obese, 49% of them were African American. Overfat/obese participants consumed significantly more sugar than the heathy group and the underfat group (p < 0.001). Saccharin consumption was significantly higher in the overfat/obese group compare to the underfat and normal groups (p < 0.001). The odds of becoming overfat/obese increased 1.2 times for excessive total sugar consumption (95% CI: 1.0-1.8, p < 0.05). CONCLUSION Excessive sugar and saccharin consumption were associated with higher body fat percentage in young adults.
Collapse
Affiliation(s)
- Pradtana Tapanee
- Department of Food Science, Nutrition, and Health Promotion, College of Agriculture and Life Sciences, Mississippi State University, Starkville, Mississippi, USA
- Institute of Nutrition, Mahidol University, Salaya, Nakhon Pathom, Thailand
| | - Nicole Reeder
- Department of Food Science, Nutrition, and Health Promotion, College of Agriculture and Life Sciences, Mississippi State University, Starkville, Mississippi, USA
| | - Rebecca Christensen
- Department of Food Science, Nutrition, and Health Promotion, College of Agriculture and Life Sciences, Mississippi State University, Starkville, Mississippi, USA
| | - Terezie Tolar-Peterson
- Department of Food Science, Nutrition, and Health Promotion, College of Agriculture and Life Sciences, Mississippi State University, Starkville, Mississippi, USA
| |
Collapse
|
10
|
Guo C, Shen W, Jin W, Jia X, Ji Z, Jinling L, Bin L. Effects of epigallocatechin gallate, caffeine, and their combination on fat accumulation in high-glucose diet-fed Caenorhabditis elegans. Biosci Biotechnol Biochem 2023; 87:898-906. [PMID: 37363872 DOI: 10.1093/bbb/zbad068] [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: 03/09/2023] [Accepted: 05/26/2023] [Indexed: 06/28/2023]
Abstract
Epigallocatechin gallate (EGCG) and caffeine are inevitable to be ingested together in the process of drinking green tea. This study used Caenorhabditis elegans as an organism model to examine whether the binding of EGCG and caffeine could influence the fat-reduction effect. The results revealed that EGCG significantly reduced the Nile Red fluorescence intensity and the triglyceride/protein ratio of the C. elegans obesity model by 14.7% and 16.5%, respectively, while the effect of caffeine was not significant. Moreover, the degree of reduction in fluorescence intensity and triglyceride/protein ratio by EGCG + caffeine was comparable to that of EGCG. In the exploration of underlying mechanism, we found that EGCG and EGCG + caffeine treatments had no influence on food intake and energy expenditure of C. elegans. Their fat-reduction effects were dependent on the regulation of lipogenesis, as shown by the decreased expression of the sbp-1, fat-7, and daf-16 genes.
Collapse
Affiliation(s)
- Cheng Guo
- College of Food Science and Engineering, Wuhan Polytechnic University, Wuhan, China
- Key Laboratory for Deep Processing of Major Grain and Oil (Wuhan Polytechnic University), Ministry of Education, Wuhan, China
- Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Wuhan Polytechnic University, Wuhan, China
| | - Wangyang Shen
- College of Food Science and Engineering, Wuhan Polytechnic University, Wuhan, China
- Key Laboratory for Deep Processing of Major Grain and Oil (Wuhan Polytechnic University), Ministry of Education, Wuhan, China
- Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Wuhan Polytechnic University, Wuhan, China
| | - Weiping Jin
- College of Food Science and Engineering, Wuhan Polytechnic University, Wuhan, China
- Key Laboratory for Deep Processing of Major Grain and Oil (Wuhan Polytechnic University), Ministry of Education, Wuhan, China
- Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Wuhan Polytechnic University, Wuhan, China
| | - Xiwu Jia
- College of Food Science and Engineering, Wuhan Polytechnic University, Wuhan, China
- Key Laboratory for Deep Processing of Major Grain and Oil (Wuhan Polytechnic University), Ministry of Education, Wuhan, China
- Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Wuhan Polytechnic University, Wuhan, China
| | - Zhili Ji
- College of Food Science and Engineering, Wuhan Polytechnic University, Wuhan, China
- Key Laboratory for Deep Processing of Major Grain and Oil (Wuhan Polytechnic University), Ministry of Education, Wuhan, China
- Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Wuhan Polytechnic University, Wuhan, China
| | - Li Jinling
- College of Food Science and Engineering, Wuhan Polytechnic University, Wuhan, China
- Key Laboratory for Deep Processing of Major Grain and Oil (Wuhan Polytechnic University), Ministry of Education, Wuhan, China
- Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Wuhan Polytechnic University, Wuhan, China
| | - Li Bin
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural University), Ministry of Education, Wuhan, China
| |
Collapse
|
11
|
Thakkar P, Pauza AG, Murphy D, Paton JFR. Carotid body: an emerging target for cardiometabolic co-morbidities. Exp Physiol 2023; 108:661-671. [PMID: 36999224 PMCID: PMC10988524 DOI: 10.1113/ep090090] [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: 10/04/2021] [Accepted: 03/03/2023] [Indexed: 04/01/2023]
Abstract
NEW FINDINGS What is the topic of this review? Regarding the global metabolic syndrome crisis, this review focuses on common mechanisms for high blood sugar and high blood pressure. Connections are made between the homeostatic regulation of blood pressure and blood sugar and their dysregulation to reveal signalling mechanisms converging on the carotid body. What advances does it highlight? The carotid body plays a major part in the generation of excessive sympathetic activity in diabetes and also underpins diabetic hypertension. As treatment of diabetic hypertension is notoriously difficult, we propose that novel receptors within the carotid body may provide a novel treatment strategy. ABSTRACT The maintenance of glucose homeostasis is obligatory for health and survival. It relies on peripheral glucose sensing and signalling between the brain and peripheral organs via hormonal and neural responses that restore euglycaemia. Failure of these mechanisms causes hyperglycaemia or diabetes. Current anti-diabetic medications control blood glucose but many patients remain with hyperglycemic condition. Diabetes is often associated with hypertension; the latter is more difficult to control in hyperglycaemic conditions. We ask whether a better understanding of the regulatory mechanisms of glucose control could improve treatment of both diabetes and hypertension when they co-exist. With the involvement of the carotid body (CB) in glucose sensing, metabolic regulation and control of sympathetic nerve activity, we consider the CB as a potential treatment target for both diabetes and hypertension. We provide an update on the role of the CB in glucose sensing and glucose homeostasis. Physiologically, hypoglycaemia stimulates the release of hormones such as glucagon and adrenaline, which mobilize or synthesize glucose; however, these counter-regulatory responses were markedly attenuated after denervation of the CBs in animals. Also, CB denervation prevents and reverses insulin resistance and glucose intolerance. We discuss the CB as a metabolic regulator (not just a sensor of blood gases) and consider recent evidence of novel 'metabolic' receptors within the CB and putative signalling peptides that may control glucose homeostasis via modulation of the sympathetic nervous system. The evidence presented may inform future clinical strategies in the treatment of patients with both diabetes and hypertension, which may include the CB.
Collapse
Affiliation(s)
- Pratik Thakkar
- Manaaki Manawa – the Centre for Heart Research, Department of Physiology, Faculty of Medical and Health SciencesUniversity of AucklandAucklandNew Zealand
| | - Audrys G. Pauza
- Manaaki Manawa – the Centre for Heart Research, Department of Physiology, Faculty of Medical and Health SciencesUniversity of AucklandAucklandNew Zealand
| | - David Murphy
- Molecular Neuroendocrinology Research Group, Bristol Medical School: Translational Health SciencesUniversity of BristolBristolUK
| | - Julian F. R. Paton
- Manaaki Manawa – the Centre for Heart Research, Department of Physiology, Faculty of Medical and Health SciencesUniversity of AucklandAucklandNew Zealand
| |
Collapse
|
12
|
Westbury S, Oyebode O, van Rens T, Barber TM. Obesity Stigma: Causes, Consequences, and Potential Solutions. Curr Obes Rep 2023; 12:10-23. [PMID: 36781624 PMCID: PMC9985585 DOI: 10.1007/s13679-023-00495-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/20/2022] [Indexed: 02/15/2023]
Abstract
PURPOSE OF REVIEW This review aims to examine (i) the aetiology of obesity; (ii) how and why a perception of personal responsibility for obesity so dominantly frames this condition and how this mindset leads to stigma; (iii) the consequences of obesity stigma for people living with obesity, and for the public support for interventions to prevent and manage this condition; and (iv) potential strategies to diminish our focus on personal responsibility for the development of obesity, to enable a reduction of obesity stigma, and to move towards effective interventions to prevent and manage obesity within the population. RECENT FINDINGS We summarise literature which shows that obesity stems from a complex interplay of genetic and environment factors most of which are outside an individual's control. Despite this, evidence of obesity stigmatisation remains abundant throughout areas of media, entertainment, social media and the internet, advertising, news outlets, and the political and public health landscape. This has damaging consequences including psychological, physical, and socioeconomic harm. Obesity stigma does not prevent obesity. A combined, concerted, and sustained effort from multiple stakeholders and key decision-makers within society is required to dispel myths around personal responsibility for body weight, and to foster more empathy for people living in larger bodies. This also sets the scene for more effective policies and interventions, targeting the social and environmental drivers of health, to ultimately improve population health.
Collapse
Affiliation(s)
- Susannah Westbury
- School of Public Health and Preventative Medicine, Monash University, Melbourne, VIC, Australia.
| | - Oyinlola Oyebode
- Wolfson Institute of Population Health, Queen Mary University of London, London, UK
| | - Thijs van Rens
- Department of Economics, University of Warwick, Coventry, UK
| | | |
Collapse
|
13
|
O'Sullivan JW, Ashley EA, Elliott PM. Polygenic risk scores for the prediction of cardiometabolic disease. Eur Heart J 2023; 44:89-99. [PMID: 36478054 DOI: 10.1093/eurheartj/ehac648] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 08/28/2022] [Accepted: 10/27/2022] [Indexed: 12/12/2022] Open
Abstract
Cardiometabolic diseases contribute more to global morbidity and mortality than any other group of disorders. Polygenic risk scores (PRSs), the weighted summation of individually small-effect genetic variants, represent an advance in our ability to predict the development and complications of cardiometabolic diseases. This article reviews the evidence supporting the use of PRS in seven common cardiometabolic diseases: coronary artery disease (CAD), stroke, hypertension, heart failure and cardiomyopathies, obesity, atrial fibrillation (AF), and type 2 diabetes mellitus (T2DM). Data suggest that PRS for CAD, AF, and T2DM consistently improves prediction when incorporated into existing clinical risk tools. In other areas such as ischaemic stroke and hypertension, clinical application appears premature but emerging evidence suggests that the study of larger and more diverse populations coupled with more granular phenotyping will propel the translation of PRS into practical clinical prediction tools.
Collapse
Affiliation(s)
- Jack W O'Sullivan
- Stanford Center for Inherited Cardiovascular Disease, Stanford University School of Medicine, Stanford, CA, USA
- Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Euan A Ashley
- Stanford Center for Inherited Cardiovascular Disease, Stanford University School of Medicine, Stanford, CA, USA
- Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Perry M Elliott
- UCL Institute of Cardiovascular Science, Gower Street, London WC1E 6BT, UK
- St. Bartholomew's Hospital, W Smithfield, London EC1A 7BE, UK
| |
Collapse
|
14
|
Muller YL, Saporito M, Day S, Bandesh K, Koroglu C, Kobes S, Knowler WC, Hanson RL, Van Hout CV, Shuldiner AR, Bogardus C, Baier LJ. Functional characterization of a novel p.Ser76Thr variant in IGFBP4 that associates with body mass index in American Indians. Eur J Hum Genet 2022; 30:1159-1166. [PMID: 35688891 PMCID: PMC9554187 DOI: 10.1038/s41431-022-01129-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 04/18/2022] [Accepted: 05/26/2022] [Indexed: 12/15/2022] Open
Abstract
Insulin-like growth factor binding protein 4 (IGFBP4) is involved in adipogenesis, and IGFBP4 null mice have decreased body fat through decreased PPAR-γ expression. In the current study, we assessed whether variation in the IGFBP4 coding region influences body mass index (BMI) in American Indians who are disproportionately affected by obesity. Whole exome sequence data from a population-based sample of 6779 American Indians with longitudinal measures of BMI were used to identify variation in IGFBP4 that associated with BMI. A novel variant that predicts a p.Ser76Thr in IGFBP4 (Thr-allele frequency = 0.02) was identified which associated with the maximum BMI measured during adulthood (BMI 39.8 kg/m2 for Thr-allele homozygotes combined with heterozygotes vs. 36.2 kg/m2 for Ser-allele homozygotes, β = 6.7% per Thr-allele, p = 8.0 × 10-5, adjusted for age, sex, birth-year and the first five genetic principal components) and the maximum age- and sex-adjusted BMI z-score measured during childhood/adolescence (z-score 0.70 SD for Thr-allele heterozygotes vs. 0.32 SD for Ser-allele homozygotes, β = 0.37 SD per Thr-allele, p = 8.8 × 10-6). In vitro functional studies showed that IGFBP4 with the Thr-allele (BMI-increasing) had a 55% decrease (p = 0.0007) in FOXO-induced transcriptional activity, reflecting increased activation of the PI3K/AKT pathway mediated through increased IGF signaling. Over-expression and knock-down of IGFBP4 in OP9 cells during differentiation showed that IGFBP4 upregulates adipogenesis through PPARγ, CEBPα, AGPAT2 and SREBP1 expression. We propose that this American Indian specific variant in IGFBP4 affects obesity via an increase of IGF signaling.
