51
|
Ortega Martínez de Victoria E, Pané Vila A, Jiménez Pineda A. Obesity in Spain: an open book that must be read. REVISTA ESPANOLA DE CARDIOLOGIA (ENGLISH ED.) 2024; 77:819-820. [PMID: 38750932 DOI: 10.1016/j.rec.2024.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 03/07/2024] [Indexed: 05/26/2024]
Affiliation(s)
- Emilio Ortega Martínez de Victoria
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Madrid, Spain; Servicio de Endocrinología y Nutrición, Hospital Clínic de Barcelona, Spain; Fundació Clínic per la Recerca Biomèdica - Institut d'Investigacions Biomèdiques August Pi Sunyer (FCRB-IDIBAPS), Barcelona, Spain.
| | - Adriana Pané Vila
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Madrid, Spain; Servicio de Endocrinología y Nutrición, Hospital Clínic de Barcelona, Spain; Fundació Clínic per la Recerca Biomèdica - Institut d'Investigacions Biomèdiques August Pi Sunyer (FCRB-IDIBAPS), Barcelona, Spain
| | - Amanda Jiménez Pineda
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Madrid, Spain; Servicio de Endocrinología y Nutrición, Hospital Clínic de Barcelona, Spain; Fundació Clínic per la Recerca Biomèdica - Institut d'Investigacions Biomèdiques August Pi Sunyer (FCRB-IDIBAPS), Barcelona, Spain
| |
Collapse
|
52
|
Grannell A, le Roux C. Obesity as a disease: a pressing need for alignment. Int J Obes (Lond) 2024; 48:1361-1362. [PMID: 38987635 PMCID: PMC11420073 DOI: 10.1038/s41366-024-01582-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 06/24/2024] [Accepted: 07/02/2024] [Indexed: 07/12/2024]
Affiliation(s)
- Andrew Grannell
- Sidekick Health, Research & Development Unit, Kópavogur, Iceland.
| | - Carel le Roux
- Diabetes Complications Research Centre, University College Dublin, Dublin, Ireland
| |
Collapse
|
53
|
Robinson KM, Robinson KA, Scherer AM, Mackin ML. Patient Perceptions of Weight Stigma Experiences in Healthcare: A Qualitative Analysis. Health Expect 2024; 27:e70013. [PMID: 39223786 PMCID: PMC11369018 DOI: 10.1111/hex.70013] [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: 03/07/2024] [Revised: 08/05/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Weight stigma is the social devaluation and denigration of individuals because of their excess body weight, resulting in poorer physical and mental health and healthcare avoidance. Attribution Theory and Goffman's theory of spoiled identity provided a general overarching framework for understanding weight stigma experiences. OBJECTIVE Our purpose was to explore weight stigma experiences from a broad range of perspectives emphasizing identities typically excluded in the weight stigma literature. DESIGN We conducted a qualitative descriptive study with data drawn from 73 substantive narrative comments from participants who responded to a larger survey. RESULTS Analysis developed five themes: Working on weight, Not being overweight, Lack of help and empathy, Exposure and embarrassment and Positive experiences. Individuals who would be clinically assessed as overweight, especially men, often did not identify with having a weight problem and found the framing of personal responsibility for weight empowering. Participants with larger body sizes more often attributed embarrassment and shame about weight to treatment in the clinical setting. Older participants were more likely to have positive experiences. CONCLUSIONS The findings suggest ongoing tension between the framing of weight as a personal responsibility as opposed to a multifactorial condition with many uncontrollable aspects. Gender, age and body size shaped respondent perspectives, with some young male respondents finding empowerment through perceived personal control of weight. The healthcare system perpetuates weight stigma through lack of adequate equipment and excessively weight-centric medical counselling. Recommending a healthy lifestyle to patients without support or personalized medical assessment may perpetuate weight stigma and associated detrimental health outcomes. PATIENT OR PUBLIC CONTRIBUTION Patients with obesity and overweight were integral to this study, providing comments for our qualitative analyses.
Collapse
Affiliation(s)
- Kathleen M. Robinson
- Department of Internal MedicineUniversity of Iowa Hospitals and ClinicsIowa CityIowaUSA
- Division of EndocrinologyUniversity of Iowa Hospitals and ClinicsIowa CityIowaUSA
| | | | - Aaron M. Scherer
- Department of Internal MedicineUniversity of Iowa Hospitals and ClinicsIowa CityIowaUSA
| | - Melissa Lehan Mackin
- Health Science Campus, College of NursingUniversity of New Mexico College of NursingAlbuquerqueNew MexicoUSA
| |
Collapse
|
54
|
Siddiqui J, Kinney CE, Han JC. The Genetics of Obesity. Pediatr Clin North Am 2024; 71:897-917. [PMID: 39343500 DOI: 10.1016/j.pcl.2024.06.001] [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] [Indexed: 10/01/2024]
Abstract
Understanding the genetic causes of obesity permits anticipatory guidance and targeted treatments. Children with hyperphagia and severe early-onset obesity should receive genetic testing for rare monogenic and syndromic disorders caused by pathogenic variants involving a single gene or single chromosomal region. Gene panels covering the leptin pathway, the key regulator of energy balance, are becoming more widely available and at lower cost. Polygenic obesity is much more common and involves multiple genes throughout the genome, although the overlap in genes for rare and common disorders suggests a spectrum of severity and the potential of shared precision medicine approaches for treatment.
Collapse
Affiliation(s)
- Juwairriyyah Siddiqui
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, Mount Sinai Hospital, Diabetes, Obesity, and Metabolism Institute, Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - Clint E Kinney
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, Mount Sinai Hospital, Diabetes, Obesity, and Metabolism Institute, Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - Joan C Han
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, Mount Sinai Hospital, Diabetes, Obesity, and Metabolism Institute, Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA.
| |
Collapse
|
55
|
Loeb GB, Kathail P, Shuai RW, Chung R, Grona RJ, Peddada S, Sevim V, Federman S, Mader K, Chu AY, Davitte J, Du J, Gupta AR, Ye CJ, Shafer S, Przybyla L, Rapiteanu R, Ioannidis NM, Reiter JF. Variants in tubule epithelial regulatory elements mediate most heritable differences in human kidney function. Nat Genet 2024; 56:2078-2092. [PMID: 39256582 DOI: 10.1038/s41588-024-01904-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 08/12/2024] [Indexed: 09/12/2024]
Abstract
Kidney failure, the decrease of kidney function below a threshold necessary to support life, is a major cause of morbidity and mortality. We performed a genome-wide association study (GWAS) of 406,504 individuals in the UK Biobank, identifying 430 loci affecting kidney function in middle-aged adults. To investigate the cell types affected by these loci, we integrated the GWAS with human kidney candidate cis-regulatory elements (cCREs) identified using single-cell assay for transposase-accessible chromatin sequencing (scATAC-seq). Overall, 56% of kidney function heritability localized to kidney tubule epithelial cCREs and an additional 7% to kidney podocyte cCREs. Thus, most heritable differences in adult kidney function are a result of altered gene expression in these two cell types. Using enhancer assays, allele-specific scATAC-seq and machine learning, we found that many kidney function variants alter tubule epithelial cCRE chromatin accessibility and function. Using CRISPRi, we determined which genes some of these cCREs regulate, implicating NDRG1, CCNB1 and STC1 in human kidney function.
Collapse
Affiliation(s)
- Gabriel B Loeb
- Division of Nephrology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA.
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA.
| | - Pooja Kathail
- Center for Computational Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Richard W Shuai
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, Berkeley, CA, USA
| | - Ryan Chung
- Center for Computational Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Reinier J Grona
- Division of Nephrology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Sailaja Peddada
- Laboratory for Genomics Research, San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
| | - Volkan Sevim
- Laboratory for Genomics Research, San Francisco, CA, USA
- Target Discovery, GSK, San Francisco, CA, USA
| | - Scot Federman
- Laboratory for Genomics Research, San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
| | - Karl Mader
- Laboratory for Genomics Research, San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
| | - Audrey Y Chu
- Human Genetics and Genomics, GSK, Cambridge, MA, USA
| | | | - Juan Du
- Department of Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Alexander R Gupta
- Department of Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Chun Jimmie Ye
- Division of Rheumatology, Department of Medicine; Bakar Computational Health Sciences Institute; Parker Institute for Cancer Immunotherapy; Institute for Human Genetics; Department of Epidemiology & Biostatistics; Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, USA
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Arc Institute, Palo Alto, CA, USA
| | - Shawn Shafer
- Laboratory for Genomics Research, San Francisco, CA, USA
- Target Discovery, GSK, San Francisco, CA, USA
| | - Laralynne Przybyla
- Laboratory for Genomics Research, San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
| | - Radu Rapiteanu
- Genome Biology, Research Technologies, GSK, Stevenage, UK
| | - Nilah M Ioannidis
- Center for Computational Biology, University of California, Berkeley, Berkeley, CA, USA
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, Berkeley, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Jeremy F Reiter
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA.
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
| |
Collapse
|
56
|
Tyler AL, Mahoney JM, Keller MP, Baker CN, Gaca M, Srivastava A, Gerdes Gyuricza I, Braun MJ, Rosenthal NA, Attie AD, Churchill GA, Carter GW. Transcripts with high distal heritability mediate genetic effects on complex metabolic traits. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.26.613931. [PMID: 39386475 PMCID: PMC11463413 DOI: 10.1101/2024.09.26.613931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Although many genes are subject to local regulation, recent evidence suggests that complex distal regulation may be more important in mediating phenotypic variability. To assess the role of distal gene regulation in complex traits, we combined multi-tissue transcriptomes with physiological outcomes to model diet-induced obesity and metabolic disease in a population of Diversity Outbred mice. Using a novel high-dimensional mediation analysis, we identified a composite transcriptome signature that summarized genetic effects on gene expression and explained 30% of the variation across all metabolic traits. The signature was heritable, interpretable in biological terms, and predicted obesity status from gene expression in an independently derived mouse cohort and multiple human studies. Transcripts contributing most strongly to this composite mediator frequently had complex, distal regulation distributed throughout the genome. These results suggest that trait-relevant variation in transcription is largely distally regulated, but is nonetheless identifiable, interpretable, and translatable across species.
Collapse
|
57
|
Dudek MF, Wenz BM, Brown CD, Voight BF, Almasy L, Grant SF. Characterization of non-coding variants associated with transcription factor binding through ATAC-seq-defined footprint QTLs in liver. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.24.614730. [PMID: 39386531 PMCID: PMC11463493 DOI: 10.1101/2024.09.24.614730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Non-coding variants discovered by genome-wide association studies (GWAS) are enriched in regulatory elements harboring transcription factor (TF) binding motifs, strongly suggesting a connection between disease association and the disruption of cis-regulatory sequences. Occupancy of a TF inside a region of open chromatin can be detected in ATAC-seq where bound TFs block the transposase Tn5, leaving a pattern of relatively depleted Tn5 insertions known as a "footprint". Here, we sought to identify variants associated with TF-binding, or "footprint quantitative trait loci" (fpQTLs) in ATAC-seq data generated from 170 human liver samples. We used computational tools to scan the ATAC-seq reads to quantify TF binding likelihood as "footprint scores" at variants derived from whole genome sequencing generated in the same samples. We tested for association between genotype and footprint score and observed 693 fpQTLs associated with footprint-inferred TF binding (FDR < 5%). Given that Tn5 insertion sites are measured with base-pair resolution, we show that fpQTLs can aid GWAS and QTL fine-mapping by precisely pinpointing TF activity within broad trait-associated loci where the underlying causal variant is unknown. Liver fpQTLs were strongly enriched across ChIP-seq peaks, liver expression QTLs (eQTLs), and liver-related GWAS loci, and their inferred effect on TF binding was concordant with their effect on underlying sequence motifs in 80% of cases. We conclude that fpQTLs can reveal causal GWAS variants, define the role of TF binding site disruption in disease and provide functional insights into non-coding variants, ultimately informing novel treatments for common diseases.