Collapse
Affiliation(s)
- Yunhua L Muller
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ, USA.
| | - Michael Saporito
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ, USA
| | - Samantha Day
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ, USA
| | - Khushdeep Bandesh
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ, USA
| | - Cigdem Koroglu
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ, USA
| | - Sayuko Kobes
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ, USA
| | - William C Knowler
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ, USA
| | - Robert L Hanson
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ, USA
| | - Cristopher V Van Hout
- Regeneron Genetics Center, Regeneron Pharmaceuticals, Tarrytown, NY, USA
- Laboratorio Internacional de Investigation sobre el Genoma Humano, Campus Juriquilla de la Universidad Nacional Autonoma de Mexico, Queretaro, QRO, Mexico
| | - Alan R Shuldiner
- Regeneron Genetics Center, Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Clifton Bogardus
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ, USA
| | - Leslie J Baier
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ, USA
| |
Collapse
|
15
|
Miola A, De Filippis E, Veldic M, Ho AMC, Winham SJ, Mendoza M, Romo-Nava F, Nunez NA, Gardea Resendez M, Prieto ML, McElroy SL, Biernacka JM, Frye MA, Cuellar-Barboza AB. The genetics of bipolar disorder with obesity and type 2 diabetes. J Affect Disord 2022; 313:222-231. [PMID: 35780966 PMCID: PMC9703971 DOI: 10.1016/j.jad.2022.06.084] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 06/25/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Bipolar disorder (BD) presents with high obesity and type 2 diabetes (T2D) and pathophysiological and phenomenological abnormalities shared with cardiometabolic disorders. Genomic studies may help define if they share genetic liability. This selective review of BD with obesity and T2D will focus on genomic studies, stress their current limitations and guide future steps in developing the field. METHODS We searched electronic databases (PubMed, Scopus) until December 2021 to identify genome-wide association studies, polygenic risk score analyses, and functional genomics of BD accounting for body mass index (BMI), obesity, or T2D. RESULTS The first genome-wide association studies (GWAS) of BD accounting for obesity found a promising genome-wide association in an intronic gene variant of TCF7L2 that was further replicated. Polygenic risk scores of obesity and T2D have also been associated with BD, yet, no genetic correlations have been demonstrated. Finally, human-induced stem cell studies of the intronic variant in TCF7L2 show a potential biological impact of the products of this genetic variant in BD risk. LIMITATIONS The narrative nature of this review. CONCLUSIONS Findings from BD GWAS accounting for obesity and their functional testing, have prompted potential biological insights. Yet, BD, obesity, and T2D display high phenotypic, genetic, and population-related heterogeneity, limiting our ability to detect genetic associations. Further studies should refine cardiometabolic phenotypes, test gene-environmental interactions and add population diversity.
Collapse
Affiliation(s)
- Alessandro Miola
- Department of Neuroscience (DNS), University of Padova, Padua, Italy
| | | | - Marin Veldic
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, MN, USA
| | - Ada Man-Choi Ho
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, MN, USA
| | - Stacey J Winham
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Mariana Mendoza
- Department of Psychiatry, Universidad Autonoma de Nuevo Leon, Monterrey, Mexico
| | - Francisco Romo-Nava
- Lindner Center of HOPE, Mason, OH, USA; Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Nicolas A Nunez
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, MN, USA
| | | | - Miguel L Prieto
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, MN, USA; Department of Psychiatry, Facultad de Medicina, Universidad de los Andes, Santiago, Chile; Mental Health Service, Clínica Universidad de los Andes, Santiago, Chile; Center for Biomedical Research and Innovation, Universidad de los Andes, Santiago, Chile
| | - Susan L McElroy
- Lindner Center of HOPE, Mason, OH, USA; Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Joanna M Biernacka
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, MN, USA; Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Mark A Frye
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, MN, USA
| | - Alfredo B Cuellar-Barboza
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, MN, USA; Department of Psychiatry, Universidad Autonoma de Nuevo Leon, Monterrey, Mexico.
| |
Collapse
|
16
|
Speakman JR, Elmquist JK. Obesity: an evolutionary context. LIFE METABOLISM 2022; 1:10-24. [PMID: 36394061 PMCID: PMC9642988 DOI: 10.1093/lifemeta/loac002] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/21/2022] [Accepted: 03/09/2022] [Indexed: 05/07/2023]
Abstract
People completely lacking body fat (lipodystrophy/lipoatrophy) and those with severe obesity both show profound metabolic and other health issues. Regulating levels of body fat somewhere between these limits would, therefore, appear to be adaptive. Two different models might be contemplated. More traditional is a set point (SP) where the levels are regulated around a fixed level. Alternatively, dual-intervention point (DIP) is a system that tolerates fairly wide variation but is activated when critically high or low levels are breached. The DIP system seems to fit our experience much better than an SP, and models suggest that it is more likely to have evolved. A DIP system may have evolved because of two contrasting selection pressures. At the lower end, we may have been selected to avoid low levels of fat as a buffer against starvation, to avoid disease-induced anorexia, and to support reproduction. At the upper end, we may have been selected to avoid excess storage because of the elevated risks of predation. This upper limit of control seems to have malfunctioned because some of us deposit large fat stores, with important negative health effects. Why has evolution not protected us against this problem? One possibility is that the protective system slowly fell apart due to random mutations after we dramatically reduced the risk of being predated during our evolutionary history. By chance, it fell apart more in some people than others, and these people are now unable to effectively manage their weight in the face of the modern food glut. To understand the evolutionary context of obesity, it is important to separate the adaptive reason for storing some fat (i.e. the lower intervention point), from the nonadaptive reason for storing lots of fat (a broken upper intervention point). The DIP model has several consequences, showing how we understand the obesity problem and what happens when we attempt to treat it.
Collapse
Affiliation(s)
- John R Speakman
- Shenzhen Key Laboratory of Metabolic Health, Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- School of Biological Sciences, University of Aberdeen, Aberdeen, UK
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental biology, Chinese Academy of Sciences, Beijing, China
- CAS Center of Excellence in Animal Evolution and Genetics, Kunming, China
| | - Joel K Elmquist
- Departments of Internal Medicine and Pharmacology, Center for Hypothalamic Research, University of Texas Southwestern, Dallas, TX, USA
| |
Collapse
|
17
|
Ontawong A, Duangjai A, Sukpondma Y, Tadpetch K, Muanprasat C, Rukachaisirikul V, Inchai J, Vaddhanaphuti CS. Cholesterol-Lowering Effects of Asperidine B, a Pyrrolidine Derivative from the Soil-Derived Fungus Aspergillus sclerotiorum PSU-RSPG178: A Potential Cholesterol Absorption Inhibitor. Pharmaceuticals (Basel) 2022; 15:ph15080955. [PMID: 36015103 PMCID: PMC9414821 DOI: 10.3390/ph15080955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/21/2022] [Accepted: 07/29/2022] [Indexed: 11/30/2022] Open
Abstract
Isolated secondary metabolites asperidine B (preussin) and asperidine C, produced by the soil-derived fungus Aspergillus sclerotiorum PSU-RSPG178, were found to exhibit inhibitory effects against 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase and oxidative stress in an in vitro assay. Whether or not the known pyrrolidine asperidine B and the recently isolated piperidine asperidine C have lipid-lowering effects remains unknown. Thus, this study aimed to investigate the hypocholesterolemic effects of asperidines B and C and identify the mechanisms involved in using in vitro, ex vivo, and in vivo models. The results show that both compounds interfered with cholesterol micelle formation by increasing bile acid binding capacity, similar to the action of the bile acid sequestrant drug cholestyramine. However, only asperidine B, but not asperidine C, was found to inhibit cholesterol uptake in Caco-2 cells by up-regulating LXRα without changing cholesterol transporter NPC1L1 protein expression. Likewise, reduced cholesterol absorption via asperidine-B-mediated activation of LXRα was also observed in isolated rat jejunal loops. Asperidine B consistently decreases plasma cholesterol absorption, similar to the effect of ezetimibe in rats. Therefore, asperidine B, the pyrrolidine derivative, has therapeutic potential to be developed into a type of cholesterol absorption inhibitor for the treatment of hypercholesterolemia.
Collapse
Affiliation(s)
- Atcharaporn Ontawong
- Division of Physiology, School of Medical Sciences, University of Phayao, Phayao 56000, Thailand; (A.O.); (A.D.)
| | - Acharaporn Duangjai
- Division of Physiology, School of Medical Sciences, University of Phayao, Phayao 56000, Thailand; (A.O.); (A.D.)
| | - Yaowapa Sukpondma
- Division of Physical Science and Center of Excellence for Innovation in Chemistry, Faculty of Science, Prince of Songkla University, Songkhla 90112, Thailand; (Y.S.); (K.T.); (V.R.)
| | - Kwanruthai Tadpetch
- Division of Physical Science and Center of Excellence for Innovation in Chemistry, Faculty of Science, Prince of Songkla University, Songkhla 90112, Thailand; (Y.S.); (K.T.); (V.R.)
| | - Chatchai Muanprasat
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10540, Thailand;
| | - Vatcharin Rukachaisirikul
- Division of Physical Science and Center of Excellence for Innovation in Chemistry, Faculty of Science, Prince of Songkla University, Songkhla 90112, Thailand; (Y.S.); (K.T.); (V.R.)
| | - Jakkapong Inchai
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Chutima S. Vaddhanaphuti
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand;
- Correspondence: ; Tel.:+66-533-935-362
| |
Collapse
|
18
|
Wu Y, Hu S, Yang D, Li L, Li B, Wang L, Li M, Wang G, Li J, Xu Y, Zhang X, Niu C, Speakman JR. Increased Variation in Body Weight and Food Intake Is Related to Increased Dietary Fat but Not Increased Carbohydrate or Protein in Mice. Front Nutr 2022; 9:835536. [PMID: 35360679 PMCID: PMC8963818 DOI: 10.3389/fnut.2022.835536] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 01/28/2022] [Indexed: 12/23/2022] Open
Abstract
A variety of inbred mouse strains have been used for research in metabolic disorders. Despite being inbred, they display large inter-individual variability for many traits like food intake and body weight. However, the relationship between dietary macronutrients and inter-individual variation in body weight and food intake of different mouse strains is still unclear. We investigated the association between macronutrient content of the diet and variations in food intake, body composition, and glucose tolerance by exposing five different mouse strains (C57BL/6, BALB/c, C3H, DBA/2, and FVB) to 24 different diets with variable protein, fat, and carbohydrate contents. We found only increasing dietary fat, but not protein or carbohydrate had a significant association (positive) with variation in both food intake and body weight. The highest variation in both body weight and food intake occurred with 50% dietary fat. However, there were no significant relationships between the variation in fat and lean mass with dietary protein, fat, or carbohydrate levels. In addition, none of the dietary macronutrients had significant impacts on the variation in glucose tolerance ability in C57BL/6 mice. In conclusion, the variations in food intake and body weight changes increased with the elevation of dietary fat levels.
Collapse
Affiliation(s)
- Yingga Wu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Sumei Hu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, National Soybean Processing Industry Technology Innovation Center, Beijing Technology and Business University, Beijing, China
| | - Dengbao Yang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Li Li
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Baoguo Li
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Lu Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Min Li
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, United Kingdom
- Shenzhen Key Laboratory of Metabolic Health, Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Guanlin Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | | | - Yanchao Xu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Xueying Zhang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, United Kingdom
- Shenzhen Key Laboratory of Metabolic Health, Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Chaoqun Niu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- Shenzhen Key Laboratory of Metabolic Health, Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - John R. Speakman
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, United Kingdom
- Shenzhen Key Laboratory of Metabolic Health, Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- CAS Center of Excellence in Animal Evolution and Genetics, Kunming, China
- *Correspondence: John R. Speakman
| |
Collapse
|
19
|
Ahluwalia MK. Nutrigenetics and nutrigenomics-A personalized approach to nutrition. ADVANCES IN GENETICS 2021; 108:277-340. [PMID: 34844714 DOI: 10.1016/bs.adgen.2021.08.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The prevalence of non-communicable diseases has been on an upward trajectory for some time and this puts an enormous burden on the healthcare expenditure. Lifestyle modifications including dietary interventions hold an immense promise to manage and prevent these diseases. Recent advances in genomic research provide evidence that focussing these efforts on individual variations in abilities to metabolize nutrients (nutrigenetics) and exploring the role of dietary compounds on gene expression (nutrigenomics and nutri-epigenomics) can lead to more meaningful personalized dietary strategies to promote optimal health. This chapter aims to provide examples on these gene-diet interactions at multiple levels to support the need of embedding targeted dietary interventions as a way forward to prevent, avoid and manage diseases.
Collapse
|
20
|
Hulme KD, Noye EC, Short KR, Labzin LI. Dysregulated Inflammation During Obesity: Driving Disease Severity in Influenza Virus and SARS-CoV-2 Infections. Front Immunol 2021; 12:770066. [PMID: 34777390 PMCID: PMC8581451 DOI: 10.3389/fimmu.2021.770066] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 09/30/2021] [Indexed: 12/15/2022] Open
Abstract
Acute inflammation is a critical host defense response during viral infection. When dysregulated, inflammation drives immunopathology and tissue damage. Excessive, damaging inflammation is a hallmark of both pandemic influenza A virus (IAV) infections and Severe Acute Respiratory Syndrome-Coronavirus-2 (SARS-CoV-2) infections. Chronic, low-grade inflammation is also a feature of obesity. In recent years, obesity has been recognized as a growing pandemic with significant mortality and associated costs. Obesity is also an independent risk factor for increased disease severity and death during both IAV and SARS-CoV-2 infection. This review focuses on the effect of obesity on the inflammatory response in the context of viral respiratory infections and how this leads to increased viral pathology. Here, we will review the fundamentals of inflammation, how it is initiated in IAV and SARS-CoV-2 infection and its link to disease severity. We will examine how obesity drives chronic inflammation and trained immunity and how these impact the immune response to IAV and SARS-CoV-2. Finally, we review both medical and non-medical interventions for obesity, how they impact on the inflammatory response and how they could be used to prevent disease severity in obese patients. As projections of global obesity numbers show no sign of slowing down, future pandemic preparedness will require us to consider the metabolic health of the population. Furthermore, if weight-loss alone is insufficient to reduce the risk of increased respiratory virus-related mortality, closer attention must be paid to a patient’s history of health, and new therapeutic options identified.
Collapse
Affiliation(s)
- Katina D Hulme
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| | - Ellesandra C Noye
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| | - Kirsty R Short
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia.,Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia
| | - Larisa I Labzin
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia.,Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
21
|
de Ceglia M, Decara J, Gaetani S, Rodríguez de Fonseca F. Obesity as a Condition Determined by Food Addiction: Should Brain Endocannabinoid System Alterations Be the Cause and Its Modulation the Solution? Pharmaceuticals (Basel) 2021; 14:ph14101002. [PMID: 34681224 PMCID: PMC8538206 DOI: 10.3390/ph14101002] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/24/2021] [Accepted: 09/26/2021] [Indexed: 12/11/2022] Open
Abstract
Obesity is a complex disorder, and the number of people affected is growing every day. In recent years, research has confirmed the hypothesis that food addiction is a determining factor in obesity. Food addiction is a behavioral disorder characterized by disruptions in the reward system in response to hedonic eating. The endocannabinoid system (ECS) plays an important role in the central and peripheral control of food intake and reward-related behaviors. Moreover, both obesity and food addiction have been linked to impairments in the ECS function in various brain regions integrating peripheral metabolic signals and modulating appetite. For these reasons, targeting the ECS could be a valid pharmacological therapy for these pathologies. However, targeting the cannabinoid receptors with inverse agonists failed when used in clinical contexts as a consequence of the induction of affective disorders. In this context, new classes of drugs acting either on CB1 and/or CB2 receptors or on synthetic and degradation enzymes of endogenous cannabinoids are being studied. However, further investigation is necessary to find safe and effective treatments that can exert anti-obesity effects, normalizing reward-related behaviors without causing important adverse mood effects.
Collapse
Affiliation(s)
- Marialuisa de Ceglia
- UGC Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga-Hospital Universitario Regional de Málaga, 29010 Málaga, Spain;
- Correspondence: (M.d.C.); (F.R.d.F.)
| | - Juan Decara
- UGC Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga-Hospital Universitario Regional de Málaga, 29010 Málaga, Spain;
| | - Silvana Gaetani
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University of Rome, 00185 Rome, Italy;
| | - Fernando Rodríguez de Fonseca
- UGC Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga-Hospital Universitario Regional de Málaga, 29010 Málaga, Spain;
- Correspondence: (M.d.C.); (F.R.d.F.)
| |
Collapse
|
22
|
Lee DH, Ku EJ, Oh TK, Jeon HJ. RANTES 59029A/G Polymorphisms Associated with Diabetic Compilations in Korean Patients with Type 2 Diabetes for over 15 Years. Genes (Basel) 2021; 12:genes12091445. [PMID: 34573427 PMCID: PMC8468929 DOI: 10.3390/genes12091445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/14/2021] [Accepted: 09/18/2021] [Indexed: 11/25/2022] Open
Abstract
Background: Polymorphisms in the RANTES gene are known to be associated with several diseases related to insulin resistance. In this study, we investigated the association between RANTES 59029A/G polymorphisms and the prevalence of diabetic complications relative to obesity in Korean patients who had type 2 diabetes (T2D) for over 15 years. Methods: A single-center, retrospective case-control study was performed. We included 271 patients with a duration of diabetes greater than 15 years. Polymerase chain reaction-restriction fragment length polymorphism was used to analyze RANTES polymorphisms, identifying genotypes as GG, AG, or AA. Obesity was defined using the body mass index with a cutoff value of 25 kg/m2. Both microvascular (retinopathy and nephropathy) and macrovascular (coronary artery disease and cerebrovascular disease) complications were evaluated. Results: The duration of T2D and hemoglobin A1c values at enrollment were 24.4 ± 5.0 years and 7.8 ± 1.6%, respectively, in the non-obese group, and 25.4 ± 6.1 years and 7.7 ± 1.7%, respectively, in the obese group. The prevalence of microvascular complications was significantly higher in the obese group compared with that in the non-obese group (83.5% vs. 72.0%, p = 0.039). Compared to the non-obese group, the obese group showed a higher proportion of the patients with AA or AG genotypes (64.3% vs. 84.5%, p = 0.001). Conclusions: The A allele of the RANTES gene is associated with obesity and may affect diabetic microvascular complications in patients with T2D for over 15 years.
Collapse
Affiliation(s)
| | | | | | - Hyun-Jeong Jeon
- Correspondence: ; Tel.: +82-43-269-6352; Fax: +82-43-273-3252
| |
Collapse
|
23
|
Ding M, Zheng L, Li QF, Wang WL, Peng WD, Zhou M. Exercise-Training Regulates Apolipoprotein B in Drosophila to Improve HFD-Mediated Cardiac Function Damage and Low Exercise Capacity. Front Physiol 2021; 12:650959. [PMID: 34305631 PMCID: PMC8294119 DOI: 10.3389/fphys.2021.650959] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 05/21/2021] [Indexed: 12/02/2022] Open
Abstract
Apolipoprotein B plays an essential role in systemic lipid metabolism, and it is closely related to cardiovascular diseases. Exercise-training can regulate systemic lipid metabolism, improve heart function, and improve exercise capacity, but the molecular mechanisms involved are poorly understood. We used a Drosophila model to demonstrate that exercise-training regulates the expression of apoLpp (a homolog of apolipoprotein B) in cardiomyocytes, thereby resisting heart insufficiency and low exercise capacity caused by obesity. The apoLpp is an essential lipid carrier produced in the heart and fat body of Drosophila. In a Drosophila genetic screen, low expression of apoLpp reduced obesity and cardiac dysfunction induced by a high-fat diet (HFD). Cardiac-specific inhibition indicated that reducing apoLpp in the heart during HFD reduced the triglyceride content of the whole-body and reduced heart function damage caused by HFD. In exercise-trained flies, the result was similar to the knockdown effect of apoLpp. Therefore, the inhibition of apoLpp plays an important role in HFD-induced cardiac function impairment and low exercise capacity. Although the apoLpp knockdown of cardiomyocytes alleviated damage to heart function, it did not reduce the arrhythmia and low exercise capacity caused by HFD. Exercise-training can improve this condition more effectively, and the possible reason for this difference is that exercise-training regulates climbing ability in ways to promote metabolism. Exercise-training during HFD feeding can down-regulate the expression of apoLpp, reduce the whole-body TG levels, improve cardiac recovery, and improve exercise capacity. Exercise-training can downregulate the expression of apoLpp in cardiomyocytes to resist cardiac function damage and low exercise capacity caused by HFD. The results revealed the relationship between exercise-training and apoLpp and their essential roles in regulating heart function and climbing ability.
Collapse
Affiliation(s)
- Meng Ding
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, Hunan Normal University, Changsha, China
| | - Lan Zheng
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, Hunan Normal University, Changsha, China
| | - Qiu Fang Li
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, Hunan Normal University, Changsha, China
| | - Wan Li Wang
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, Hunan Normal University, Changsha, China
| | - Wan Da Peng
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, Hunan Normal University, Changsha, China
| | - Meng Zhou
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, Hunan Normal University, Changsha, China
| |
Collapse
|
24
|
Rana S, Bhatti AA. Predicting anthropometric and metabolic traits with a genetic risk score for obesity in a sample of Pakistanis. Sci Rep 2021; 11:8320. [PMID: 33859285 PMCID: PMC8050295 DOI: 10.1038/s41598-021-87702-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 04/01/2021] [Indexed: 12/18/2022] Open
Abstract
Obesity is an outcome of multiple factors including environmental and genetic influences. Common obesity is a polygenic trait indicating that multiple genetic variants act synergistically to influence its expression. We constructed a genetic risk score (GRS) based on five genetic variants (MC4R rs17782313, BDNF rs6265, FTO rs1421085, TMEM18 rs7561317, and NEGR1 rs2815752) and examined its association with obesity-related traits in a sample of Pakistanis. The study involved 306 overweight/obese (OW/OB) and 300 normal-weight (NW) individuals. The age range of the study participants was 12-63 years. All anthropometric and metabolic parameters were measured for each participant via standard procedures and biochemical assays, respectively. The genetic variants were genotyped by allelic discrimination assays. The age- and gender-adjusted associations between the GRS and obesity-related anthropometric and metabolic measures were determined using linear regression analyses. The results showed that OW/OB individuals had significantly higher mean ranks of GRS than NW individuals. Moreover, a significant association of the GRS with obesity-related anthropometric traits was seen. However, the GRS did not appear to affect any obesity-related metabolic parameter. In conclusion, our findings indicate the combined effect of multiple genetic variants on the obesity-related anthropometric phenotypes in Pakistanis.