Collapse
Affiliation(s)
- Max F. Dudek
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Graduate Group in Genomics and Computational Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Brandon M. Wenz
- Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christopher D. Brown
- Graduate Group in Genomics and Computational Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Benjamin F. Voight
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Laura Almasy
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Lifespan Brain Institute, Children’s Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Biomedical and Health Informatics, Children’s Hospital of Philadelphia
| | - Struan F.A. Grant
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of Endocrinology and Diabetes, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| |
Collapse
|
58
|
Placzek M, Chinnaiya K, Kim DW, Blackshaw S. Control of tuberal hypothalamic development and its implications in metabolic disorders. Nat Rev Endocrinol 2024:10.1038/s41574-024-01036-1. [PMID: 39313573 DOI: 10.1038/s41574-024-01036-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/29/2024] [Indexed: 09/25/2024]
Abstract
The tuberal hypothalamus regulates a range of crucial physiological processes, including energy homeostasis and metabolism. In this Review, we explore the intricate molecular mechanisms and signalling pathways that control the development of the tuberal hypothalamus, focusing on aspects that shape metabolic outcomes. Major developmental events are discussed in the context of their effect on the establishment of both functional hypothalamic neuronal circuits and brain-body interfaces that are pivotal to the control of metabolism. Emerging evidence indicates that aberrations in molecular pathways during tuberal hypothalamic development contribute to metabolic dysregulation. Understanding the molecular underpinnings of tuberal hypothalamic development provides a comprehensive view of neurodevelopmental processes and offers a promising avenue for future targeted interventions to prevent and treat metabolic disorders.
Collapse
Affiliation(s)
- Marysia Placzek
- School of Biosciences, University of Sheffield, Sheffield, UK.
- Bateson Centre, University of Sheffield, Sheffield, UK.
- Neuroscience Institute, University of Sheffield, Sheffield, UK.
| | | | - Dong Won Kim
- Danish Research Institute of Translational Neuroscience (DANDRITE), Nordic EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Seth Blackshaw
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
59
|
Baya NA, Erdem IS, Venkatesh SS, Reibe S, Charles PD, Navarro-Guerrero E, Hill B, Lassen FH, Claussnitzer M, Palmer DS, Lindgren CM. Combining evidence from human genetic and functional screens to identify pathways altering obesity and fat distribution. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.19.24313913. [PMID: 39371160 PMCID: PMC11451655 DOI: 10.1101/2024.09.19.24313913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Overall adiposity and body fat distribution are heritable traits associated with altered risk of cardiometabolic disease and mortality. Performing rare variant (minor allele frequency<1%) association testing using exome-sequencing data from 402,375 participants in the UK Biobank (UKB) for nine overall and tissue-specific fat distribution traits, we identified 19 genes where putatively damaging rare variation associated with at least one trait (Bonferroni-adjusted P<1.58×10-7) and 52 additional genes at FDR≤1% (P≤4.37×10-5). These 71 genes exhibited higher (P=3.58×10-18) common variant prioritisation scores than genes not significantly enriched for rare putatively damaging variation, with evidence of monotonic allelic series (dose-response relationships) among ultra-rare variants (minor allele count≤10) in 22 genes. Five of the 71 genes have cognate protein UKB Olink data available; all five associated (P<3.80×10-6) with three or more analysed traits. Combining rare and common variation evidence, allelic series and proteomics, we selected 17 genes for CRISPR knockout in human white adipose tissue cell lines. In three previously uncharacterised target genes, knockout increased (two-sided t-test P<0.05) lipid accumulation, a cellular phenotype relevant for fat mass traits, compared to Cas9-empty negative controls: COL5A3 (fold change [FC]=1.72, P=0.0028), EXOC7 (FC=1.35, P=0.0096), and TRIP10 (FC=1.39, P=0.0157); furthermore, knockout of SLTM resulted in reduced lipid accumulation (FC=0.51, P=1.91×10-4). Integrating across population-based genetic and in vitro functional evidence, we highlight therapeutic avenues for altering obesity and body fat distribution by modulating lipid accumulation.
Collapse
Affiliation(s)
- Nikolas A Baya
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford OX3 7LF, United Kingdom
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, United Kingdom
| | - Ilknur Sur Erdem
- Nuffield Department of Women's and Reproductive Health, Medical Sciences Division, University of Oxford, United Kingdom
- Chinese Academy for Medical Sciences Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, United Kingdom
| | - Samvida S Venkatesh
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford OX3 7LF, United Kingdom
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, United Kingdom
| | - Saskia Reibe
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford OX3 7LF, United Kingdom
- Nuffield Department of Population Health, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Philip D Charles
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford OX3 7LF, United Kingdom
| | - Elena Navarro-Guerrero
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, United Kingdom
| | - Barney Hill
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford OX3 7LF, United Kingdom
- Nuffield Department of Population Health, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Frederik Heymann Lassen
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford OX3 7LF, United Kingdom
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, United Kingdom
| | - Melina Claussnitzer
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
- Broad Institute of MIT and Harvard, Novo Nordisk Foundation Center for Genomic Mechanisms of Disease & Type 2 Diabetes Systems Genomics Initiative, Cambridge, Massachusetts, United States of America
- Center for Genomic Medicine and Endocrine Division, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Harvard Medical School, Harvard University, Boston, Massachusetts, United States of America
| | - Duncan S Palmer
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford OX3 7LF, United Kingdom
- Nuffield Department of Population Health, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Cecilia M Lindgren
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford OX3 7LF, United Kingdom
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, United Kingdom
- Nuffield Department of Women's and Reproductive Health, Medical Sciences Division, University of Oxford, United Kingdom
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| |
Collapse
|
60
|
Huang Y, Sulek K, Stinson SE, Holm LA, Kim M, Trost K, Hooshmand K, Lund MAV, Fonvig CE, Juel HB, Nielsen T, Ängquist L, Rossing P, Thiele M, Krag A, Holm JC, Legido-Quigley C, Hansen T. Lipid profiling identifies modifiable signatures of cardiometabolic risk in children and adolescents with obesity. Nat Med 2024:10.1038/s41591-024-03279-x. [PMID: 39304782 DOI: 10.1038/s41591-024-03279-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 08/30/2024] [Indexed: 09/22/2024]
Abstract
Pediatric obesity is a progressive, chronic disease that can lead to serious cardiometabolic complications. Here we investigated the peripheral lipidome in 958 children and adolescents with overweight or obesity and 373 with normal weight, in a cross-sectional study. We also implemented a family-based, personalized program to assess the effects of obesity management on 186 children and adolescents in a clinical setting. Using mass spectrometry-based lipidomics, we report an increase in ceramides, alongside a decrease in lysophospholipids and omega-3 fatty acids with obesity metabolism. Ceramides, phosphatidylethanolamines and phosphatidylinositols were associated with insulin resistance and cardiometabolic risk, whereas sphingomyelins showed inverse associations. Additionally, a panel of three lipids predicted hepatic steatosis as effectively as liver enzymes. Lipids partially mediated the association between obesity and cardiometabolic traits. The nonpharmacological management reduced levels of ceramides, phospholipids and triglycerides, indicating that lowering the degree of obesity could partially restore a healthy lipid profile in children and adolescents.
Collapse
Affiliation(s)
- Yun Huang
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | | | - Sara E Stinson
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Louise Aas Holm
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- The Children's Obesity Clinic, accredited European Centre for Obesity Management, Department of Paediatrics, Copenhagen University Hospital Holbæk, Holbæk, Denmark
| | - Min Kim
- Steno Diabetes Center Copenhagen, Copenhagen, Denmark
| | - Kajetan Trost
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- Steno Diabetes Center Copenhagen, Copenhagen, Denmark
| | | | - Morten Asp Vonsild Lund
- The Children's Obesity Clinic, accredited European Centre for Obesity Management, Department of Paediatrics, Copenhagen University Hospital Holbæk, Holbæk, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Cilius E Fonvig
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- The Children's Obesity Clinic, accredited European Centre for Obesity Management, Department of Paediatrics, Copenhagen University Hospital Holbæk, Holbæk, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Helene Bæk Juel
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Trine Nielsen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Medical Department, Zealand University Hospital, Roskilde, Denmark
| | - Lars Ängquist
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Peter Rossing
- Steno Diabetes Center Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Maja Thiele
- Center for Liver Research, Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Aleksander Krag
- Center for Liver Research, Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Jens-Christian Holm
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark.
- The Children's Obesity Clinic, accredited European Centre for Obesity Management, Department of Paediatrics, Copenhagen University Hospital Holbæk, Holbæk, Denmark.
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Cristina Legido-Quigley
- Steno Diabetes Center Copenhagen, Copenhagen, Denmark.
- Institute of Pharmaceutical Science, King's College London, London, United Kingdom.
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
61
|
Jovanovic VM, Narisu N, Bonnycastle LL, Tharakan R, Mesch KT, Glover HJ, Yan T, Sinha N, Sen C, Castellano D, Yang S, Blivis D, Ryu S, Bennett DF, Rosales-Soto G, Inman J, Ormanoglu P, LeClair CA, Xia M, Schneider M, Hernandez-Ochoa EO, Erdos MR, Simeonov A, Chen S, Collins FS, Doege CA, Tristan CA. Scalable Hypothalamic Arcuate Neuron Differentiation from Human Pluripotent Stem Cells Suitable for Modeling Metabolic and Reproductive Disorders. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.27.601062. [PMID: 39005353 PMCID: PMC11244856 DOI: 10.1101/2024.06.27.601062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
The hypothalamus, composed of several nuclei, is essential for maintaining our body's homeostasis. The arcuate nucleus (ARC), located in the mediobasal hypothalamus, contains neuronal populations with eminent roles in energy and glucose homeostasis as well as reproduction. These neuronal populations are of great interest for translational research. To fulfill this promise, we used a robotic cell culture platform to provide a scalable and chemically defined approach for differentiating human pluripotent stem cells (hPSCs) into pro-opiomelanocortin (POMC), somatostatin (SST), tyrosine hydroxylase (TH) and gonadotropin-releasing hormone (GnRH) neuronal subpopulations with an ARC-like signature. This robust approach is reproducible across several distinct hPSC lines and exhibits a stepwise induction of key ventral diencephalon and ARC markers in transcriptomic profiling experiments. This is further corroborated by direct comparison to human fetal hypothalamus, and the enriched expression of genes implicated in obesity and type 2 diabetes (T2D). Genome-wide chromatin accessibility profiling by ATAC-seq identified accessible regulatory regions that can be utilized to predict candidate enhancers related to metabolic disorders and hypothalamic development. In depth molecular, cellular, and functional experiments unveiled the responsiveness of the hPSC-derived hypothalamic neurons to hormonal stimuli, such as insulin, neuropeptides including kisspeptin, and incretin mimetic drugs such as Exendin-4, highlighting their potential utility as physiologically relevant cellular models for disease studies. In addition, differential glucose and insulin treatments uncovered adaptability within the generated ARC neurons in the dynamic regulation of POMC and insulin receptors. In summary, the establishment of this model represents a novel, chemically defined, and scalable platform for manufacturing large numbers of hypothalamic arcuate neurons and serves as a valuable resource for modeling metabolic and reproductive disorders.