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.
| | - 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
|
25
|
Dessein A. Clinical utility of polygenic risk scores for predicting NAFLD disorders. J Hepatol 2021; 74:769-770. [PMID: 33653592 DOI: 10.1016/j.jhep.2021.02.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 02/04/2021] [Accepted: 02/14/2021] [Indexed: 01/03/2023]
Affiliation(s)
- Alain Dessein
- GENEPRED Biotechnologies, Bilhigenetics, Marseille, France.
| |
Collapse
|
26
|
Zaghlool SB, Sharma S, Molnar M, Matías-García PR, Elhadad MA, Waldenberger M, Peters A, Rathmann W, Graumann J, Gieger C, Grallert H, Suhre K. Revealing the role of the human blood plasma proteome in obesity using genetic drivers. Nat Commun 2021; 12:1279. [PMID: 33627659 PMCID: PMC7904950 DOI: 10.1038/s41467-021-21542-4] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 01/29/2021] [Indexed: 12/21/2022] Open
Abstract
Blood circulating proteins are confounded readouts of the biological processes that occur in different tissues and organs. Many proteins have been linked to complex disorders and are also under substantial genetic control. Here, we investigate the associations between over 1000 blood circulating proteins and body mass index (BMI) in three studies including over 4600 participants. We show that BMI is associated with widespread changes in the plasma proteome. We observe 152 replicated protein associations with BMI. 24 proteins also associate with a genome-wide polygenic score (GPS) for BMI. These proteins are involved in lipid metabolism and inflammatory pathways impacting clinically relevant pathways of adiposity. Mendelian randomization suggests a bi-directional causal relationship of BMI with LEPR/LEP, IGFBP1, and WFIKKN2, a protein-to-BMI relationship for AGER, DPT, and CTSA, and a BMI-to-protein relationship for another 21 proteins. Combined with animal model and tissue-specific gene expression data, our findings suggest potential therapeutic targets further elucidating the role of these proteins in obesity associated pathologies.
Collapse
Affiliation(s)
- Shaza B Zaghlool
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Sapna Sharma
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Bavaria, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Bavaria, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Megan Molnar
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Bavaria, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Bavaria, Germany
| | - Pamela R Matías-García
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Bavaria, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Bavaria, Germany
- TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Mohamed A Elhadad
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Bavaria, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Bavaria, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Melanie Waldenberger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Bavaria, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Bavaria, Germany
- German Research Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Bavaria, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- German Research Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Wolfgang Rathmann
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute of Biometrics and Epidemiology, German Diabetes Center, Düsseldorf, Germany
| | - Johannes Graumann
- Scientific Service Group Biomolecular Mass Spectrometry, Max Planck Institute for Heart and Lung Research, W.G. Kerckhoff Institute, Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Max Planck Institute of Heart and Lung Research, Bad Nauheim, Germany
| | - Christian Gieger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Bavaria, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Bavaria, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Harald Grallert
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Bavaria, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Bavaria, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Karsten Suhre
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Doha, Qatar.
| |
Collapse
|
27
|
Khattab A, Ahmed-Farid O, Nasr S. Enhanced brain biodistribution of Ginsenoside Rg1 based self-nanoemulsifying drug delivery system to ameliorate metabolic syndromes and keep homeostatic balance. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2020.102276] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
28
|
Fujimi TJ, Sate M, Tsuchiya M, Hirono M, Asahi R, Suzuki R, Nakajima S, Yokoyama H, Matsuura T, Kanzawa N. Gene Expression and Histochemical Analyses in the Fatty Livers of Rats Fed a Histidine-Excess Diet. J Nutr Sci Vitaminol (Tokyo) 2021; 66:561-570. [PMID: 33390398 DOI: 10.3177/jnsv.66.561] [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] [Indexed: 11/27/2022]
Abstract
Triglyceride (TG) and cholesterol accumulation are known to occur in the liver of rats fed a histidine-excess (5%) diet, but there are few studies reporting histochemical and molecular biological analyses of the rat liver. The aim of this study was to elucidate the molecular basis of this lipid-accumulation mechanism. Lipid accumulations, tissue section images, and gene expression levels were compared in the livers of rats fed a control or histidine-excess diet for 5 wk (n=8/group). Serum levels of TGs, free fatty acids, total cholesterol, high-density lipoprotein cholesterol, glucose, albumin, and the enzyme activities of aspartate aminotransferase and alanine aminotransferase were also analyzed. In the livers of rats fed a histidine-excess diet, histochemical analyses showed what appeared to be a preliminary stage of nonalcoholic fatty liver, characterized by lipid accumulation around the central vein area and minor fibrosis. However, there were no changes in serum TG or free fatty acid levels. Quantitative PCR analyses showed the up-regulation of FAT/CD36, which is related to the uptake of fatty acids into cells, and the downregulation of two apolipoprotein genes, ApoC3 and ApoE. The mRNA levels of PPARγ, LXRα, and AMPKα in the liver were also reduced by excess histidine intake. The results of this study suggest that steatosis caused by excess histidine intake may be the result of an imbalance between lipid transport from the liver and the uptake of free fatty acids into hepatocytes.
Collapse
Affiliation(s)
- Takahiko J Fujimi
- Department of Registered Dietitians, Faculty of Health and Nutrition, Bunkyo University
| | - Mikako Sate
- Department of Registered Dietitians, Faculty of Health and Nutrition, Bunkyo University
| | - Minori Tsuchiya
- Department of Registered Dietitians, Faculty of Health and Nutrition, Bunkyo University
| | - Mayuko Hirono
- Department of Registered Dietitians, Faculty of Health and Nutrition, Bunkyo University
| | - Riku Asahi
- Department of Registered Dietitians, Faculty of Health and Nutrition, Bunkyo University
| | - Ryuta Suzuki
- Department of Registered Dietitians, Faculty of Health and Nutrition, Bunkyo University
| | - Shigeru Nakajima
- Department of Registered Dietitians, Faculty of Health and Nutrition, Bunkyo University
| | - Hiroshi Yokoyama
- Department of Laboratory Medicine, The Jikei University School of Medicine
| | - Tomokazu Matsuura
- Department of Laboratory Medicine, The Jikei University School of Medicine
| | - Nobuyuki Kanzawa
- Department of Material and Life Science, Faculty of Science and Technology, Sophia University
| |
Collapse
|
29
|
Nam SW, Choi J, Jeon HJ, Oh TK, Lee DH. The Associations Between Vitamin D Receptor BsmI and ApaI Polymorphisms and Obesity in Korean Patients with Type 2 Diabetes Mellitus. Diabetes Metab Syndr Obes 2021; 14:557-564. [PMID: 33603421 PMCID: PMC7882455 DOI: 10.2147/dmso.s293032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 01/27/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Vitamin D receptor (VDR) polymorphisms are associated with osteoporosis, diabetes, immunological diseases, and cancers. However, the association of obesity with VDR polymorphisms has shown inconsistent results, and perhaps it depends upon the characteristics of a population. Therefore, we evaluated the association between BsmI (rs1544410) and ApaI (rs7975232) polymorphisms of VDR and obesity in Korean patients with type 2 diabetes mellitus (T2DM). METHODS A total of 506 patients with T2DM participated in the study. Polymerase chain reaction-restriction fragment length polymorphism was used to analyze BsmI and ApaI polymorphisms; the genotypes were presented as BB, Bb, or bb for BsmI and AA, Aa, or aa for ApaI. Obesity was defined using the body mass index (BMI) with a cutoff level of 25 kg/m2. RESULTS The prevalence of obesity was higher in patients with the bb genotype than in those with BB or Bb genotypes (48.4% vs 33.9%, P = 0.031). The mean BMI was 25.2 ± 3.5 kg/m2 in patients with bb genotype and 24.1 ± 3.1 kg/m2 in patients with BB or Bb genotypes. Patients with Aa or aa genotypes showed a higher prevalence of obesity than patients with AA genotype (47.6% vs 26.1%, P = 0.043). Glycemic control parameters and lipid profiles did not show significant differences with either polymorphism. CONCLUSION To our knowledge, this is the first study to assess the association between VDR polymorphisms and obesity in Korean patients with T2DM. Further studies in larger populations and multiethnic cohorts are needed to validate our findings.
Collapse
Affiliation(s)
- Sang Won Nam
- Department of Internal Medicine, Chungbuk National University Hospital, Cheongju, South Korea
| | - Jinwoo Choi
- Department of Internal Medicine, Chungbuk National University Hospital, Cheongju, South Korea
| | - Hyun Jeong Jeon
- Department of Internal Medicine, Chungbuk National University College of Medicine, Cheongju, South Korea
| | - Tae Keun Oh
- Department of Internal Medicine, Chungbuk National University College of Medicine, Cheongju, South Korea
| | - Dong-Hwa Lee
- Department of Internal Medicine, Chungbuk National University Hospital, Cheongju, South Korea
- Correspondence: Dong-Hwa Lee Department of Internal Medicine, Chungbuk National University Hospital, 776, 1sunhwan-Ro, Seowon-Gu, Cheongju-si, Chungcheongbuk-do, 28644, South KoreaTel +82-43-269-7504Fax +82-43-273-3252 Email
| |
Collapse
|
30
|
Chalmers J, Tung YCL, Liu CH, O'Kane CJ, O'Rahilly S, Yeo GSH. A multicomponent screen for feeding behaviour and nutritional status in Drosophila to interrogate mammalian appetite-related genes. Mol Metab 2021; 43:101127. [PMID: 33242659 PMCID: PMC7753202 DOI: 10.1016/j.molmet.2020.101127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/18/2020] [Accepted: 11/19/2020] [Indexed: 02/08/2023] Open
Abstract
OBJECTIVE More than 300 genetic variants have been robustly associated with measures of human adiposity. Highly penetrant mutations causing human obesity do so largely by disrupting satiety pathways in the brain and increasing food intake. Most of the common obesity-predisposing variants are in, or near, genes expressed highly in the brain, but little is known of their function. Exploring the biology of these genes at scale in mammalian systems is challenging. We sought to establish and validate the use of a multicomponent screen for feeding behaviour phenotypes, taking advantage of the tractable model organism Drosophila melanogaster. METHODS We validated a screen for feeding behaviour in Drosophila by comparing results after disrupting the expression of centrally expressed genes that influence energy balance in flies to those of 10 control genes. We then used this screen to explore the effects of disrupted expression of genes either a) implicated in energy homeostasis through human genome-wide association studies (GWAS) or b) expressed and nutritionally responsive in specific populations of hypothalamic neurons with a known role in feeding/fasting. RESULTS Using data from the validation study to classify responses, we studied 53 Drosophila orthologues of genes implicated by human GWAS in body mass index and found that 15 significantly influenced feeding behaviour or energy homeostasis in the Drosophila screen. We then studied 50 Drosophila homologues of 47 murine genes reciprocally nutritionally regulated in POMC and agouti-related peptide neurons. Seven of these 50 genes were found by our screen to influence feeding behaviour in flies. CONCLUSION We demonstrated the utility of Drosophila as a tractable model organism in a high-throughput genetic screen for food intake phenotypes. This simple, cost-efficient strategy is ideal for high-throughput interrogation of genes implicated in feeding behaviour and obesity in mammals and will facilitate the process of reaching a functional understanding of obesity pathogenesis.
Collapse
Affiliation(s)
- J Chalmers
- Medical Research Council (MRC) Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK.
| | - Y C L Tung
- Medical Research Council (MRC) Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK.
| | - C H Liu
- Department of Physiology, Development and Neuroscience, Cambridge University, Downing St, Cambridge, CB2 3EG, UK.
| | - C J O'Kane
- Department of Genetics, University of Cambridge, Downing Street, Cambridge, CB2 3EH, UK.
| | - S O'Rahilly
- Medical Research Council (MRC) Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK.
| | - G S H Yeo
- Medical Research Council (MRC) Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK.
| |
Collapse
|
31
|
Herring Milt and Herring Milt Protein Hydrolysate Are Equally Effective in Improving Insulin Sensitivity and Pancreatic Beta-Cell Function in Diet-Induced Obese- and Insulin-Resistant Mice. Mar Drugs 2020; 18:md18120635. [PMID: 33322303 PMCID: PMC7763884 DOI: 10.3390/md18120635] [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: 10/24/2020] [Revised: 12/02/2020] [Accepted: 12/08/2020] [Indexed: 02/06/2023] Open
Abstract
Although genetic predisposition influences the onset and progression of insulin resistance and diabetes, dietary nutrients are critical. In general, protein is beneficial relative to carbohydrate and fat but dependent on protein source. Our recent study demonstrated that 70% replacement of dietary casein protein with the equivalent quantity of protein derived from herring milt protein hydrolysate (HMPH; herring milt with proteins being enzymatically hydrolyzed) significantly improved insulin resistance and glucose homeostasis in high-fat diet-induced obese mice. As production of protein hydrolysate increases the cost of the product, it is important to determine whether a simply dried and ground herring milt product possesses similar benefits. Therefore, the current study was conducted to investigate the effect of herring milt dry powder (HMDP) on glucose control and the associated metabolic phenotypes and further to compare its efficacy with HMPH. Male C57BL/6J mice on a high-fat diet for 7 weeks were randomized based on body weight and blood glucose into three groups. One group continued on the high-fat diet and was used as the insulin-resistant/diabetic control and the other two groups were given the high-fat diet modified to have 70% of casein protein being replaced with the same amount of protein from HMDP or HMPH. A group of mice on a low-fat diet all the time was used as the normal control. The results demonstrated that mice on the high-fat diet increased weight gain and showed higher blood concentrations of glucose, insulin, and leptin, as well as impaired glucose tolerance and pancreatic β-cell function relative to those on the normal control diet. In comparison with the high-fat diet, the replacement of 70% dietary casein protein with the same amount of HMDP or HMPH protein decreased weight gain and significantly improved the aforementioned biomarkers, insulin sensitivity or resistance, and β-cell function. The HMDP and HMPH showed similar effects on every parameter except blood lipids where HMDP decreased total cholesterol and non-HDL-cholesterol levels while the effect of HMPH was not significant. The results demonstrate that substituting 70% of dietary casein protein with the equivalent amount of HMDP or HMPH protein protects against obesity and diabetes, and HMDP is also beneficial to cholesterol homeostasis.
Collapse
|
32
|
Regan JA, Shah SH. Obesity Genomics and Metabolomics: a Nexus of Cardiometabolic Risk. Curr Cardiol Rep 2020; 22:174. [PMID: 33040225 DOI: 10.1007/s11886-020-01422-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/14/2020] [Indexed: 12/24/2022]
Abstract
PURPOSE OF REVIEW Obesity is a significant international public health epidemic with major downstream consequences on morbidity and mortality. While lifestyle factors contribute, there is an evolving understanding of genomic and metabolomic pathways involved with obesity and its relationship with cardiometabolic risk. This review will provide an overview of some of these important findings from both a biologic and clinical perspective. RECENT FINDINGS Recent studies have identified polygenic risk scores and metabolomic biomarkers of obesity and related outcomes, which have also highlighted biological pathways, such as the branched-chain amino acid (BCAA) pathway that is dysregulated in this disease. These biomarkers may help in personalizing obesity interventions and for mitigation of future cardiometabolic risk. A multifaceted approach is necessary to impact the growing epidemic of obesity and related diseases. This will likely include incorporating precision medicine approaches with genomic and metabolomic biomarkers to personalize interventions and improve risk prediction.
Collapse
Affiliation(s)
- Jessica A Regan
- Department of Medicine, Duke University, Durham, NC, USA.,Duke Molecular Physiology Institute, Duke University, 300 N. Duke Street, DUMC, Box 104775, Durham, NC, 27701, USA
| | - Svati H Shah
- Department of Medicine, Duke University, Durham, NC, USA. .,Duke Molecular Physiology Institute, Duke University, 300 N. Duke Street, DUMC, Box 104775, Durham, NC, 27701, USA.
| |
Collapse
|
33
|
Monnier L, Schlienger JL, Colette C, Bonnet F. The obesity treatment dilemma: Why dieting is both the answer and the problem? A mechanistic overview. DIABETES & METABOLISM 2020; 47:101192. [PMID: 33002604 DOI: 10.1016/j.diabet.2020.09.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 09/05/2020] [Indexed: 12/23/2022]
Abstract
Restricted-calorie diets are the most worldwide used treatments for obesity. Although such strategies are based on the first law of thermodynamics, the real life clinical practice demonstrates that the observed weight losses are divergent from those theoretically predicted. Loosely adherence to recommendations is one of the main causes for the limited efficacy of dieting, but many additional factors can be involved in the hurdles to weight loss. According to the second law of thermodynamics any restriction in dietary energy intake results in energy sparing with a diminution in the basal metabolic rate and a concomitant loss in the lean body mass. This "thrifty" energetic adaptation is associated with a progressive reduction in the difference between levels of energy intake and expenditure, thus resulting in a drastic fall in weight loss rates on the medium and long-term regardless of the dietary carbohydrate/fat ratio. This loss of efficacy is aggravated by the misadaptation of the production and action of anti-obesity hormones such as leptin. During the latest past decades the discovery of changes in the gut microbiota of obese people referred to as "obese dysbiosis" has raised the question as to whether these alterations can participate to diet-resistance. Combined with the behavioral and psychological barriers to low-calorie diets, there is a broad physiologic spectrum of evidence indicating that weight loss is a hard challenge. Consequently, the answer would be primarily to prevent the development of obesity and at worst to avoid its ominous progression from metabolically healthy to unhealthy stages.
Collapse
Affiliation(s)
- Louis Monnier
- Institute of Clinical Research. University of Montpellier, 641 Avenue du doyen Giraud, 34093 Montpellier cedex 5, France.
| | - Jean-Louis Schlienger
- University of Strasbourg, Medical School, 4 rue Kirschleger 67000 Strasbourg, France.
| | - Claude Colette
- Institute of Clinical Research. University of Montpellier, 641 Avenue du doyen Giraud, 34093 Montpellier cedex 5, France.
| | - Fabrice Bonnet
- University of Rennes, University Hospital of Rennes, Department of Endocrinology-Diabetes-Nutrition, 16 Bd de Bulgarie, 35200 Rennes, France.
| |
Collapse
|
34
|
Hossain M, Park DS, Rahman MS, Ki SJ, Lee YR, Imran KM, Yoon D, Heo J, Lee TJ, Kim YS. Bifidobacterium longum DS0956 and Lactobacillus rhamnosus DS0508 culture-supernatant ameliorate obesity by inducing thermogenesis in obese-mice. Benef Microbes 2020; 11:361-373. [PMID: 32755263 DOI: 10.3920/bm2019.0179] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Excessive body fat and the related dysmetabolic diseases affect both developed and developing countries. The aim of this study was to investigate the beneficial role of a bacterial culture supernatant (hereafter: BS) of Lactobacillus and Bifidobacterium and their potential mechanisms of action on white-fat browning and lipolysis. For selection of four candidates among 55 Lactic acid producing bacteria (LAB) from human infant faeces, we evaluated by Oil Red O staining and Ucp1 mRNA quantitation in 3T3-L1 preadipocytes. The expression of browning and lipolysis markers was examined along with in vitro assays. The possible mechanism was revealed by molecular and biological experiments including inhibitor and small interfering RNA (siRNA) assays. In a mouse model, physiological, histological, and biochemical parameters and expression of some thermogenesis-related genes were compared among six experimental groups fed a high-fat diet and one normal-diet control group. The results allow us to speculate that BS treatment promotes browning and lipolysis both in vitro and in vivo. Moreover, the BS may activate thermogenic programs via a mechanism involving PKA-CREB signaling in 3T3-L1 cells. According to our data, we can propose that two LAB strains, Bifidobacterium longum DS0956 and Lactobacillus rhamnosus DS0508, may be good candidates for a dietary supplement against obesity and metabolic diseases; however, further research is required for the development as dietary supplements or drugs.
Collapse
Affiliation(s)
- M Hossain
- Institute of Tissue Regeneration, College of Medicine, Soonchunhyang University, Cheonan Chung-nam 31151, Republic of Korea.,Department of Microbiology, College of Medicine, Soonchunhyang University, Soonchunhyang 6 gil 31, Dongnam-Gu, Cheonan Chung-nam 31151, Republic of Korea
| | - D-S Park
- Biological Resource Center, Korea Research Institute of Bioscience and Biotechnology, 181 Ipsin-gil, Jeongeup-si, Jeollabuk-do 580-185, Republic of Korea
| | - M S Rahman
- Institute of Tissue Regeneration, College of Medicine, Soonchunhyang University, Cheonan Chung-nam 31151, Republic of Korea.,Department of Microbiology, College of Medicine, Soonchunhyang University, Soonchunhyang 6 gil 31, Dongnam-Gu, Cheonan Chung-nam 31151, Republic of Korea
| | - S-J Ki
- Biological Resource Center, Korea Research Institute of Bioscience and Biotechnology, 181 Ipsin-gil, Jeongeup-si, Jeollabuk-do 580-185, Republic of Korea
| | - Y R Lee
- Biological Resource Center, Korea Research Institute of Bioscience and Biotechnology, 181 Ipsin-gil, Jeongeup-si, Jeollabuk-do 580-185, Republic of Korea
| | - K M Imran
- Institute of Tissue Regeneration, College of Medicine, Soonchunhyang University, Cheonan Chung-nam 31151, Republic of Korea.,Department of Microbiology, College of Medicine, Soonchunhyang University, Soonchunhyang 6 gil 31, Dongnam-Gu, Cheonan Chung-nam 31151, Republic of Korea
| | - D Yoon
- Institute of Tissue Regeneration, College of Medicine, Soonchunhyang University, Cheonan Chung-nam 31151, Republic of Korea.,Department of Microbiology, College of Medicine, Soonchunhyang University, Soonchunhyang 6 gil 31, Dongnam-Gu, Cheonan Chung-nam 31151, Republic of Korea
| | - J Heo
- International Agricultural Development and Cooperation Center, Chonbuk National University, Jeonju 54896, Republic of Korea
| | - T-J Lee
- Department of Anatomy, College of Medicine, Yeungnam University, Daegu 42415, Republic of Korea
| | - Y-S Kim
- Institute of Tissue Regeneration, College of Medicine, Soonchunhyang University, Cheonan Chung-nam 31151, Republic of Korea.,Department of Microbiology, College of Medicine, Soonchunhyang University, Soonchunhyang 6 gil 31, Dongnam-Gu, Cheonan Chung-nam 31151, Republic of Korea
| |
Collapse
|
35
|
You W, Xu Z, Sun Y, Valencak TG, Wang Y, Shan T. GADD45α drives brown adipose tissue formation through upregulating PPARγ in mice. Cell Death Dis 2020; 11:585. [PMID: 32719383 PMCID: PMC7385159 DOI: 10.1038/s41419-020-02802-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 07/13/2020] [Accepted: 07/14/2020] [Indexed: 02/07/2023]
Abstract
Stress can lead to obesity and metabolic dysfunction, but the underlying mechanisms are unclear. Here we identify GADD45α, a stress-inducible histone folding protein, as a potential regulator for brown adipose tissue biogenesis. Unbiased transcriptomics data indicate a positive correlation between adipose Gadd45a mRNA level and obesity. At the cellular level, Gadd45a knockdown promoted proliferation and lipolysis of brown adipocytes, while Gadd45a overexpression had the opposite effects. Consistently, using a knockout (Gadd45a−/−) mouse line, we found that GADD45α deficiency inhibited lipid accumulation and promoted expression of thermogenic genes in brown adipocytes, leading to improvements in insulin sensitivity, glucose uptake, energy expenditure. At the molecular level, GADD45α deficiency increased proliferation through upregulating expression of cell cycle related genes. GADD45α promoted brown adipogenesis via interacting with PPARγ and upregulating its transcriptional activity. Our new data suggest that GADD45α may be targeted to promote non-shivering thermogenesis and metabolism while counteracting obesity.
Collapse
Affiliation(s)
- Wenjing You
- College of Animal Sciences, Zhejiang University, Hangzhou, China.,The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China.,Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China
| | - Ziye Xu
- College of Animal Sciences, Zhejiang University, Hangzhou, China.,The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China.,Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China
| | - Ye Sun
- College of Animal Sciences, Zhejiang University, Hangzhou, China.,The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China.,Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China
| | | | - Yizhen Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, China.,The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China.,Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China
| | - Tizhong Shan
- College of Animal Sciences, Zhejiang University, Hangzhou, China. .,The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China. .,Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China.
| |
Collapse
|
36
|
Sebastian M, Hsiao CJ, Futch HS, Eisinger RS, Dumeny L, Patel S, Gobena M, Katikaneni DS, Cohen J, Carpenter AM, Spiryda L, Heldermon CD, Jin L, Brantly ML. Obesity and STING1 genotype associate with 23-valent pneumococcal vaccination efficacy. JCI Insight 2020; 5:136141. [PMID: 32376795 PMCID: PMC7253011 DOI: 10.1172/jci.insight.136141] [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: 01/10/2020] [Accepted: 04/02/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Obesity has been associated with attenuated vaccine responses and an increased risk of contracting pneumococcal pneumonia, but no study to our knowledge has assessed the impact of obesity and genetics on 23-valent pneumococcal vaccine (PPSV23) efficacy. We assessed the relationship of obesity (primary analysis) and stimulator of interferon genes (STING1) genotype (secondary analysis) on PPSV23 efficacy. METHODS Nonobese (BMI 22–25 kg/m2) and obese participants (BMI ≥30 kg/m2) were given a single dose of PPSV23. Blood was drawn immediately prior to and 4–6 weeks after vaccination. Serum samples were used to assess PPSV23-specific antibodies. STING1 genotypes were identified using PCR on DNA extracted from peripheral blood samples. RESULTS Forty-six participants were categorized as nonobese (n = 23; 56.5% women; mean BMI 23.3 kg/m2) or obese (n = 23; 65.2% women; mean BMI 36.3 kg/m2). Obese participants had an elevated fold change in vaccine-specific responses compared with nonobese participants (P < 0.0001). The WT STING1 group (R232/R232) had a significantly higher PPSV23 response than individuals with a single copy of HAQ-STING1 regardless of BMI (P = 0.0025). When WT was assessed alone, obese participants had a higher fold serotype-specific response compared with nonobese participants (P < 0.0001), but no difference was observed between obese and nonobese individuals with 1 HAQ allele (P = 0.693). CONCLUSIONS These observations demonstrate a positive association between obesity and PPSV23 efficacy specifically in participants with the WT STING1 genotype. TRIAL REGISTRATION ClinicalTrials.gov NCT02471014. FUNDING This research was supported by the NIH and the University of Florida MD-PhD Training Program. Obesity and a WT STING1 genotype are positively associated with efficacy of the 23-valent pneumococcal vaccine in a small cohort of subjects.