Collapse
Affiliation(s)
- Vukasin M. Jovanovic
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation Rockville, MD 20850, USA
- Hypothalamus Consortium
| | - Narisu Narisu
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
- Hypothalamus Consortium
| | - Lori L. Bonnycastle
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
- Hypothalamus Consortium
| | - Ravi Tharakan
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation Rockville, MD 20850, USA
| | - Kendall T. Mesch
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation Rockville, MD 20850, USA
- Hypothalamus Consortium
| | - Hannah J. Glover
- Naomi Berrie Diabetes Center, Columbia Stem Cell Initiative, Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
- Hypothalamus Consortium
| | - Tingfen Yan
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
- Hypothalamus Consortium
| | - Neelam Sinha
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
- Hypothalamus Consortium
| | - Chaitali Sen
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation Rockville, MD 20850, USA
- Hypothalamus Consortium
| | - David Castellano
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation Rockville, MD 20850, USA
| | - Shu Yang
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation Rockville, MD 20850, USA
| | - Dvir Blivis
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation Rockville, MD 20850, USA
| | - Seungmi Ryu
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation Rockville, MD 20850, USA
| | - Daniel F. Bennett
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Giovanni Rosales-Soto
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Jason Inman
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation Rockville, MD 20850, USA
| | - Pinar Ormanoglu
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation Rockville, MD 20850, USA
| | - Christopher A. LeClair
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation Rockville, MD 20850, USA
| | - Menghang Xia
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation Rockville, MD 20850, USA
| | - Martin Schneider
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Erick O. Hernandez-Ochoa
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Michael R. Erdos
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
- Hypothalamus Consortium
| | - Anton Simeonov
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation Rockville, MD 20850, USA
| | - Shuibing Chen
- Department of Surgery, Center for Genomic Health, Weill Cornell Medicine, New York, NY 10065, USA
- Hypothalamus Consortium
| | - Francis S. Collins
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
- Hypothalamus Consortium
| | - Claudia A. Doege
- Naomi Berrie Diabetes Center, Columbia Stem Cell Initiative, Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
- Hypothalamus Consortium
| | - Carlos A. Tristan
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation Rockville, MD 20850, USA
- Hypothalamus Consortium
| |
Collapse
|
62
|
Politei JM, Patrono A. Clinically Meaningful Outcomes after 1 Year of Treatment with Setmelanotide in an Adult Patient with a Variant in SH2B1. Obes Facts 2024; 17:646-651. [PMID: 39284294 DOI: 10.1159/000541267] [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: 03/08/2024] [Accepted: 08/26/2024] [Indexed: 10/12/2024] Open
Abstract
INTRODUCTION Monogenic obesity is caused by a unique genetic dysfunction, often appears in childhood, and can be accompanied by neuroendocrine, skeletal, developmental, and behavioral disorders, among other manifestations. Some variants in the SH2B1 gene have been suggested as strong candidates for the development of autosomal dominant obesity. CASE PRESENTATION We describe here the clinical response after 1 year of setmelanotide treatment in a 22-year-old patient with an SH2B1 variant. After 3 months of treatment, our patient lost 5.4% of body weight. This period was followed by a 3-month period of noncompliance, in which the patient gained 4% body weight. After reinstating daily drug administration, the patient showed a 19.5% reduction in body weight and a clear improvement in all hunger scales after 1 year of treatment. CONCLUSION These results indicate that the changes seen are drug dependent and provide positive evidence for the administration of setmelanotide in adult patients with heterozygous variants in the SH2B1 gene.
Collapse
Affiliation(s)
- Juan M Politei
- Neurology Department, SPINE Foundation, Buenos Aires, Argentina
| | - Andrea Patrono
- Nutrition Department, Trinity Clinic, Buenos Aires, Argentina
| |
Collapse
|
63
|
Lingvay I, Cohen RV, Roux CWL, Sumithran P. Obesity in adults. Lancet 2024; 404:972-987. [PMID: 39159652 DOI: 10.1016/s0140-6736(24)01210-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 04/29/2024] [Accepted: 06/06/2024] [Indexed: 08/21/2024]
Abstract
Obesity has increased in prevalence worldwide and WHO has declared it a global epidemic. Population-level preventive interventions have been insufficient to slow down this trajectory. Obesity is a complex, heterogeneous, chronic, and progressive disease, which substantially affects health, quality of life, and mortality. Lifestyle and behavioural interventions are key components of obesity management; however, when used alone, they provide substantial and durable response in a minority of people. Bariatric (metabolic) surgery remains the most effective and durable treatment, with proven benefits beyond weight loss, including for cardiovascular and renal health, and decreased rates of obesity-related cancers and mortality. Considerable progress has been made in the development of pharmacological agents that approach the weight loss efficacy of metabolic surgery, and relevant outcome data related to these agents' use are accumulating. However, all treatment approaches to obesity have been vastly underutilised.
Collapse
Affiliation(s)
- Ildiko Lingvay
- Division of Endocrinology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA; Peter O'Donnel Jr School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Ricardo V Cohen
- The Center for Obesity and Diabetes, Oswaldo Cruz German Hospital, São Paulo, Brazil
| | - Carel W le Roux
- Diabetes Complications Research Centre, Conway Institute, School of Medicine, University College Dublin, Dublin, Ireland; Diabetes Research Centre, Ulster University, Coleraine, UK
| | - Priya Sumithran
- Department of Surgery, School of Translational Medicine, Monash University, Melbourne, VIC, Australia; Department of Endocrinology and Diabetes, Alfred Health, Melbourne, VIC, Australia
| |
Collapse
|
64
|
Saeed S, Bonnefond A, Froguel P. Obesity: exploring its connection to brain function through genetic and genomic perspectives. Mol Psychiatry 2024:10.1038/s41380-024-02737-9. [PMID: 39237720 DOI: 10.1038/s41380-024-02737-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 08/27/2024] [Accepted: 08/29/2024] [Indexed: 09/07/2024]
Abstract
Obesity represents an escalating global health burden with profound medical and economic impacts. The conventional perspective on obesity revolves around its classification as a "pure" metabolic disorder, marked by an imbalance between calorie consumption and energy expenditure. Present knowledge, however, recognizes the intricate interaction of rare or frequent genetic factors that favor the development of obesity, together with the emergence of neurodevelopmental and mental abnormalities, phenotypes that are modulated by environmental factors such as lifestyle. Thirty years of human genetic research has unveiled >20 genes, causing severe early-onset monogenic obesity and ~1000 loci associated with common polygenic obesity, most of those expressed in the brain, depicting obesity as a neurological and mental condition. Therefore, obesity's association with brain function should be better recognized. In this context, this review seeks to broaden the current perspective by elucidating the genetic determinants that contribute to both obesity and neurodevelopmental and mental dysfunctions. We conduct a detailed examination of recent genetic findings, correlating them with clinical and behavioral phenotypes associated with obesity. This includes how polygenic obesity, influenced by a myriad of genetic variants, impacts brain regions associated with addiction and reward, differentiating it from monogenic forms. The continuum between non-syndromic and syndromic monogenic obesity, with evidence from neurodevelopmental and cognitive assessments, is also addressed. Current therapeutic approaches that target these genetic mechanisms, yielding improved clinical outcomes and cognitive advantages, are discussed. To sum up, this review corroborates the genetic underpinnings of obesity, affirming its classification as a neurological disorder that may have broader implications for neurodevelopmental and mental conditions. It highlights the promising intersection of genetics, genomics, and neurobiology as a foundation for developing tailored medical approaches to treat obesity and its related neurological aspects.
Collapse
Affiliation(s)
- Sadia Saeed
- INSERM UMR 1283, CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Lille, France
- University of Lille, Lille University Hospital, Lille, France
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Amélie Bonnefond
- INSERM UMR 1283, CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Lille, France
- University of Lille, Lille University Hospital, Lille, France
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Philippe Froguel
- INSERM UMR 1283, CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Lille, France.
- University of Lille, Lille University Hospital, Lille, France.
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.
| |
Collapse
|
65
|
Kelly AS, Armstrong SC, Michalsky MP, Fox CK. Obesity in Adolescents: A Review. JAMA 2024; 332:738-748. [PMID: 39102244 DOI: 10.1001/jama.2024.11809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/06/2024]
Abstract
Importance Obesity affects approximately 21% of US adolescents and is associated with insulin resistance, hypertension, dyslipidemia, sleep disorders, depression, and musculoskeletal problems. Obesity during adolescence has also been associated with an increased risk of mortality from cardiovascular disease and type 2 diabetes in adulthood. Observations Obesity in adolescents aged 12 to younger than 18 years is commonly defined as a body mass index (BMI) at the 95th or greater age- and sex-adjusted percentile. Comprehensive treatment in adolescents includes lifestyle modification therapy, pharmacotherapy, and metabolic and bariatric surgery. Lifestyle modification therapy, which includes dietary, physical activity, and behavioral counseling, is first-line treatment; as monotherapy, lifestyle modification requires more than 26 contact hours over 1 year to elicit approximately 3% mean BMI reduction. Newer antiobesity medications, such as liraglutide, semaglutide, and phentermine/topiramate, in combination with lifestyle modification therapy, can reduce mean BMI by approximately 5% to 17% at 1 year of treatment. Adverse effects vary, but severe adverse events from these newer antiobesity medications are rare. Surgery (Roux-en-Y gastric bypass and vertical sleeve gastrectomy) for severe adolescent obesity (BMI ≥120% of the 95th percentile) reduces mean BMI by approximately 30% at 1 year. Minor and major perioperative complications, such as reoperation and hospital readmission for dehydration, are experienced by approximately 15% and 8% of patients, respectively. Determining the long-term durability of all obesity treatments warrants future research. Conclusions and Relevance The prevalence of adolescent obesity is approximately 21% in the US. Treatment options for adolescents with obesity include lifestyle modification therapy, pharmacotherapy, and metabolic and bariatric surgery. Intensive lifestyle modification therapy reduces BMI by approximately 3% while pharmacotherapy added to lifestyle modification therapy can attain BMI reductions ranging from 5% to 17%. Surgery is the most effective intervention for adolescents with severe obesity and has been shown to achieve BMI reduction of approximately 30%.
Collapse
Affiliation(s)
- Aaron S Kelly
- Department of Pediatrics and Center for Pediatric Obesity Medicine, University of Minnesota Medical School, Minneapolis
| | - Sarah C Armstrong
- Department of Pediatrics, Department of Population Health Sciences, Duke University, Durham, North Carolina
- Duke Clinical Research Institute, Duke Center for Childhood Obesity Research, Durham, North Carolina
| | - Marc P Michalsky
- Department of Pediatric Surgery, Nationwide Children's Hospital and The Ohio State University, College of Medicine, Columbus
| | - Claudia K Fox
- Department of Pediatrics and Center for Pediatric Obesity Medicine, University of Minnesota Medical School, Minneapolis
| |
Collapse
|
66
|
Dumitrescu A, Vitcu GM, Stoica S, Susa SR, Stoicescu ER. Cardiovascular Risk Factors in Socioeconomically Disadvantaged Populations in a Suburb of the Largest City in Western Romania. Biomedicines 2024; 12:1989. [PMID: 39335503 PMCID: PMC11429410 DOI: 10.3390/biomedicines12091989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 08/27/2024] [Accepted: 08/30/2024] [Indexed: 09/30/2024] Open
Abstract
Background and Objectives: Cardiovascular disease (CVD) remains a major public health issue worldwide, disproportionately affecting socioeconomically disadvantaged populations due to the social determinants of health (SDOHs). In Western Romania, these populations are particularly vulnerable to CVD. This study aims to investigate the prevalence and impact of cardiovascular risk factors (CVRFs) among socioeconomically disadvantaged individuals in Western Romania and identify the primary CVRFs contributing to the high incidence of CVD within this population. Materials and Methods: A retrospective observational design was employed, utilizing data from the medical records of 1433 eligible individuals. The inclusion criteria were based on Eurostat's EU-SILC benchmarks, focusing on severe material deprivation, at-risk-of-poverty rates, and low work intensity. Data on demographics, familial and personal medical history, smoking status, blood pressure, glucose, cholesterol, triglycerides, and HbA1c levels were collected. Results: Of the 1433 subjects, 34.75% were male, with a median age of 52 years. Significant conditions included diabetes (7.39%), coronary disease (3.83%), arterial hypertension (35.58%), and dyslipidemia (21.28%). Median ages were higher for those with diabetes (65 vs. 51 years, p < 0.0001), coronary disease (64 vs. 51 years, p < 0.0001), arterial hypertension (65 vs. 43 years, p < 0.0001), and dyslipidemia (66 vs. 47 years, p < 0.0001). BMI (Body Mass Index) classifications showed 33.77% were overweight, 21.21% obese, and 15.07% morbidly obese. Smokers were younger than non-smokers (48 vs. 54 years, p < 0.0001). Conclusions: The findings highlight the significant prevalence of CVRFs among socioeconomically disadvantaged populations in Western Romania. Socioeconomically disadvantaged populations exhibit a significantly higher prevalence of cardiovascular risk factors such as diabetes, impaired glucose regulation, hypertension, and dyslipidemia compared to their before known status.