Collapse
Affiliation(s)
- Mathew Sebastian
- MD-PhD Training Program and.,Lillian S. Wells Department of Neurosurgery, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Chu J Hsiao
- MD-PhD Training Program and.,Genetics Institute, University of Florida, Gainesville, Florida, USA.,Department of Anthropology, University of Florida College of Liberal Arts and Sciences, Gainesville, Florida, USA
| | - Hunter S Futch
- MD-PhD Training Program and.,Norman Fixel Institute for Neurological Diseases, Department of Neuroscience
| | - Robert S Eisinger
- MD-PhD Training Program and.,Norman Fixel Institute for Neurological Diseases, Department of Neuroscience
| | - Leanne Dumeny
- MD-PhD Training Program and.,Genetics Institute, University of Florida, Gainesville, Florida, USA.,Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics and Precision Medicine
| | - Seema Patel
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine
| | - Mesfin Gobena
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine
| | - Divya S Katikaneni
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine
| | - Joel Cohen
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine
| | | | - Lisa Spiryda
- Department of Obstetrics and Gynecology, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Coy D Heldermon
- Division of Hematology and Oncology, Department of Medicine, University of Florida, Gainesville, Florida, USA
| | - Lei Jin
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine
| | - Mark L Brantly
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine
| |
Collapse
|
37
|
Zhao ZJ, Derous D, Gerrard A, Wen J, Liu X, Tan S, Hambly C, Speakman JR. Limits to sustained energy intake. XXX. Constraint or restraint? Manipulations of food supply show peak food intake in lactation is constrained. J Exp Biol 2020; 223:jeb208314. [PMID: 32139473 DOI: 10.1242/jeb.208314] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 02/27/2020] [Indexed: 11/20/2022]
Abstract
Lactating mice increase food intake 4- to 5-fold, reaching an asymptote in late lactation. A key question is whether this asymptote reflects a physiological constraint, or a maternal investment strategy (a 'restraint'). We exposed lactating mice to periods of food restriction, hypothesizing that if the limit reflected restraint, they would compensate by breaching the asymptote when refeeding. In contrast, if it was a constraint, they would by definition be unable to increase their intake on refeeding days. Using isotope methods, we found that during food restriction, the females shut down milk production, impacting offspring growth. During refeeding, food intake and milk production rose again, but not significantly above unrestricted controls. These data provide strong evidence that asymptotic intake in lactation reflects a physiological/physical constraint, rather than restraint. Because hypothalamic neuropeptide Y (Npy) was upregulated under both states of restriction, this suggests the constraint is not imposed by limits in the capacity to upregulate hunger signalling (the saturated neural capacity hypothesis). Understanding the genetic basis of the constraint will be a key future goal and will provide us additional information on the nature of the constraining factors on reproductive output, and their potential links to life history strategies.
Collapse
Affiliation(s)
- Zhi-Jun Zhao
- School of Life and Environmental Sciences, Wenzhou University, Wenzhou, Zhejiang 325035, China
| | - Davina Derous
- School of Biological Sciences, University of Aberdeen, Aberdeen AB24 2TZ, UK
| | - Abby Gerrard
- School of Biological Sciences, University of Aberdeen, Aberdeen AB24 2TZ, UK
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100100, China
| | - Jing Wen
- School of Life and Environmental Sciences, Wenzhou University, Wenzhou, Zhejiang 325035, China
| | - Xue Liu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100100, China
| | - Song Tan
- School of Life and Environmental Sciences, Wenzhou University, Wenzhou, Zhejiang 325035, China
| | - Catherine Hambly
- School of Biological Sciences, University of Aberdeen, Aberdeen AB24 2TZ, UK
| | - John R Speakman
- School of Biological Sciences, University of Aberdeen, Aberdeen AB24 2TZ, UK
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100100, China
- CAS Center of Excellence for Animal Evolution and Genetics, Kunming 650223, China
| |
Collapse
|
38
|
Cahyadi M, Park HB, Seo DW, Jin S, Choi N, Heo KN, Kang BS, Jo C, Lee JH. Association of the thyroid hormone responsive spot 14 alpha gene with growth-related traits in Korean native chicken. ASIAN-AUSTRALASIAN JOURNAL OF ANIMAL SCIENCES 2020; 33:1755-1762. [PMID: 32106653 PMCID: PMC7649070 DOI: 10.5713/ajas.19.0541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 02/10/2020] [Indexed: 11/28/2022]
Abstract
Objective Thyroid hormone responsive spot 14 alpha (THRSP) has been used to investigate the regulation of de novo lipogenesis because the variation of THRSP mRNA content in the tissue affects directly the ability of that tissue to synthetize lipids. Also, this gene responds to thyroid hormone stimulation and high level of carbohydrate feeding or insulin-injection. This study was carried out to investigate variations within THRSP and their effects on body and carcass weights in Korean native chicken (KNC). Methods A total of 585 chickens which represent the five lines of KNC (Black, Gray-Brown, Red-Brown, White, and Yellow-Brown) were reared and body weight data were recorded every two weeks from hatch until 20 weeks of age. Polymerase chain reaction-restriction fragment length polymorphism, DNA chips for Agilent 2100 Bioanalyzer, and Fluidigm Genotyping Technology, were applied to genotype selected markers. A linear mixed-effect model was used to access association between these single nucleotide polymorphism (SNP) markers and growth-related traits. Results A total of 30 polymorphisms were investigated in THRSP. Of these, nine SNPs for loci were selected to perform association analyses. Significant associations were detected between g.-49G>T SNP with body weight at 20 weeks of age (BW20), g.451T>C SNP with growth at 10 to 12 weeks of age (GR10-12), and g.1432A>C SNP with growth at 14 to 16 weeks trait (GR14-16) and body weight at 18 weeks of age (BW18). Moreover, diplotype of the THRSP gene significantly affected body weight at 12 weeks of age (BW12) and GR10-12 traits. Diplotype of ht1/ht2 was favorable for BW12 and GR10-12 traits. Conclusion These results suggest that THRSP can be regarded as a candidate gene for growth traits in KNC.
Collapse
Affiliation(s)
- Muhammad Cahyadi
- Department of Animal Science, Faculty of Agriculture, Universitas Sebelas Maret, Surakarta 57126, Indonesia
| | - Hee-Bok Park
- Department of Animal Resources Science, Kongju National University, Yesan 32439, Korea
| | - Dong Won Seo
- Department of Animal and Dairy Science, College of Agriculture and Life Sciences, Chungnam National University, Daejeon 34134, Korea
| | - Shil Jin
- Department of Animal and Dairy Science, College of Agriculture and Life Sciences, Chungnam National University, Daejeon 34134, Korea.,Hanwoo Research Institute, National Institute of Animal Science, RDA, Pyeongchang 25340, Korea
| | - Nuri Choi
- Division of Biotechnology, College of Environmental and Bioresource Sciences, Chonbuk National University, Iksan 54596, Korea
| | - Kang Nyeong Heo
- Poultry Research Institute, National Institute of Animal Science, RDA, Pyeongchang 25340, Korea
| | - Bo Seok Kang
- Poultry Research Institute, National Institute of Animal Science, RDA, Pyeongchang 25340, Korea
| | - Cheorun Jo
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul 151921, Korea
| | - Jun Heon Lee
- Department of Animal and Dairy Science, College of Agriculture and Life Sciences, Chungnam National University, Daejeon 34134, Korea
| |
Collapse
|
39
|
Yu K, Li L, Zhang L, Guo L, Wang C. Association between MC4R rs17782313 genotype and obesity: A meta-analysis. Gene 2020; 733:144372. [PMID: 31954858 DOI: 10.1016/j.gene.2020.144372] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 01/08/2020] [Accepted: 01/13/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND Obesity is a huge burden of the world. It is commonly recognized that dietary structure and physical inactivity is essential in the progress of obesity. However, some individuals still face the trouble of obese even though they live a healthy life. Except for the combination of diseases, the operation of both lifestyle and genetic features contributes to obesity. Melanocortin-4-receptor (MC4R) gene is one of the known hereditary factors of obesity. rs17782313, a single nucleotide variant in MC4R gene, has been reported unclear results in whether it plays a role in obesity. This meta-analysis is to estimate the association between MC4R rs17782313 genotype and obesity. METHOD A systematic literature retrieval was conducted in four databases: PubMed, Embase, Web of Science and Cochrane Library with specific search strategy. Select qualified studies to identify relevant studies. Odds ratios (ORs) with 95% confidence intervals (CI), P value and I2 value were used to assess the strength of the association in meta-analysis and adjusted with ethnicity, quality and single nucleotide polymorphism (SNP) testing method. RESULT 6 eligible studies involving 3133 obese cases and 3123 normal-weight participants were selected from 378 articles. Allele B of MC4R rs17782313 present a statistically significant association with obesity under allele contrast model (OR = 1.325, 95%CI: 1.219-1.439), dominant model (OR = 1.320, 95%CI: 1.184-1.472), recessive model (OR = 1.690, 95%CI: 1.420-2.011) and homozygous type of co-dominant model (OR = 1.925, 95%CI: 1.590-2.330), respectively, and P < 0.05. CONCLUSION Mutated MC4R rs17782313 is associated with higher risk of obesity. People with homozygous mutant genotype of MC4R rs17782313 would be more likely to suffer from obesity, while heterozygous mutant genotype needs further studies to clarify.
Collapse
Affiliation(s)
- Keping Yu
- Department of Endocrinology, Chongqing General Hospital, Chongqing 400013, PR China
| | - Li Li
- Department of Endocrinology, Chongqing General Hospital, Chongqing 400013, PR China
| | - Lan Zhang
- Department of Endocrinology, Chongqing General Hospital, Chongqing 400013, PR China
| | - Li Guo
- Department of Endocrinology, Chongqing General Hospital, Chongqing 400013, PR China
| | - Chengjian Wang
- Department of Endocrinology, Chongqing General Hospital, Chongqing 400013, PR China.
| |
Collapse
|
40
|
Perez-Calahorra S, Civeira F, Guallar-Castillón P, Pinto X, Banegas JR, Pedro-Botet J, Suarez-Tembra M, Mauri M, Soler C, Rodriguez-Artalejo F, Laclaustra M. Behavioural cardiovascular risk factors and prevalence of diabetes in subjects with familial hypercholesterolaemia. Eur J Prev Cardiol 2020; 27:1649-1660. [PMID: 31914797 DOI: 10.1177/2047487319896138] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
A low prevalence of type 2 diabetes mellitus has been reported in familial hypercholesterolaemia. Whether a healthier lifestyle could explain it has not been explored. This cross-sectional study determines the prevalence of lifestyle-related cardiovascular risk factors in heterozygous familial hypercholesterolaemia (HeFH) from the Dyslipidaemia Registry of the Spanish Atherosclerosis Society and in the ENRICA study, a representative sample of the adult Spanish general population, weighted to match the age and sex distribution of the HeFH sample. A total of 2185 HeFH patients and 11,856 individuals from ENRICA were included. HeFH had lower body mass index and fewer of them were smokers than in the reference population. A model adjusted for age, sex and body mass index showed that HeFH more frequently had cardiovascular disease (odds ratio (OR) 23.98; 95% confidence interval (CI) 18.40-31.23) and hypertension (OR 1.20; 95% CI 1.07-1.35), and took anti-hypertensive medication (OR 1.36; 95% CI 1.18-1.56) and anti-diabetic medication (OR 1.25; 95% CI 1.00-1.56), but less frequently were smokers (OR 0.79; 95% CI 0.71-0.89). In a HeFH subsample (n = 513) with complete blood glucose information, those patients without cardiovascular disease showed lower prevalence of smoking and type 2 diabetes mellitus, lower body mass index and glucose, and higher diastolic blood pressure than the Spanish population. The differences in type 2 diabetes mellitus were justified mostly by the difference in body mass index. Body mass index adjustment also showed higher prevalence of hypertension and use of anti-hypertensive drugs in HeFH. In summary, HeFH patients had lower body mass index, which may contribute to explaining the lower prevalence of diabetes, and lower current smoking but higher hypertension.
Collapse
Affiliation(s)
- Sofia Perez-Calahorra
- Lipid Unit, Hospital Universitario Miguel Servet, IIS Aragon, CIBERCV, Zaragoza, Spain
| | - Fernando Civeira
- Lipid Unit, Hospital Universitario Miguel Servet, IIS Aragon, CIBERCV, Zaragoza, Spain.,Universidad de Zaragoza, Spain
| | - Pilar Guallar-Castillón
- Department of Preventive Medicine and Public Health, School of Medicine, University Autonoma of Madrid/Research Institute of University Hospital La Paz (IdiPAZ) and CIBER de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain.,IMDEA Food Institute, CEI UAM+CSIC, Madrid, Spain
| | - Xavier Pinto
- Lipid Unit and Vascular Risk Unit, Internal Medicine Service, Hospital de Bellvitge, CIBEROBN, Hospitalet de Llobregat, Barcelona, Spain
| | - José R Banegas
- Department of Preventive Medicine and Public Health, School of Medicine, University Autonoma of Madrid/Research Institute of University Hospital La Paz (IdiPAZ) and CIBER de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Juan Pedro-Botet
- Servicio Endocrinología y Nutrición, Hospital del Mar and Departamento de Medicina, Universitat Autònoma de Barcelona, Spain
| | | | - Marta Mauri
- Lipid Unit, Consorci Sanitari de Terrassa-Hospital de Terrassa, Spain
| | - Cristina Soler
- Internal Medicine Department, Hospital de Santa Caterina de Salt, Parc Hospitalari Martí i Julià, Girona, Spain
| | - Fernando Rodriguez-Artalejo
- Department of Preventive Medicine and Public Health, School of Medicine, University Autonoma of Madrid/Research Institute of University Hospital La Paz (IdiPAZ) and CIBER de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain.,IMDEA Food Institute, CEI UAM+CSIC, Madrid, Spain
| | - Martín Laclaustra
- Lipid Unit, Hospital Universitario Miguel Servet, IIS Aragon, CIBERCV, Zaragoza, Spain.,Fundación Agencia Aragonesa para la Investigación y Desarrollo (ARAID), Zaragoza, Spain
| |
Collapse
|
41
|
Zhang X, Li W, Tang Y, Lin C, Cao Y, Chen Y. Mechanism of Pentagalloyl Glucose in Alleviating Fat Accumulation in Caenorhabditis elegans. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:14110-14120. [PMID: 31789033 DOI: 10.1021/acs.jafc.9b06167] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Pentagalloyl glucose (PGG) has been studied for its valuable biological activities. However, the functional role of PGG in lipid metabolism in vivo is unclear. Here, we investigated the effects of PGG on lipid metabolism and its underlying mechanism in Caenorhabditis elegans. PGG decreased the accumulation of reactive oxygen species at 800 μM and remarkably increased the activities of antioxidant enzymes. PGG decreased significantly fat accumulation in wild-type worms (39.7 ± 5.7% in the normal group and 19.9 ± 4.5% in the high-fat group by Oil red O; 21.2 ± 2.7% in the high-fat group by Nile red; p < 0.001), but fat reduction by PGG was eliminated in the skn-1 mutant. The amount and size of lipid droplets in the ZXW618 mutant were decreased by PGG. The proportions of unsaturated fatty acids in both conditions were increased by PGG. In addition, the expression levels of fat metabolism genes were significantly changed in both conditions by PGG, which include mdt-15, pod-2, elo-2, fat-6, and fat-7 genes modulated fat synthesis; aak-2 and nhr-49 genes participated in fat consumption; and tub-1 gene regulated fat storage. However, fat-5 and acs-2 were downregulated in high-fat worms only, and vit-2 and lipl-4 were downregulated in normal worms only. Our study provided new insights into the role of PGG in alleviating fat accumulation and its underlying mechanism of action in C. elegans.
Collapse
Affiliation(s)
- Xiaoying Zhang
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science , South China Agricultural University , Guangzhou 510642 , Guangdong , China
| | - Wei Li
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science , South China Agricultural University , Guangzhou 510642 , Guangdong , China
| | - Yunzhou Tang
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science , South China Agricultural University , Guangzhou 510642 , Guangdong , China
| | - Chunxiu Lin
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science , South China Agricultural University , Guangzhou 510642 , Guangdong , China
| | - Yong Cao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science , South China Agricultural University , Guangzhou 510642 , Guangdong , China
| | - Yunjiao Chen
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science , South China Agricultural University , Guangzhou 510642 , Guangdong , China
| |
Collapse
|
42
|
Abstract
The discovery of leptin changed the view of adipose tissue from that of a passive vessel that stores fat to that of a dynamic endocrine organ that actively regulates behaviour and metabolism. Secreted by adipose tissue, leptin functions as an afferent signal in a negative feedback loop, acting primarily on neurons in the hypothalamus and regulating feeding and many other functions. The leptin endocrine system serves a critical evolutionary function by maintaining the relative constancy of adipose tissue mass, thereby protecting individuals from the risks associated with being too thin (starvation and infertility) or too obese (predation). In this Review, the biology of leptin is summarized, and a conceptual framework is established for studying the pathogenesis of obesity, which, analogously to diabetes, can result from either leptin hyposecretion or leptin resistance. Herein, these two states are distinguished with the terms 'type 1 obesity' and 'type 2 obesity': type 1 obesity describes a subset of obese individuals with low endogenous plasma leptin levels who respond to leptin therapy, whereas type 2 obesity describes most obese individuals, who are leptin resistant but might respond to leptin therapy in combination with other drugs, such as leptin sensitizers.
Collapse
Affiliation(s)
- Jeffrey M Friedman
- Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA.
| |
Collapse
|
43
|
Role of Personalized Nutrition in Chronic-Degenerative Diseases. Nutrients 2019; 11:nu11081707. [PMID: 31344895 PMCID: PMC6723746 DOI: 10.3390/nu11081707] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 07/15/2019] [Accepted: 07/20/2019] [Indexed: 02/07/2023] Open
Abstract
Human nutrition is a branch of medicine based on foods biochemical interactions with the human body. The phenotypic transition from health to disease status can be attributed to changes in genes and/or protein expression. For this reason, a new discipline has been developed called “-omic science”. In this review, we analyzed the role of “-omics sciences” (nutrigenetics, nutrigenomics, proteomics and metabolomics) in the health status and as possible therapeutic tool in chronic degenerative diseases. In particular, we focused on the role of nutrigenetics and the relationship between eating habits, changes in the DNA sequence and the onset of nutrition-related diseases. Moreover, we examined nutrigenomics and the effect of nutrients on gene expression. We perused the role of proteomics and metabolomics in personalized nutrition. In this scenario, we analyzed also how dysbiosis of gut microbiota can influence the onset and progression of chronic degenerative diseases. Moreover, nutrients influencing and regulating gene activity, both directly and indirectly, paves the way for personalized nutrition that plays a key role in the prevention and treatment of chronic degenerative diseases.
Collapse
|
44
|
Lakra P, Aditi K, Agrawal N. Peripheral Expression of Mutant Huntingtin is a Critical Determinant of Weight Loss and Metabolic Disturbances in Huntington's Disease. Sci Rep 2019; 9:10127. [PMID: 31300691 PMCID: PMC6626032 DOI: 10.1038/s41598-019-46470-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 06/26/2019] [Indexed: 12/31/2022] Open
Abstract
Deteriorating weight loss in patients with Huntington's disease (HD) is a complicated peripheral manifestation and the cause remains poorly understood. Studies suggest that body weight strongly influences the clinical progression rate of HD and thereby offers a valuable target for therapeutic interventions. Mutant huntingtin (mHTT) is ubiquitously expressed and could induce toxicity by directly acting in the peripheral tissues. We investigated the effects of selective expression of mHTT exon1 in fat body (FB; functionally equivalent to human adipose tissue and liver) using transgenic Drosophila. We find that FB-autonomous expression of mHTT exon1 is intrinsically toxic and causes chronic weight loss in the flies despite progressive hyperphagia, and early adult death. Moreover, flies exhibit loss of intracellular lipid stores, and decline in the systemic levels of lipids and carbohydrates which aggravates over time, representing metabolic defects. At the cellular level, besides impairment, cell death also occurs with the formation of mHTT aggregates in the FB. These findings indicate that FB-autonomous expression of mHTT alone is sufficient to cause metabolic abnormalities and emaciation in vivo without any neurodegenerative cues.
Collapse
Affiliation(s)
- Priya Lakra
- Department of Zoology, University of Delhi, Delhi, 110007, India
| | - Kumari Aditi
- Department of Zoology, University of Delhi, Delhi, 110007, India
| | - Namita Agrawal
- Department of Zoology, University of Delhi, Delhi, 110007, India.
| |
Collapse
|
45
|
Muller YL, Hanson RL, Piaggi P, Chen P, Wiessner G, Okani C, Skelton G, Kobes S, Hsueh WC, Knowler WC, Bogardus C, Baier LJ. Assessing the Role of 98 Established Loci for BMI in American Indians. Obesity (Silver Spring) 2019; 27:845-854. [PMID: 30887699 PMCID: PMC6478540 DOI: 10.1002/oby.22433] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 01/10/2019] [Indexed: 12/26/2022]
Abstract
OBJECTIVE Meta-analyses of genome-wide association studies in Europeans have identified > 98 loci for BMI. Transferability of these established associations in Pima Indians was analyzed. METHODS Among 98 lead single nucleotide polymorphisms (SNPs), 82 had minor allele frequency ≥ 0.01 in Pima Indians and were analyzed for association with the maximum BMI in adulthood (n = 3,491) and BMI z score in childhood (n = 1,958). Common tag SNPs across 98 loci were also analyzed for additional signals. RESULTS Among the lead SNPs, 13 (TMEM18, TCF7L2, MRPS33P4, PRKD1, ZFP64, FTO, TAL1, CALCR, GNPDA2, CREB1, LMX1B, ADCY9, NLRC3) were associated with BMI (P ≤ 0.05) in Pima adults. A multi-allelic genetic risk score (GRS), which summed the risk alleles for 82 lead SNPs, showed a significant trend for a positive relationship between GRS and BMI in adulthood (beta = 0.48% per risk allele; P = 1.6 × 10-9 ) and BMI z score in childhood (beta = 0.024 SD; P = 1.7 × 10-7 ). GRS was significantly associated with BMI across all age groups ≥ 5 years, except for those ≥ 50 years. The strongest association was seen in adolescence (age 14-16 years; P = 1.84 × 10-9 ). CONCLUSIONS In aggregate, European-derived lead SNPs had a notable effect on BMI in Pima Indians. Polygenic obesity in this population manifests strongly in childhood and adolescence and persists throughout much of adult life.