Collapse
Affiliation(s)
- Andreea Dumitrescu
- Neoclinic Concept SRL, Calea Dorobantilor 3, 300134 Timisoara, Romania; (A.D.); (G.M.V.)
- Department of Kinetotherapy, Faculty of Physical Education and Sport, West Univestity of Timisoara, 300223 Timisoara, Romania
| | - Gabriela Mut Vitcu
- Neoclinic Concept SRL, Calea Dorobantilor 3, 300134 Timisoara, Romania; (A.D.); (G.M.V.)
| | - Svetlana Stoica
- Neoclinic Concept SRL, Calea Dorobantilor 3, 300134 Timisoara, Romania; (A.D.); (G.M.V.)
- Cardiology University Clinic, Department VI, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
- Research Centre of Timisoara Institute of Cardiovascular Diseases, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
| | - Septimiu Radu Susa
- Doctoral School, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania;
| | - Emil Robert Stoicescu
- Radiology and Medical Imaging University Clinic, ‘Victor Babes’ University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania;
- Research Center for Pharmaco-Toxicological Evaluations, ‘Victor Babes’ University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
- Field of Applied Engineering Sciences, Specialization Statistical Methods and Techniques in Health and Clinical Research, Faculty of Mechanics, ‘Politehnica’ University Timisoara, Mihai Viteazul Boulevard No. 1, 300222 Timisoara, Romania
| |
Collapse
|
67
|
Wang M, Chao M, Han H, Zhao T, Yan W, Yang G, Pang W, Cai R. Hinokiflavone resists HFD-induced obesity by promoting apoptosis in an IGF2BP2-mediated Bim m 6A modification dependent manner. J Biol Chem 2024; 300:107721. [PMID: 39214307 PMCID: PMC11465056 DOI: 10.1016/j.jbc.2024.107721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 07/29/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024] Open
Abstract
Obesity has emerged as a major health risk on a global scale. Hinokiflavone (HF), a natural small molecule, extracted from plants like cypress, exhibits diverse chemical structures and low synthesis costs. Using high-fat diet-induced obese mice models, we found that HF suppresses obesity by inducing apoptosis in adipose tissue. Adipocyte apoptosis helps maintain tissue health by removing aging, damaged, or excess cells in adipose tissue, which is crucial in preventing obesity and metabolic diseases. We found that HF can specifically bind to insulin-like growth factor 2 mRNA binding protein 2 to promote the stability of N6-methyladenosine-modified Bim, inducing mitochondrial outer membrane permeabilization. Mitochondrial outer membrane permeabilization leads to Caspase9/3-mediated adipocyte mitochondrial apoptosis, alleviating obesity induced by a high-fat diet. The proapoptotic effect of HF offers a controlled means for weight loss. This study reveals the potential of small molecule HF in developing new therapeutic approaches in drug development and biomedical research.
Collapse
Affiliation(s)
- Mingyu Wang
- Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Mingkun Chao
- Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Haozhe Han
- Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Tiantian Zhao
- Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Wenyong Yan
- Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Gongshe Yang
- Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Weijun Pang
- Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China.
| | - Rui Cai
- Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China.
| |
Collapse
|
68
|
Keigan J, De Los Santos B, Gaither SE, Walker DC. The relationship between racial/ethnic identification and body ideal internalization, hair satisfaction, and skin tone satisfaction in black and black/white biracial women. Body Image 2024; 50:101719. [PMID: 38788592 DOI: 10.1016/j.bodyim.2024.101719] [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: 07/03/2023] [Revised: 04/03/2024] [Accepted: 04/25/2024] [Indexed: 05/26/2024]
Abstract
Eurocentric physical characteristics, including a thin, tall physique, long straight hair, and fair skin, typify Western beauty standards. Past research indicates that for Black women, greater identification with one's racial/ethnic culture may buffer against internalizing Eurocentric beauty standards, specifically the thin ideal. Black/White Biracial women often experience different appearance pressures from each of their racial identity's sociocultural appearance ideals. Unfortunately, body image research is limited among Bi/Multiracial individuals. Participants were recruited online via Prime Panels, a high-quality data recruitment service provided by CloudResearch. Participants, M(SD)Age= 34.64 (12.85), self-reported their racial/ethnic identification, thin and thick/curvy ideal internalization, and hair and skin tone satisfaction. Using linear regression analyses, we assessed whether racial/ethnic identification buffered against monoracial Black (n = 317) and Black/White Biracial (n = 254) women's thin ideal internalization. Additionally, we assessed whether stronger racial/ethnic identity was associated with stronger thick/curvy ideal internalization and hair and skin tone satisfaction. Supporting hypotheses, greater racial/ethnic identification was associated with higher thick/curvy ideal internalization and hair and skin tone satisfaction among both Black and Biracial women. Contrary to hypotheses, greater racial/ethnic identification was not associated with lower thin ideal internalization in either group. Our results stress the need to use racially and culturally sensitive measurements of body image.
Collapse
Affiliation(s)
- Jessica Keigan
- Union College, Department of Psychology, 807 Union Street, Schenectady, NY 12308, USA
| | - Bonelyn De Los Santos
- Union College, Department of Psychology, 807 Union Street, Schenectady, NY 12308, USA
| | - Sarah E Gaither
- Duke University, Department of Psychology, 417 Chapel Dr, Durham, NC 27708, USA
| | - D Catherine Walker
- Union College, Department of Psychology, 807 Union Street, Schenectady, NY 12308, USA.
| |
Collapse
|
69
|
M JN, Bharadwaj D. The complex web of obesity: from genetics to precision medicine. Expert Rev Endocrinol Metab 2024; 19:403-418. [PMID: 38869356 DOI: 10.1080/17446651.2024.2365785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 06/05/2024] [Indexed: 06/14/2024]
Abstract
INTRODUCTION Obesity is a growing public health concern affecting both children and adults. Since it involves both genetic and environmental components, the management of obesity requires both, an understanding of the underlying genetics and changes in lifestyle. The knowledge of obesity genetics will enable the possibility of precision medicine in anti-obesity medications. AREAS COVERED Here, we explore health complications and the prevalence of obesity. We discuss disruptions in energy balance as a symptom of obesity, examining evolutionary theories, its multi-factorial origins, and heritability. Additionally, we discuss monogenic and polygenic obesity, the converging biological pathways, potential pharmacogenomics applications, and existing anti-obesity medications - specifically focussing on the leptin-melanocortin and incretin pathways. Comparisons between childhood and adult obesity genetics are made, along with insights into structural variants, epigenetic changes, and environmental influences on epigenetic signatures. EXPERT OPINION With recent advancements in anti-obesity drugs, genetic studies pinpoint new targets and allow for repurposing existing drugs. This creates opportunities for genotype-informed treatment options. Also, lifestyle interventions can help in the prevention and treatment of obesity by altering the epigenetic signatures. The comparison of genetic architecture in adults and children revealed a significant overlap. However, more robust studies with diverse ethnic representation is required in childhood obesity.
Collapse
Affiliation(s)
- Janaki Nair M
- Systems Genomics Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Dwaipayan Bharadwaj
- Systems Genomics Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
70
|
Mas-Bermejo P, Azcona-Granada N, Peña E, Lecube A, Ciudin A, Simó R, Luna A, Rigla M, Arenas C, Caixàs A, Rosa A. Genetic risk score based on obesity-related genes and progression in weight loss after bariatric surgery: a 60-month follow-up study. Surg Obes Relat Dis 2024; 20:814-821. [PMID: 38744640 DOI: 10.1016/j.soard.2024.04.002] [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: 10/26/2023] [Revised: 04/02/2024] [Accepted: 04/02/2024] [Indexed: 05/16/2024]
Abstract
BACKGROUND Obesity is a polygenic multifactorial disease. Recent genome-wide association studies have identified several common loci associated with obesity-related phenotypes. Bariatric surgery (BS) is the most effective long-term treatment for patients with severe obesity. The huge variability in BS outcomes between patients suggests a moderating effect of several factors, including the genetic architecture of the patients. OBJECTIVE To examine the role of a genetic risk score (GRS) based on 7 polymorphisms in 5 obesity-candidate genes (FTO, MC4R, SIRT1, LEP, and LEPR) on weight loss after BS. SETTING University hospital in Spain. METHODS We evaluated a cohort of 104 patients with severe obesity submitted to BS (Roux-en-Y gastric bypass or sleeve gastrectomy) followed up for >60 months (lost to follow-up, 19.23%). A GRS was calculated for each patient, considering the number of carried risk alleles for the analyzed genes. During the postoperative period, the percentage of excess weight loss total weight loss and changes in body mass index were evaluated. Generalized estimating equation models were used for the prospective analysis of the variation of these variables in relation to the GRS. RESULTS The longitudinal model showed a significant effect of the GRS on the percentage of excess weight loss (P = 1.5 × 10-5), percentage of total weight loss (P = 3.1 × 10-8), and change in body mass index (P = 7.8 × 10-16) over time. Individuals with a low GRS seemed to experience better outcomes at 24 and 60 months after surgery than those with a higher GRS. CONCLUSION The use of the GRS in considering the polygenic nature of obesity seems to be a useful tool to better understand the outcome of patients with obesity after BS.
Collapse
Affiliation(s)
- Patricia Mas-Bermejo
- Secció de Zoologia i Antropologia Biòlogica, Departament de Biologia Evolutiva, Ecologia i Ciències Ambientals, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain; Institut de Biomedicina de la Universitat de Barcelona, Barcelona, Spain
| | - Natalia Azcona-Granada
- Secció de Zoologia i Antropologia Biòlogica, Departament de Biologia Evolutiva, Ecologia i Ciències Ambientals, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain; Department of Biological Psychology, Vrije Universiteit, Amsterdam, Netherlands; Amsterdam Public Health Research Institute, Amsterdam University Medical Centre, Amsterdam, Netherlands
| | - Elionora Peña
- Secció de Zoologia i Antropologia Biòlogica, Departament de Biologia Evolutiva, Ecologia i Ciències Ambientals, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain; Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Albert Lecube
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Madrid, Spain; Department of Endocrinology and Nutrition, Hospital Universitari Arnau de Vilanova, IRBLleida, Universitat de Lleida, Lleida, Spain
| | - Andreea Ciudin
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Madrid, Spain; Institut de Recerca Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Rafael Simó
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Madrid, Spain; Institut de Recerca Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alexis Luna
- Institut d'Investigació i Innovació Parc Taulí (I3PT-CERCA-ISCIII), Sabadell, Spain; Department of Surgery, Esofago-gastric Surgery Section, Hospital Universitari Parc Taulí, Sabadell, Spain
| | - Mercedes Rigla
- Institut d'Investigació i Innovació Parc Taulí (I3PT-CERCA-ISCIII), Sabadell, Spain; Department of Endocrinology and Nutrition, Hospital Universitari Parc Taulí, and Department of Medicine, Universitat Autònoma de Barcelona, Sabadell, Spain
| | - Concepción Arenas
- Secció d'Estadística, Department de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | - Assumpta Caixàs
- Institut d'Investigació i Innovació Parc Taulí (I3PT-CERCA-ISCIII), Sabadell, Spain; Department of Endocrinology and Nutrition, Hospital Universitari Parc Taulí, and Department of Medicine, Universitat Autònoma de Barcelona, Sabadell, Spain.
| | - Araceli Rosa
- Secció de Zoologia i Antropologia Biòlogica, Departament de Biologia Evolutiva, Ecologia i Ciències Ambientals, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain; Institut de Biomedicina de la Universitat de Barcelona, Barcelona, Spain; CIBER de Salud Mental, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
71
|
Xu Y, He C, Fan J, Zhou Y, Cheng C, Meng R, Cui Y, Li W, Gamazon ER, Zhou D. A multi-modal framework improves prediction of tissue-specific gene expression from a surrogate tissue. EBioMedicine 2024; 107:105305. [PMID: 39180788 PMCID: PMC11388271 DOI: 10.1016/j.ebiom.2024.105305] [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: 02/13/2024] [Revised: 08/08/2024] [Accepted: 08/09/2024] [Indexed: 08/26/2024] Open
Abstract
BACKGROUND Tissue-specific analysis of the transcriptome is critical to elucidating the molecular basis of complex traits, but central tissues are often not accessible. We propose a methodology, Multi-mOdal-based framework to bridge the Transcriptome between PEripheral and Central tissues (MOTPEC). METHODS Multi-modal regulatory elements in peripheral blood are incorporated as features for gene expression prediction in 48 central tissues. To demonstrate the utility, we apply it to the identification of BMI-associated genes and compare the tissue-specific results with those derived directly from surrogate blood. FINDINGS MOTPEC models demonstrate superior performance compared with both baseline models in blood and existing models across the 48 central tissues. We identify a set of BMI-associated genes using the central tissue MOTPEC-predicted transcriptome data. The MOTPEC-based differential gene expression (DGE) analysis of BMI in the central tissues (including brain caudate basal ganglia and visceral omentum adipose tissue) identifies 378 genes overlapping the results from a TWAS of BMI, while only 162 overlapping genes are identified using gene expression in blood. Cellular perturbation analysis further supports the utility of MOTPEC for identifying trait-associated gene sets and narrowing the effect size divergence between peripheral blood and central tissues. INTERPRETATION The MOTPEC framework improves the gene expression prediction accuracy for central tissues and enhances the identification of tissue-specific trait-associated genes. FUNDING This research is supported by the National Natural Science Foundation of China 82204118 (D.Z.), the seed funding of the Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province (2020E10004), the National Institutes of Health (NIH) Genomic Innovator Award R35HG010718 (E.R.G.), NIH/NHGRI R01HG011138 (E.R.G.), NIH/NIA R56AG068026 (E.R.G.), NIH Office of the Director U24OD035523 (E.R.G.), and NIH/NIGMS R01GM140287 (E.R.G.).
Collapse
Affiliation(s)
- Yue Xu
- School of Public Health and the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Chunfeng He
- School of Public Health and the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Jiayao Fan
- School of Public Health and the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Yuan Zhou
- School of Public Health and the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Chunxiao Cheng
- School of Public Health and the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Ran Meng
- School of Public Health and the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ya Cui
- Division of Computational Biomedicine, Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Wei Li
- Division of Computational Biomedicine, Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Eric R Gamazon
- Vanderbit Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA; Data Science Institute, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Dan Zhou
- School of Public Health and the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou, Zhejiang, China.
| |
Collapse
|
72
|
Fitch AK, Malhotra S, Conroy R. Differentiating monogenic and syndromic obesities from polygenic obesity: Assessment, diagnosis, and management. OBESITY PILLARS 2024; 11:100110. [PMID: 38766314 PMCID: PMC11101890 DOI: 10.1016/j.obpill.2024.100110] [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: 02/07/2024] [Revised: 04/18/2024] [Accepted: 04/18/2024] [Indexed: 05/22/2024]
Abstract
Background Obesity is a multifactorial neurohormonal disease that results from dysfunction within energy regulation pathways and is associated with increased morbidity, mortality, and reduced quality of life. The most common form is polygenic obesity, which results from interactions between multiple gene variants and environmental factors. Highly penetrant monogenic and syndromic obesities result from rare genetic variants with minimal environmental influence and can be differentiated from polygenic obesity depending on key symptoms, including hyperphagia; early-onset, severe obesity; and suboptimal responses to nontargeted therapies. Timely diagnosis of monogenic or syndromic obesity is critical to inform management strategies and reduce disease burden. We outline the physiology of weight regulation, role of genetics in obesity, and differentiating characteristics between polygenic and rare genetic obesity to facilitate diagnosis and transition toward targeted therapies. Methods In this narrative review, we focused on case reports, case studies, and natural history studies of patients with monogenic and syndromic obesities and clinical trials examining the efficacy, safety, and quality of life impact of nontargeted and targeted therapies in these populations. We also provide comprehensive algorithms for diagnosis of patients with suspected rare genetic causes of obesity. Results Patients with monogenic and syndromic obesities commonly present with hyperphagia (ie, pathologic, insatiable hunger) and early-onset, severe obesity, and the presence of hallmark characteristics can inform genetic testing and diagnostic approach. Following diagnosis, specialized care teams can address complex symptoms, and hyperphagia is managed behaviorally. Various pharmacotherapies show promise in these patient populations, including setmelanotide and glucagon-like peptide-1 receptor agonists. Conclusion Understanding the pathophysiology and differentiating characteristics of monogenic and syndromic obesities can facilitate diagnosis and management and has led to development of targeted pharmacotherapies with demonstrated efficacy for reducing body weight and hunger in the affected populations.
Collapse
Affiliation(s)
| | - Sonali Malhotra
- Harvard Medical School, Boston, MA, USA
- Rhythm Pharmaceuticals, Inc., Boston, MA, USA
- Massachussetts General Hospital, Boston, MA, USA
| | | |
Collapse
|
73
|
Rios-Lugo MJ, Serafín-Fabián JI, Hernández-Mendoza H, Klünder-Klünder M, Cruz M, Chavez-Prieto E, Martínez-Navarro I, Vilchis-Gil J, Vazquez-Moreno M. Mediation effect of body mass index on the association between serum magnesium level and insulin resistance in children from Mexico City. Eur J Clin Nutr 2024; 78:808-813. [PMID: 38745051 DOI: 10.1038/s41430-024-01447-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 04/26/2024] [Accepted: 04/29/2024] [Indexed: 05/16/2024]
Abstract
BACKGROUND/OBJECTIVES Reduced serum magnesium (Mg) levels have been associated with obesity, insulin resistance (IR), type 2 diabetes, and metabolic syndrome in adults. However, in the children population, the evidence is still limited. In this cross-sectional study, we aimed to analyze the association of serum Mg levels with the frequency of overweight and obesity and cardiometabolic traits in 189 schoolchildren (91 girls and 98 boys) between 6 and 12 years old from Mexico City. SUBJECTS/METHODS Anthropometrical data were collected and biochemical parameters were measured by enzymatic colorimetric assay. Serum Mg level was analyzed by inductively coupled plasma mass spectrometry (ICP-MS). The triglyceride-glucose (TyG) index was used as a surrogate marker to evaluate IR. RESULTS Serum Mg level was negatively associated with overweight (Odds ratio [OR] = 0.377, 95% confidence interval [CI] 0.231-0.614, p < 0.001) and obesity (OR = 0.345, 95% CI 0.202-0.589, p < 0.001). Serum Mg level resulted negatively associated with body mass index (BMI, β = -1.16 ± 0.26, p < 0.001), BMI z-score (β = -0.48 ± 0.10, p < 0.001) and TyG index (β = -0.04 ± 0.04, p = 0.041). Through a mediation analysis was estimated that BMI z-score accounts for 60.5% of the negative association of serum Mg level with IR (Sobel test: z = 2.761; p = 0.005). CONCLUSION Our results evidence that BMI z-score mediate part of the negative association of serum Mg level and IR in Mexican schoolchildren.
Collapse
Affiliation(s)
- María Judith Rios-Lugo
- Facultad de Enfermería y Nutrición, Universidad Autónoma de San Luis Potosí, Avda. Niño Artillero 130, CP 78210, San Luis Potosí, SLP, México
- Sección de Medicina Molecular y Traslacional, Centro de Investigación en Ciencias de Salud y Biomedicina. Universidad Autónoma de San Luis Potosí, Avda. Sierra Leona 550, CP 78210, San Luis, SLP, México
| | - Jesús Isimar Serafín-Fabián
- Unidad de Investigación Médica en Bioquímica, Hospital de Especialidades, Centro Médico Nacional Siglo XXI. Instituto Mexicano del Seguro Social, México City, México
- Doctorado en Ciencias Biomédicas, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero. Chilpancingo de los Bravo, Guerrero, México
| | - Héctor Hernández-Mendoza
- Instituto de Investigación de Zonas Desérticas, Universidad Autónoma de San Luis Potosí, Altair 200, CP 78377, San Luis, SLP, México
- Universidad del Centro de México, Capitán Caldera 75, CP 78250, San Luis, SLP, México
| | - Miguel Klünder-Klünder
- Unidad de Investigación Epidemiológica en Endocrinología y Nutrición, Hospital Infantil de México Federico Gómez, Secretaría de Salud, CP 06720, Ciudad de México, México
| | - Miguel Cruz
- Unidad de Investigación Médica en Bioquímica, Hospital de Especialidades, Centro Médico Nacional Siglo XXI. Instituto Mexicano del Seguro Social, México City, México
| | - Estefania Chavez-Prieto
- Programa Multidisciplinario de Posgrado en Ciencias Ambientales, Universidad Autónoma de San Luis Potosí, Zona Universitaria, Av. Manuel Nava 201, CP 78210, San Luis Potosí, SLP, México
| | - Israel Martínez-Navarro
- Sección de Medicina Molecular y Traslacional, Centro de Investigación en Ciencias de Salud y Biomedicina. Universidad Autónoma de San Luis Potosí, Avda. Sierra Leona 550, CP 78210, San Luis, SLP, México
| | - Jenny Vilchis-Gil
- Unidad de Investigación Epidemiológica en Endocrinología y Nutrición, Hospital Infantil de México Federico Gómez, Secretaría de Salud, CP 06720, Ciudad de México, México.
| | - Miguel Vazquez-Moreno
- Unidad de Investigación Médica en Bioquímica, Hospital de Especialidades, Centro Médico Nacional Siglo XXI. Instituto Mexicano del Seguro Social, México City, México.
| |
Collapse
|
74
|
Patterson E, Nyberg G, Norman Å, Schäfer Elinder L. Universal healthy school start intervention reduced the body mass index of young children with obesity. Acta Paediatr 2024; 113:2119-2125. [PMID: 38381539 DOI: 10.1111/apa.17164] [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: 09/02/2023] [Revised: 01/25/2024] [Accepted: 02/13/2024] [Indexed: 02/23/2024]
Abstract
AIM To evaluate the effect of a universal, school-based family support programme on body mass index (BMI) of children aged 5-7 years, using pooled data from three trials. METHODS The programme has three to four components and is delivered during the first school year. It aims to promote healthy dietary and physical activity behaviours, and secondarily prevent unhealthy weight gain. Three cluster-randomised controlled trials were conducted between 2010 and 2018 in low and mixed socioeconomic status areas in Sweden. Weight and height were measured. Multiple mixed linear regression analysis was performed on the pooled data. RESULTS In total, 961 children were included (50% girls, mean age 6.3 years). The post-intervention effect on BMI z-score in all children was small, but in those with obesity at baseline, we observed a significant, clinically relevant, decrease in BMI z-score (-0.21). This was most pronounced in children with a non-Nordic born parent (-0.24). Five to six months after the intervention, decreases were no longer statistically significant. CONCLUSION The intervention resulted in changes in BMI comparable to obesity treatment programmes focusing on behaviour change. However, the effect attenuated with time suggesting the programme should be sustained and evaluated for a longer time.
Collapse
Affiliation(s)
- Emma Patterson
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden
- Division for Risk and Benefit Assessment, Swedish Food Agency, Uppsala, Sweden
| | - Gisela Nyberg
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden
- The Swedish School of Sport and Health Sciences (GIH), Stockholm, Sweden
| | - Åsa Norman
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Liselotte Schäfer Elinder
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden
- Centre for Epidemiology and Community Medicine, Region Stockholm, Stockholm, Sweden
| |
Collapse
|
75
|
Bailin SS, Koethe JR. Weight Gain and Antiretroviral Therapy. Infect Dis Clin North Am 2024; 38:499-515. [PMID: 38871568 PMCID: PMC11305935 DOI: 10.1016/j.idc.2024.04.005] [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] [Indexed: 06/15/2024]
Abstract
Antiretroviral therapy (ART) agents as a determinant of body weight in ART-naïve and ART-experienced persons with human immunodeficiency virus (HIV) (PWH) has become a major focus area in research and clinical settings. Recent studies demonstrating weight-suppressing properties of efavirenz and tenofovir disoproxil fumarate led to re-evaluation of weight gain studies, and a reassessment of whether other agents are weight promoting versus weight neutral. In this review, the authors synthesize recent literature on factors related to obesity, clinical measurements of adiposity, weight gain in ART-naïve and ART-experienced PWH, metabolic consequences of ART and weight gain, and the clinical management of weight gain in PWH.