Collapse
Affiliation(s)
- Yunhua L Muller
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, Arizona, USA
| | - Robert L Hanson
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, Arizona, USA
| | - Paolo Piaggi
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, Arizona, USA
| | - Peng Chen
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, Arizona, USA
| | - Gregory Wiessner
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, Arizona, USA
| | - Chidinma Okani
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, Arizona, USA
| | - Graham Skelton
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, Arizona, USA
| | - Sayuko Kobes
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, Arizona, USA
| | - Wen-Chi Hsueh
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, Arizona, USA
| | - William C Knowler
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, Arizona, USA
| | - Clifton Bogardus
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, Arizona, USA
| | - Leslie J Baier
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, Arizona, USA
| |
Collapse
|
46
|
Khera AV, Chaffin M, Wade KH, Zahid S, Brancale J, Xia R, Distefano M, Senol-Cosar O, Haas ME, Bick A, Aragam KG, Lander ES, Smith GD, Mason-Suares H, Fornage M, Lebo M, Timpson NJ, Kaplan LM, Kathiresan S. Polygenic Prediction of Weight and Obesity Trajectories from Birth to Adulthood. Cell 2019; 177:587-596.e9. [PMID: 31002795 PMCID: PMC6661115 DOI: 10.1016/j.cell.2019.03.028] [Citation(s) in RCA: 430] [Impact Index Per Article: 86.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 11/07/2018] [Accepted: 03/12/2019] [Indexed: 12/30/2022]
Abstract
Severe obesity is a rapidly growing global health threat. Although often attributed to unhealthy lifestyle choices or environmental factors, obesity is known to be heritable and highly polygenic; the majority of inherited susceptibility is related to the cumulative effect of many common DNA variants. Here we derive and validate a new polygenic predictor comprised of 2.1 million common variants to quantify this susceptibility and test this predictor in more than 300,000 individuals ranging from middle age to birth. Among middle-aged adults, we observe a 13-kg gradient in weight and a 25-fold gradient in risk of severe obesity across polygenic score deciles. In a longitudinal birth cohort, we note minimal differences in birthweight across score deciles, but a significant gradient emerged in early childhood and reached 12 kg by 18 years of age. This new approach to quantify inherited susceptibility to obesity affords new opportunities for clinical prevention and mechanistic assessment.
Collapse
Affiliation(s)
- Amit V Khera
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Division of Cardiology, Massachusetts General Hospital, Boston, MA 02114, USA; Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA.
| | - Mark Chaffin
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Kaitlin H Wade
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol BS8 1TH, UK; Population Health Science, Bristol Medical School, Bristol, Bristol BS8 1TH, UK; Avon Longitudinal Study of Parents and Children, Bristol BS8 1TH, UK
| | - Sohail Zahid
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Joseph Brancale
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA; Obesity, Metabolism, and Nutrition Institute, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Rui Xia
- The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Marina Distefano
- Laboratory for Molecular Medicine, Partners HealthCare Personalized Medicine, Cambridge, MA 02139, USA; Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Pathology, Harvard Medical School, Cambridge, MA 02115, USA
| | - Ozlem Senol-Cosar
- Laboratory for Molecular Medicine, Partners HealthCare Personalized Medicine, Cambridge, MA 02139, USA; Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Pathology, Harvard Medical School, Cambridge, MA 02115, USA
| | - Mary E Haas
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Alexander Bick
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Krishna G Aragam
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Division of Cardiology, Massachusetts General Hospital, Boston, MA 02114, USA; Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Eric S Lander
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Program in Health Sciences and Technology, Harvard Medical School, Boston, MA 02115, USA
| | - George Davey Smith
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol BS8 1TH, UK; Population Health Science, Bristol Medical School, Bristol, Bristol BS8 1TH, UK
| | - Heather Mason-Suares
- Laboratory for Molecular Medicine, Partners HealthCare Personalized Medicine, Cambridge, MA 02139, USA; Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Pathology, Harvard Medical School, Cambridge, MA 02115, USA
| | - Myriam Fornage
- The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Matthew Lebo
- Laboratory for Molecular Medicine, Partners HealthCare Personalized Medicine, Cambridge, MA 02139, USA; Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Pathology, Harvard Medical School, Cambridge, MA 02115, USA
| | - Nicholas J Timpson
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol BS8 1TH, UK; Population Health Science, Bristol Medical School, Bristol, Bristol BS8 1TH, UK; Avon Longitudinal Study of Parents and Children, Bristol BS8 1TH, UK
| | - Lee M Kaplan
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA; Obesity, Metabolism, and Nutrition Institute, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Sekar Kathiresan
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Division of Cardiology, Massachusetts General Hospital, Boston, MA 02114, USA; Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
47
|
Ontawong A, Boonphang O, Pasachan T, Duangjai A, Pongchaidecha A, Phatsara M, Jinakote M, Amornlerdpison D, Srimaroeng C. Hepatoprotective effect of coffee pulp aqueous extract combined with simvastatin against hepatic steatosis in high-fat diet-induced obese rats. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
48
|
Shishay G, Liu G, Jiang X, Yu Y, Teketay W, Du D, Jing H, Liu C. Variation in the Promoter Region of the MC4R Gene Elucidates the Association of Body Measurement Traits in Hu Sheep. Int J Mol Sci 2019; 20:E240. [PMID: 30634446 PMCID: PMC6358852 DOI: 10.3390/ijms20020240] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 12/24/2018] [Accepted: 12/30/2018] [Indexed: 12/15/2022] Open
Abstract
The melanocortin 4 receptor (MC4R) gene is expressed in the appetite-regulating areas of the brain and is engaged in the leptin signaling pathway. Although previous studies have identified variants in the coding region of the sheep MC4R gene showing significant associations with birth weight, weaning weight, and backfat thickness, no such associations have been reported for the promoter region. Besides, the essential promoter region of the sheep MC4R has not been delineated. In this study, to better understand the transcriptional regulation of MC4R and to elucidate the association between regulatory variants and haplotypes with body measurement traits in sheep, we cloned and characterized the MC4R promoter. We found that the minimal promoter of the gene is located within the region -1207/-880 bp upstream of the first exon. Real-time quantitative PCR (RT-qPCR) data revealed the mRNA expression of the MC4R gene had a significant difference between sex and age. In the association analysis, eight single nucleotide polymorphisms (SNPs) had a significant association with one or more traits (p < 0.05); of these, two SNPs were novel. Notably, individuals with haplotype H1H2 (CT-GA-GT-GA-GT-GA-GA-CG) were heavier in body weight than other haplotypes. Altogether, variations in the MC4R gene promoter, most notably haplotype H1H2, may greatly benefit marker-assisted selection in sheep.
Collapse
Affiliation(s)
- Girmay Shishay
- Laboratory of Small Ruminant Genetics, Breeding and Reproduction, Huazhong Agricultural University, Wuhan 430070, China.
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China.
| | - Guiqiong Liu
- Laboratory of Small Ruminant Genetics, Breeding and Reproduction, Huazhong Agricultural University, Wuhan 430070, China.
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China.
| | - Xunping Jiang
- Laboratory of Small Ruminant Genetics, Breeding and Reproduction, Huazhong Agricultural University, Wuhan 430070, China.
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China.
| | - Yun Yu
- Laboratory of Small Ruminant Genetics, Breeding and Reproduction, Huazhong Agricultural University, Wuhan 430070, China.
| | - Wassie Teketay
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China.
| | - Dandan Du
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China.
| | - Huang Jing
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China.
| | - Chenghui Liu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
49
|
Böckerman P, Cawley J, Viinikainen J, Lehtimäki T, Rovio S, Seppälä I, Pehkonen J, Raitakari O. The effect of weight on labor market outcomes: An application of genetic instrumental variables. HEALTH ECONOMICS 2019; 28:65-77. [PMID: 30240095 PMCID: PMC6585973 DOI: 10.1002/hec.3828] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 07/13/2018] [Accepted: 08/23/2018] [Indexed: 05/23/2023]
Abstract
This paper contributes to the literature on the labor market consequences of obesity by using a novel instrument: genetic risk score, which reflects the predisposition to higher body mass index (BMI) across many genetic loci. We estimate instrumental variable models of the effect of BMI on labor market outcomes using Finnish data that have many strengths, for example, BMI that is measured rather than self-reported, and data on earnings and social income transfers that are from administrative tax records and are thus free of the problems associated with nonresponse, reporting error or top coding. The empirical results are sensitive to whether we use a narrower or broader genetic risk score, and to model specification. For example, models using the narrower genetic risk score as an instrument imply that a one-unit increase in BMI is associated with 6.9% lower wages, 1.8% fewer years employed, and a 3 percentage point higher probability of receiving any social income transfers. However, when we use a newer, broader genetic risk score, we cannot reject the null hypothesis of no effect. Future research using genetic risk scores should examine the sensitivity of their results to the risk score used.
Collapse
Affiliation(s)
- Petri Böckerman
- Labour Institute for Economic Research and IZAJyväskylä University School of Business and Economics, University of JyväskyläJyväskyläFinland
| | - John Cawley
- Department of Policy Analysis and Management, and Department of EconomicsCornell UniversityIthacaNew York
| | - Jutta Viinikainen
- Jyväskylä University School of Business and Economics, University of JyväskyläJyväskyläFinland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories and Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Life SciencesUniversity of TampereTampereFinland
| | - Suvi Rovio
- Research Centre of Applied and Preventive Cardiovascular MedicineUniversity of TurkuTurkuFinland
| | - Ilkka Seppälä
- Department of Clinical Chemistry, Fimlab Laboratories and Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Life SciencesUniversity of TampereTampereFinland
| | - Jaakko Pehkonen
- Jyväskylä University School of Business and Economics, University of JyväskyläJyväskyläFinland
| | - Olli Raitakari
- Research Centre of Applied and Preventive Cardiovascular MedicineUniversity of TurkuTurkuFinland
- Department of Clinical Physiology and Nuclear MedicineTurku University HospitalTurkuFinland
| |
Collapse
|
50
|
Turer EE, San Miguel M, Wang KW, McAlpine W, Ou F, Li X, Tang M, Zang Z, Wang J, Hayse B, Evers B, Zhan X, Russell J, Beutler B. A viable hypomorphic Arnt2 mutation causes hyperphagic obesity, diabetes and hepatic steatosis. Dis Model Mech 2018; 11:11/12/dmm035451. [PMID: 30563851 PMCID: PMC6307907 DOI: 10.1242/dmm.035451] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 11/20/2018] [Indexed: 12/12/2022] Open
Abstract
Aryl hydrocarbon receptor nuclear translocator 2 (ARNT2) is a member of the basic helix-loop-helix/PER-ARNT-SIM (bHLH/PAS) transcription factor family. ARNT2 heterodimerizes with several members of the family, including single-minded homolog-1 (SIM1) and neuronal PAS domain protein 4 (NPAS4), primarily in neurons of the central nervous system. We screened 64,424 third-generation germline mutant mice derived from N-ethyl-N-nitrosourea (ENU)-mutagenized great-grandsires for weight abnormalities. Among 17 elevated body weight phenotypes identified and mapped, one strongly correlated with an induced missense mutation in Arnt2 using a semidominant model of inheritance. Causation was confirmed by CRISPR/Cas9 gene targeting to recapitulate the original ENU allele, specifying Arg74Cys (R74C). The CRISPR/Cas9-targeted (Arnt2R74C/R74C) mice demonstrated hyperphagia and increased adiposity as well as hepatic steatosis and abnormalities in glucose homeostasis. The mutant ARNT2 protein showed decreased transcriptional activity when coexpressed with SIM1. These findings establish a requirement for ARNT2-dependent genes in the maintenance of the homeostatic feeding response, necessary for prevention of obesity and obesity-related diseases. Summary: A forward genetic approach identified genes involved in maintaining normal body weight. A mutation in Arnt2 caused obesity by regulating food intake.
Collapse
Affiliation(s)
- Emre E Turer
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX, 75390-8505, USA.,Department of Internal Medicine, Division of Gastroenterology, University of Texas Southwestern Medical Center, Dallas, TX, 75390-8505 USA
| | - Miguel San Miguel
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX, 75390-8505, USA.,Department of Internal Medicine, Division of Gastroenterology, University of Texas Southwestern Medical Center, Dallas, TX, 75390-8505 USA
| | - Kuan-Wen Wang
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX, 75390-8505, USA
| | - William McAlpine
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX, 75390-8505, USA
| | - Feiya Ou
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX, 75390-8505, USA.,Department of Internal Medicine, Division of Gastroenterology, University of Texas Southwestern Medical Center, Dallas, TX, 75390-8505 USA
| | - Xiaohong Li
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX, 75390-8505, USA
| | - Miao Tang
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX, 75390-8505, USA
| | - Zhao Zang
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX, 75390-8505, USA
| | - Jianhui Wang
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX, 75390-8505, USA
| | - Braden Hayse
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX, 75390-8505, USA
| | - Bret Evers
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, 75390-8505 USA
| | - Xiaoming Zhan
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX, 75390-8505, USA
| | - Jamie Russell
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX, 75390-8505, USA
| | - Bruce Beutler
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX, 75390-8505, USA
| |
Collapse
|