Collapse
Affiliation(s)
- Samuel S Bailin
- Division of Infectious Diseases, Vanderbilt University Medical Center, 1161 21st Avenue South, A2200 Medical Center North, Nashville, TN 37232, USA.
| | - John R Koethe
- Division of Infectious Diseases, Vanderbilt University Medical Center, 1161 21st Avenue South, A2200 Medical Center North, Nashville, TN 37232, USA
| |
Collapse
|
76
|
Rodriguez-Muñoz A, Motahari-Rad H, Martin-Chaves L, Benitez-Porres J, Rodriguez-Capitan J, Gonzalez-Jimenez A, Insenser M, Tinahones FJ, Murri M. A Systematic Review of Proteomics in Obesity: Unpacking the Molecular Puzzle. Curr Obes Rep 2024; 13:403-438. [PMID: 38703299 PMCID: PMC11306592 DOI: 10.1007/s13679-024-00561-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/14/2024] [Indexed: 05/06/2024]
Abstract
PURPOSE OF REVIEW The present study aims to review the existing literature to identify pathophysiological proteins in obesity by conducting a systematic review of proteomics studies. Proteomics may reveal the mechanisms of obesity development and clarify the links between obesity and related diseases, improving our comprehension of obesity and its clinical implications. RECENT FINDINGS Most of the molecular events implicated in obesity development remain incomplete. Proteomics stands as a powerful tool for elucidating the intricate interactions among proteins in the context of obesity. This methodology has the potential to identify proteins involved in pathological processes and to evaluate changes in protein abundance during obesity development, contributing to the identification of early disease predisposition, monitoring the effectiveness of interventions and improving disease management overall. Despite many non-targeted proteomic studies exploring obesity, a comprehensive and up-to-date systematic review of the molecular events implicated in obesity development is lacking. The lack of such a review presents a significant challenge for researchers trying to interpret the existing literature. This systematic review was conducted following the PRISMA guidelines and included sixteen human proteomic studies, each of which delineated proteins exhibiting significant alterations in obesity. A total of 41 proteins were reported to be altered in obesity by at least two or more studies. These proteins were involved in metabolic pathways, oxidative stress responses, inflammatory processes, protein folding, coagulation, as well as structure/cytoskeleton. Many of the identified proteomic biomarkers of obesity have also been reported to be dysregulated in obesity-related disease. Among them, seven proteins, which belong to metabolic pathways (aldehyde dehydrogenase and apolipoprotein A1), the chaperone family (albumin, heat shock protein beta 1, protein disulfide-isomerase A3) and oxidative stress and inflammation proteins (catalase and complement C3), could potentially serve as biomarkers for the progression of obesity and the development of comorbidities, contributing to personalized medicine in the field of obesity. Our systematic review in proteomics represents a substantial step forward in unravelling the complexities of protein alterations associated with obesity. It provides valuable insights into the pathophysiological mechanisms underlying obesity, thereby opening avenues for the discovery of potential biomarkers and the development of personalized medicine in obesity.
Collapse
Affiliation(s)
- Alba Rodriguez-Muñoz
- Endocrinology and Nutrition UGC, Hospital Universitario Virgen de La Victoria, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Hospital Clínico Virgen de La Victoria, Málaga, Spain
- CIBER Fisiopatología de La Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Málaga, Spain
| | - Hanieh Motahari-Rad
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Laura Martin-Chaves
- Heart Area, Hospital Universitario Virgen de La Victoria, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Malaga, Spain
- Department of Dermatology and Medicine, Faculty of Medicine, University of Malaga, Malaga, Spain
| | - Javier Benitez-Porres
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Hospital Clínico Virgen de La Victoria, Málaga, Spain
- Department of Human Physiology, Physical Education and Sport, Faculty of Medicine, University of Malaga, Malaga, Spain
| | - Jorge Rodriguez-Capitan
- Heart Area, Hospital Universitario Virgen de La Victoria, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Malaga, Spain
- Biomedical Research Network Center for Cardiovascular Diseases (CIBERCV), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | | | - Maria Insenser
- Diabetes, Obesity and Human Reproduction Research Group, Department of Endocrinology & Nutrition, Hospital Universitario Ramón y Cajal & Universidad de Alcalá & Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) & Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain.
| | - Francisco J Tinahones
- Endocrinology and Nutrition UGC, Hospital Universitario Virgen de La Victoria, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Hospital Clínico Virgen de La Victoria, Málaga, Spain
- CIBER Fisiopatología de La Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Málaga, Spain
- Department of Dermatology and Medicine, Faculty of Medicine, University of Malaga, Malaga, Spain
| | - Mora Murri
- Endocrinology and Nutrition UGC, Hospital Universitario Virgen de La Victoria, Málaga, Spain.
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Hospital Clínico Virgen de La Victoria, Málaga, Spain.
- CIBER Fisiopatología de La Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Málaga, Spain.
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
77
|
Esteves JV, Stanford KI. Exercise as a tool to mitigate metabolic disease. Am J Physiol Cell Physiol 2024; 327:C587-C598. [PMID: 38981607 PMCID: PMC11427015 DOI: 10.1152/ajpcell.00144.2024] [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: 03/12/2024] [Revised: 06/28/2024] [Accepted: 06/28/2024] [Indexed: 07/11/2024]
Abstract
Metabolic diseases, notably obesity and type 2 diabetes (T2D), have reached alarming proportions and constitute a significant global health challenge, emphasizing the urgent need for effective preventive and therapeutic strategies. In contrast, exercise training emerges as a potent intervention, exerting numerous positive effects on metabolic health through adaptations to the metabolic tissues. Here, we reviewed the major features of our current understanding with respect to the intricate interplay between metabolic diseases and key metabolic tissues, including adipose tissue, skeletal muscle, and liver, describing some of the main underlying mechanisms driving pathogenesis, as well as the role of exercise to combat and treat obesity and metabolic disease.
Collapse
Affiliation(s)
- Joao Victor Esteves
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States
- Division of General and Gastrointestinal Surgery, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States
| | - Kristin I Stanford
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States
- Division of General and Gastrointestinal Surgery, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States
| |
Collapse
|
78
|
Ulusoy-Gezer HG, Rakıcıoğlu N. The Future of Obesity Management through Precision Nutrition: Putting the Individual at the Center. Curr Nutr Rep 2024; 13:455-477. [PMID: 38806863 PMCID: PMC11327204 DOI: 10.1007/s13668-024-00550-y] [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] [Accepted: 05/18/2024] [Indexed: 05/30/2024]
Abstract
PURPOSE OF REVIEW: The prevalence of obesity continues to rise steadily. While obesity management typically relies on dietary and lifestyle modifications, individual responses to these interventions vary widely. Clinical guidelines for overweight and obesity stress the importance of personalized approaches to care. This review aims to underscore the role of precision nutrition in delivering tailored interventions for obesity management. RECENT FINDINGS: Recent technological strides have expanded our ability to detect obesity-related genetic polymorphisms, with machine learning algorithms proving pivotal in analyzing intricate genomic data. Machine learning algorithms can also predict postprandial glucose, triglyceride, and insulin levels, facilitating customized dietary interventions and ultimately leading to successful weight loss. Additionally, given that adherence to dietary recommendations is one of the key predictors of weight loss success, employing more objective methods for dietary assessment and monitoring can enhance sustained long-term compliance. Biomarkers of food intake hold promise for a more objective dietary assessment. Acknowledging the multifaceted nature of obesity, precision nutrition stands poised to transform obesity management by tailoring dietary interventions to individuals' genetic backgrounds, gut microbiota, metabolic profiles, and behavioral patterns. However, there is insufficient evidence demonstrating the superiority of precision nutrition over traditional dietary recommendations. The integration of precision nutrition into routine clinical practice requires further validation through randomized controlled trials and the accumulation of a larger body of evidence to strengthen its foundation.
Collapse
Affiliation(s)
- Hande Gül Ulusoy-Gezer
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Hacettepe University, 06100, Sıhhiye, Ankara, Türkiye
| | - Neslişah Rakıcıoğlu
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Hacettepe University, 06100, Sıhhiye, Ankara, Türkiye.
| |
Collapse
|
79
|
Delfan M, Radkia F, Juybari RA, Daneshyar S, Willems ME, Saeidi A, Hackney AC, Laher I, Zouhal H. Unveiling the Effects of Interval Resistance Training and Chlorella Vulgaris Supplementation on Meteorin-like Protein and Oxidative Stress in Obese Men. Curr Dev Nutr 2024; 8:104428. [PMID: 39279784 PMCID: PMC11402038 DOI: 10.1016/j.cdnut.2024.104428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 07/04/2024] [Accepted: 07/21/2024] [Indexed: 09/18/2024] Open
Abstract
Background Dysregulation of adipocyte function occurs in obesity. Meteorin-like protein (Metrnl) is a newly discovered modulator of inflammation, metabolism, and differentiation of human adipocytes. The dietary supplement Chlorella Vulgaris (CV) reduces hyperlipidemia, hyperglycemia, and oxidative stress in clinical trials. Objectives To explore the impact of 12 wks of interval resistance training (IRT) and CV supplementation on plasma levels of Metrnl and oxidative stress in males with obesity. Methods Forty-four obese men (BMI: 32.0 ± 1.5 kg/m2, weight: 101.1 ± 2.2 kg, age: 23-35 years) were randomly assigned into 4 groups (n = 11/group): control (CON), CV supplement (CV), IRT, and CV + IRT (CVIRT). The IRT was performed for 12 wks (3 sessions per week). The treatment consisted of a daily intake of CV (1800 mg capsule) or placebo capsules. Blood samples were collected 48 hours before and after the interventions to analyze biomedical measurements. Results The IRT and CVIRT groups had elevations in plasma Metrnl, superoxide dismutase, and total antioxidant capacity levels (all P < 0.0001), and reductions in malondialdehyde (P < 0.0001). Supplementation with CV significantly reduced malondialdehyde (P < 0.001) and increased total antioxidant capacity (P < 0.0001) but failed to alter superoxide dismutase or Metrnl (P > 0.05). Conclusions Although IRT and its combination with CV hold promise for improving Metrnl levels and oxidative status in obesity, combining IRT and CV do not yield greater benefits than IRT alone. Although standalone CV supplementation could favorably impact certain markers of oxidative stress, the effectiveness of CV supplementation appears to have a relatively limited effect across assessed biomarkers and requires further investigation.
Collapse
Affiliation(s)
- Maryam Delfan
- Department of Exercise Physiology, Faculty of Sport Sciences, Alzahra University, Tehran, Iran
| | - Fatemeh Radkia
- Department of Exercise Physiology, Faculty of Sport Sciences, Alzahra University, Tehran, Iran
| | - Raheleh Amadeh Juybari
- Department of Exercise Physiology, Faculty of Sport Sciences, Alzahra University, Tehran, Iran
| | - Saeed Daneshyar
- Department of Physical Education, Hamedan University of Technology, Hamedan, Iran
| | - Mark Et Willems
- Institute of Applied Sciences, University of Chichester, Chichester, United Kingdom
| | - Ayoub Saeidi
- Department of Physical Education and Sport Sciences, Faculty of Humanities and Social Sciences, University of Kurdistan, Sanandaj, Kurdistan, Iran
| | - Anthony C Hackney
- Department of Exercise and Sport Science, University of North Carolina, Chapel Hill, North Carolina, United States
| | - Ismail Laher
- Department of Anesthesiology, Pharmacology, and Therapeutics, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Hassane Zouhal
- Univ Rennes, M2S (Laboratoire Mouvement, Sport, Santé), Rennes, France
- Institut International des Sciences du Sport, Irodouer, France
| |
Collapse
|
80
|
Iyer M, Firkins SA, Patel R, Flora B, Staneff E, Simons-Linares R. Endoscopic and Pharmacologic Treatment of Obesity in Patients With Hereditary Polyposis Syndromes. ACG Case Rep J 2024; 11:e01514. [PMID: 39267621 PMCID: PMC11392469 DOI: 10.14309/crj.0000000000001514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 08/16/2024] [Indexed: 09/15/2024] Open
Abstract
Patients with hereditary polyposis syndromes (HPS) are among the highest risk of multiple types of cancer. This risk is further magnified by comorbid obesity; however, HPS present unique risks for bariatric surgery. The advent of endoscopic bariatric and metabolic therapies along with advancements in the realm of antiobesity medications provides potential weight loss alternatives in this vulnerable population. We present 2 cases of patients with obesity and HPS successfully treated with intragastric balloons in combination with antiobesity medications.
Collapse
Affiliation(s)
- Meghana Iyer
- Cleveland Clinic Lerner College of Medicine, Cleveland Clinic Foundation, Cleveland, OH
| | - Stephen A Firkins
- Digestive Disease and Surgery Institute, Cleveland Clinic Foundation, Cleveland, OH
| | - Roma Patel
- Digestive Disease and Surgery Institute, Cleveland Clinic Foundation, Cleveland, OH
| | - Bailey Flora
- Digestive Disease and Surgery Institute, Cleveland Clinic Foundation, Cleveland, OH
| | - Erika Staneff
- Digestive Disease and Surgery Institute, Cleveland Clinic Foundation, Cleveland, OH
| | | |
Collapse
|
81
|
Liu K, Zhou D, Chen L, Hao S. Depression and type 2 diabetes risk: a Mendelian randomization study. Front Endocrinol (Lausanne) 2024; 15:1436411. [PMID: 39268231 PMCID: PMC11390465 DOI: 10.3389/fendo.2024.1436411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 08/05/2024] [Indexed: 09/15/2024] Open
Abstract
Background Extensive observational evidence has suggested an association between depression and type 2 diabetes (T2D). However, the causal relationships between these two diseases require further investigation. This study aimed to evaluate the bidirectional causal effect between two types of depression and T2D using two-sample Mendelian randomization (MR). Methods We applied two-step MR techniques, using single-nucleotide polymorphisms (SNPs) as the genetic instruments for analysis. We utilized summary data from genome-wide association studies (GWASs) for major depression (MD), depressive status (frequency of depressed mood in the last two weeks), T2D, and other known T2D risk factors such as obesity, sedentary behavior (time spent watching television), and blood pressure. The analysis utilized inverse variance weighted (IVW), MR-Egger regression, weighted median, weighted mode, MR pleiotropy residual sum, and outlier methods to determine potential causal relationships. Results The study found that MD was positively associated with T2D, with an odds ratio (OR) of 1.26 (95% CI: 1.10-1.43, p = 5.6×10-4) using the IVW method and an OR of 1.21 (95% CI: 1.04-1.41, p = 0.01) using the weighted median method. Depressive status was also positively associated with T2D, with an OR of 2.26 (95% CI: 1.03-4.94, p = 0.04) and an OR of 3.62 (95% CI: 1.33-9.90, p = 0.01) using the IVW and weighted median methods, respectively. No causal effects of MD and depressive status on T2D risk factors were observed, and T2D did not influence these factors. Conclusion Our study demonstrates a causal relationship between depression and an increased risk of developing T2D, with both major depression and depressive status being positively associated with T2D.
Collapse
Affiliation(s)
- Kaiyuan Liu
- Department of Endocrinology, Zhejiang Integrated Traditional Chinese and Western Medicine Hospital, Hangzhou, Zhejiang, China
| | - Diyi Zhou
- Department of Endocrinology, Zhejiang Integrated Traditional Chinese and Western Medicine Hospital, Hangzhou, Zhejiang, China
| | - Lijun Chen
- Department of Endocrinology, Zhejiang Integrated Traditional Chinese and Western Medicine Hospital, Hangzhou, Zhejiang, China
| | - Sida Hao
- Department of Urology, Zhejiang Integrated Traditional Chinese and Western Medicine Hospital, Hangzhou, Zhejiang, China
| |
Collapse
|
82
|
Wang B, Hu Z, Cui L, Zhao M, Su Z, Jiang Y, Liu J, Zhao Y, Hou Y, Yang X, Zhang C, Guo B, Li D, Zhao L, Zheng S, Zhao Y, Yang W, Wang D, Yu S, Zhu S, Yan Y, Yuan G, Li K, Zhang W, Qin L, Zhang W, Sun F, Luo J, Zheng R. βAR-mTOR-lipin1 pathway mediates PKA-RIIβ deficiency-induced adipose browning. Theranostics 2024; 14:5316-5335. [PMID: 39267778 PMCID: PMC11388065 DOI: 10.7150/thno.97046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 08/16/2024] [Indexed: 09/15/2024] Open
Abstract
Background: Enhancing white adipose tissue (WAT) browning combats obesity. The RIIβ subunit of cAMP-dependent protein kinase (PKA) is primarily expressed in the brain and adipose tissue. Deletion of the hypothalamic RIIβ gene centrally induces WAT browning, yet the peripheral mechanisms mediating this process remain unexplored. Methods: This study investigates the mechanisms underlying WAT browning in RIIβ-KO mice. Genetic approaches such as β3-adrenergic receptors (β3ARs) deletion and sympathetic denervation of WAT were utilized. Genome-wide transcriptomic sequencing and bioinformatic analysis were employed to identify potential mediators of WAT browning. siRNA assays were employed to knock down mTOR and lipin1 in vitro, while AAV-shRNAs were used for the same purpose in vivo. Results: We found that WAT browning substantially contributes to the lean and obesity-resistant phenotypes of RIIβ-KO mice. The WAT browning can be dampened by β3ARs deletion or WAT sympathetic denervation. We identified that adipocytic mTOR and lipin1 may act as mediators of the WAT browning. Inhibition of mTOR or lipin1 abrogates WAT browning and hinders the lean phenotype of RIIβ-KO mice. In human subcutaneous white adipocytes and mouse white adipocytes, β3AR stimulation can activate mTOR and causes lipin1 nuclear translocation; knockdown of mTOR and Lipin1 mitigates WAT browning-associated gene expression, impedes mitochondrial activity. Moreover, mTOR knockdown reduces lipin1 level and nuclear translocation, indicating that lipin1 may act downstream of mTOR. Additionally, in vivo knockdown of mTOR and Lipin1 diminished WAT browning and increased adiposity. Conclusions: The β3AR-activated mTOR-lipin1 axis mediates WAT browning, offering new insights into the molecular basis of PKA-regulated WAT browning. These findings provide potential adipose target candidates for the development of drugs to treat obesity.
Collapse
Affiliation(s)
- Bingwei Wang
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University, Beijing, China
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, China
| | - Zhiping Hu
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Peking University, Beijing, China
- Present address: Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Long Cui
- Department of General Surgery, Peking University Third Hospital, Peking University, Beijing, China
| | - Miao Zhao
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Zhijie Su
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yong Jiang
- Department of General Surgery, Peking University First Hospital, Peking University, Beijing, China
| | - Jiarui Liu
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yun Zhao
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yujia Hou
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Xiaoning Yang
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Chenyu Zhang
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Bingbing Guo
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Daotong Li
- National Engineering Research Center for Fruit and Vegetable Processing, Key Laboratory of Fruits and Vegetables Processing, College of Food Science and Nutritional Engineering, Ministry of Agriculture, Engineering Research Centre for Fruits and Vegetables Processing, Ministry of Education, China Agricultural University, Beijing, China
| | - Liang Zhao
- Department of Obstetrics and Gynecology, Beijing Jishuitan Hospital, Peking University, Beijing, China
| | - Shengmin Zheng
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Peking University, Beijing, China
| | - Yiguo Zhao
- Department of Gastrointestinal Surgery, Peking University International Hospital, Peking University, Beijing, China
| | - Weipeng Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Dunfang Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Siwang Yu
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, Peking University, Beijing, China
| | - Shigong Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yi Yan
- Department of Sport Biochemistry, School of Sport Science, Beijing Sport University, Beijing, China
| | - Geheng Yuan
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| | - Kailong Li
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Wenqiang Zhang
- College of Engineering, China Agricultural University, Beijing, China
| | - Lihua Qin
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Weiguang Zhang
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Feng Sun
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Jianyuan Luo
- Department of Medical Genetics, School of Basic Medical Sciences, Peking University, Beijing, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Ruimao Zheng
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University, Beijing, China
- Neuroscience Research Institute, Peking University, Beijing, China
- Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing, China
- Beijing Life Science Academy, Beijing, China
| |
Collapse
|
83
|
Dayal Aggarwal D, Mishra P, Yadav G, Mitra S, Patel Y, Singh M, Sahu RK, Sharma V. Decoding the connection between lncRNA and obesity: Perspective from humans and Drosophila. Heliyon 2024; 10:e35327. [PMID: 39166041 PMCID: PMC11334870 DOI: 10.1016/j.heliyon.2024.e35327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 07/20/2024] [Accepted: 07/26/2024] [Indexed: 08/22/2024] Open
Abstract
Background Obesity is a burgeoning global health problem with an escalating prevalence and severe implications for public health. New evidence indicates that long non-coding RNAs (lncRNAs) may play a pivotal role in regulating adipose tissue function and energy homeostasis across various species. However, the molecular mechanisms underlying obesity remain elusive. Scope of review This review discusses obesity and fat metabolism in general, highlighting the emerging importance of lncRNAs in modulating adipogenesis. It describes the regulatory networks, latest tools, techniques, and approaches to enhance our understanding of obesity and its lncRNA-mediated epigenetic regulation in humans and Drosophila. Major conclusions This review analyses large datasets of human and Drosophila lncRNAs from published databases and literature with experimental evidence supporting lncRNAs role in fat metabolism. It concludes that lncRNAs play a crucial role in obesity-related metabolism. Cross-species comparisons highlight the relevance of Drosophila findings to human obesity, emphasizing their potential role in adipose tissue biology. Furthermore, it discusses how recent technological advancements and multi-omics data integration enhance our capacity to characterize lncRNAs and their function. Additionally, this review briefly touches upon innovative methodologies like experimental evolution and advanced sequencing technologies for identifying novel genes and lncRNA regulators in Drosophila, which can potentially contribute to obesity research.
Collapse
Affiliation(s)
- Dau Dayal Aggarwal
- Department of Biochemistry, University of Delhi South Campus, New Delhi, India
| | - Prachi Mishra
- Department of Biochemistry, University of Delhi South Campus, New Delhi, India
| | - Gaurav Yadav
- Department of Biochemistry, University of Delhi South Campus, New Delhi, India
| | - Shrishti Mitra
- Department of Biochemistry, University of Delhi South Campus, New Delhi, India
| | - Yashvant Patel
- Department of Zoology, Banaras Hindu University, Varanasi, India
| | - Manvender Singh
- Department of Biotechnology, UIET, MD University, Rohtak, India
| | - Ranjan Kumar Sahu
- Department of Neurology, Houston Methodist Research Insititute, Houston, Tx, USA
| | - Vijendra Sharma
- Department of Biomedical Sciences, University of Windsor, Ontario, Canada
| |
Collapse
|
84
|
Wu C, Yang F, Zhong H, Hong J, Lin H, Zong M, Ren H, Zhao S, Chen Y, Shi Z, Wang X, Shen J, Wang Q, Ni M, Chen B, Cai Z, Zhang M, Cao Z, Wu K, Gao A, Li J, Liu C, Xiao M, Li Y, Shi J, Zhang Y, Xu X, Gu W, Bi Y, Ning G, Wang W, Wang J, Liu R. Obesity-enriched gut microbe degrades myo-inositol and promotes lipid absorption. Cell Host Microbe 2024; 32:1301-1314.e9. [PMID: 38996548 DOI: 10.1016/j.chom.2024.06.012] [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: 11/29/2023] [Revised: 04/29/2024] [Accepted: 06/14/2024] [Indexed: 07/14/2024]
Abstract
Numerous studies have reported critical roles for the gut microbiota in obesity. However, the specific microbes that causally contribute to obesity and the underlying mechanisms remain undetermined. Here, we conducted shotgun metagenomic sequencing in a Chinese cohort of 631 obese subjects and 374 normal-weight controls and identified a Megamonas-dominated, enterotype-like cluster enriched in obese subjects. Among this cohort, the presence of Megamonas and polygenic risk exhibited an additive impact on obesity. Megamonas rupellensis possessed genes for myo-inositol degradation, as demonstrated in vitro and in vivo, and the addition of myo-inositol effectively inhibited fatty acid absorption in intestinal organoids. Furthermore, mice colonized with M. rupellensis or E. coli heterologously expressing the myo-inositol-degrading iolG gene exhibited enhanced intestinal lipid absorption, thereby leading to obesity. Altogether, our findings uncover roles for M. rupellensis as a myo-inositol degrader that enhances lipid absorption and obesity, suggesting potential strategies for future obesity management.
Collapse
Affiliation(s)
- Chao Wu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fangming Yang
- BGI Research, Shenzhen 518083, China; Institute of Intelligent Medical Research (IIMR), BGI Genomics, Shenzhen 518083, China
| | - Huanzi Zhong
- BGI Research, Shenzhen 518083, China; Institute of Intelligent Medical Research (IIMR), BGI Genomics, Shenzhen 518083, China
| | - Jie Hong
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huibin Lin
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingxi Zong
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huahui Ren
- BGI Research, Shenzhen 518083, China; Institute of Intelligent Medical Research (IIMR), BGI Genomics, Shenzhen 518083, China
| | - Shaoqian Zhao
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yufei Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhun Shi
- BGI Research, Shenzhen 518083, China; Institute of Intelligent Medical Research (IIMR), BGI Genomics, Shenzhen 518083, China
| | - Xingyu Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Juan Shen
- BGI Research, Shenzhen 518083, China
| | - Qiaoling Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mengshan Ni
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Banru Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhongle Cai
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Minchun Zhang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiwen Cao
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kui Wu
- BGI Research, Shenzhen 518083, China; Institute of Intelligent Medical Research (IIMR), BGI Genomics, Shenzhen 518083, China; Guangdong Provincial Key Laboratory of Human Disease Genomics, Shenzhen Key Laboratory of Genomics, BGI Research, Shenzhen 518083, China
| | - Aibo Gao
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junhua Li
- BGI Research, Shenzhen 518083, China
| | - Cong Liu
- BGI Research, Shenzhen 518083, China
| | | | - Yan Li
- BGI Research, Shenzhen 518083, China
| | - Juan Shi
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yifei Zhang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xun Xu
- BGI Research, Shenzhen 518083, China
| | - Weiqiong Gu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yufang Bi
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guang Ning
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Weiqing Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jiqiu Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Ruixin Liu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
85
|
Trang KB, Pahl MC, Pippin JA, Su C, Littleton SH, Sharma P, Kulkarni NN, Ghanem LR, Terry NA, O’Brien JM, Wagley Y, Hankenson KD, Jermusyk A, Hoskins JW, Amundadottir LT, Xu M, Brown KM, Anderson SA, Yang W, Titchenell PM, Seale P, Cook L, Levings MK, Zemel BS, Chesi A, Wells AD, Grant SF. 3D genomic features across >50 diverse cell types reveal insights into the genomic architecture of childhood obesity. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.08.30.23294092. [PMID: 37693606 PMCID: PMC10491377 DOI: 10.1101/2023.08.30.23294092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
The prevalence of childhood obesity is increasing worldwide, along with the associated common comorbidities of type 2 diabetes and cardiovascular disease in later life. Motivated by evidence for a strong genetic component, our prior genome-wide association study (GWAS) efforts for childhood obesity revealed 19 independent signals for the trait; however, the mechanism of action of these loci remains to be elucidated. To molecularly characterize these childhood obesity loci we sought to determine the underlying causal variants and the corresponding effector genes within diverse cellular contexts. Integrating childhood obesity GWAS summary statistics with our existing 3D genomic datasets for 57 human cell types, consisting of high-resolution promoter-focused Capture-C/Hi-C, ATAC-seq, and RNA-seq, we applied stratified LD score regression and calculated the proportion of genome-wide SNP heritability attributable to cell type-specific features, revealing pancreatic alpha cell enrichment as the most statistically significant. Subsequent chromatin contact-based fine-mapping was carried out for genome-wide significant childhood obesity loci and their linkage disequilibrium proxies to implicate effector genes, yielded the most abundant number of candidate variants and target genes at the BDNF, ADCY3, TMEM18 and FTO loci in skeletal muscle myotubes and the pancreatic beta-cell line, EndoC-BH1. One novel implicated effector gene, ALKAL2 - an inflammation-responsive gene in nerve nociceptors - was observed at the key TMEM18 locus across multiple immune cell types. Interestingly, this observation was also supported through colocalization analysis using expression quantitative trait loci (eQTL) derived from the Genotype-Tissue Expression (GTEx) dataset, supporting an inflammatory and neurologic component to the pathogenesis of childhood obesity. Our comprehensive appraisal of 3D genomic datasets generated in a myriad of different cell types provides genomic insights into pediatric obesity pathogenesis.
Collapse
Affiliation(s)
- Khanh B. Trang
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Matthew C. Pahl
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - James A. Pippin
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Chun Su
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sheridan H. Littleton
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Prabhat Sharma
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pathology, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Nikhil N. Kulkarni
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pathology, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Louis R. Ghanem
- Division of Gastroenterology, Hepatology, and Nutrition, Children’s Hospital of Philadelphia, PA, USA
| | - Natalie A. Terry
- Division of Gastroenterology, Hepatology, and Nutrition, Children’s Hospital of Philadelphia, PA, USA
| | - Joan M. O’Brien
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, PA, USA
- Penn Medicine Center for Ophthalmic Genetics in Complex Disease
| | - Yadav Wagley
- Department of Orthopedic Surgery University of Michigan Medical School Ann Arbor, MI, USA
| | - Kurt D. Hankenson
- Department of Orthopedic Surgery University of Michigan Medical School Ann Arbor, MI, USA
| | - Ashley Jermusyk
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Jason W. Hoskins
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Laufey T. Amundadottir
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Mai Xu
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Kevin M Brown
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Stewart A. Anderson
- Department of Child and Adolescent Psychiatry, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Wenli Yang
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Paul M. Titchenell
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Patrick Seale
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Laura Cook
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Department of Critical Care, Melbourne Medical School, University of Melbourne, Melbourne, VIC, Australia
- Division of Infectious Diseases, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Megan K. Levings
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada
- BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Babette S. Zemel
- Division of Gastroenterology, Hepatology, and Nutrition, Children’s Hospital of Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alessandra Chesi
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pathology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrew D. Wells
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pathology, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pathology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Struan F.A. Grant
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division Endocrinology and Diabetes, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
86
|
Vranceanu M, Filip L, Hegheș SC, de Lorenzo D, Cozma-Petruț A, Ghitea TC, Stroia CM, Banc R, Mîrza OM, Miere D, Cozma V, Popa DS. Genes Involved in Susceptibility to Obesity and Emotional Eating Behavior in a Romanian Population. Nutrients 2024; 16:2652. [PMID: 39203789 PMCID: PMC11357152 DOI: 10.3390/nu16162652] [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: 07/15/2024] [Revised: 08/06/2024] [Accepted: 08/08/2024] [Indexed: 09/03/2024] Open
Abstract
Obesity, a significant public health concern with high prevalence in both adults and children, is a complex disorder arising from the interaction of multiple genes and environmental factors. Advances in genome-wide association studies (GWAS) and sequencing technologies have identified numerous polygenic causes of obesity, particularly genes involved in hunger, satiety signals, adipocyte differentiation, and energy expenditure. This study investigates the relationship between six obesity-related genes (CLOCK, FTO, GHRL, LEP, LEPR, MC4R) and their impact on BMI, WC, HC, WHR, and emotional eating behavior in 220 Romanian adults. Emotional eating was assessed using the validated Emotional Eating Questionnaire (EEQ). Our analysis revealed significant variability in obesity-related phenotypes and emotional eating behaviors across different genotypes. Specifically, CLOCK/CC, FTO/AA, and LEP/AA genotypes were strongly associated with higher obesity metrics and emotional eating scores, while GHRL/TT and MC4R/CC were linked to increased BMI and WHR. The interplay between genetic predisposition and emotional eating behavior significantly influenced BMI and WHR, indicating a complex relationship between genetic and behavioral factors. This study, the first of its kind in Romania, provides a foundation for targeted interventions to prevent and reduce obesity and suggests potential strategies for gene expression modulation to mitigate the effects of emotional eating. Adopting a 'One Health' approach by creating an evidence base derived from both human and animal studies is crucial for understanding how to control obesity.
Collapse
Affiliation(s)
- Maria Vranceanu
- Department of Toxicology, Faculty of Pharmacy, “Iuliu Hatieganu” University of Medicine and Pharmacy, 6 Pasteur Street, 400349 Cluj-Napoca, Romania (D.-S.P.)
| | - Lorena Filip
- Department of Bromatology, Hygiene, Nutrition, Faculty of Pharmacy, “Iuliu Hatieganu” University of Medicine and Pharmacy, 6 Pasteur Street, 400349 Cluj-Napoca, Romania; (A.C.-P.); (R.B.); (O.M.M.); (D.M.)
- Academy of Romanian Scientists (AOSR), 3 Ilfov St, 050044 Bucharest, Romania
| | - Simona-Codruța Hegheș
- Department of Drug Analysis, Facullty of Pharmacy, “Iuliu Hatieganu” University of Medicine and Pharmacy, 6 Pasteur Street, 400349 Cluj-Napoca, Romania;
| | - David de Lorenzo
- UCL Great Ormond Street Institute of Child Health, 30 Guilford St, London WC1N 1EH, UK;
| | - Anamaria Cozma-Petruț
- Department of Bromatology, Hygiene, Nutrition, Faculty of Pharmacy, “Iuliu Hatieganu” University of Medicine and Pharmacy, 6 Pasteur Street, 400349 Cluj-Napoca, Romania; (A.C.-P.); (R.B.); (O.M.M.); (D.M.)
| | - Timea Claudia Ghitea
- Doctoral Scool of Biomedical Sciences, Faculty of Medicine and Pharmacy, University of Oradea, 1 Universităţii Street, 410087 Oradea, Romania; (T.C.G.)
| | - Carmina Mariana Stroia
- Doctoral Scool of Biomedical Sciences, Faculty of Medicine and Pharmacy, University of Oradea, 1 Universităţii Street, 410087 Oradea, Romania; (T.C.G.)
| | - Roxana Banc
- Department of Bromatology, Hygiene, Nutrition, Faculty of Pharmacy, “Iuliu Hatieganu” University of Medicine and Pharmacy, 6 Pasteur Street, 400349 Cluj-Napoca, Romania; (A.C.-P.); (R.B.); (O.M.M.); (D.M.)
| | - Oana Maria Mîrza
- Department of Bromatology, Hygiene, Nutrition, Faculty of Pharmacy, “Iuliu Hatieganu” University of Medicine and Pharmacy, 6 Pasteur Street, 400349 Cluj-Napoca, Romania; (A.C.-P.); (R.B.); (O.M.M.); (D.M.)
| | - Doina Miere
- Department of Bromatology, Hygiene, Nutrition, Faculty of Pharmacy, “Iuliu Hatieganu” University of Medicine and Pharmacy, 6 Pasteur Street, 400349 Cluj-Napoca, Romania; (A.C.-P.); (R.B.); (O.M.M.); (D.M.)
| | - Vasile Cozma
- Department of Parasitology and Parasitic Diseases, Faculty of Veterinary Medicine, University of Agricultural Sciences and Veterinary Medicine of Cluj-Napoca, 3–5, Mănăştur Street, 400372 Cluj-Napoca, Romania;
- Academy of Agricultural and Forestry Sciences Gheorghe Ionescu-Siseşti (A.S.A.S.), 61, Mărăști Boulevard, 011464 Bucharest, Romania
| | - Daniela-Saveta Popa
- Department of Toxicology, Faculty of Pharmacy, “Iuliu Hatieganu” University of Medicine and Pharmacy, 6 Pasteur Street, 400349 Cluj-Napoca, Romania (D.-S.P.)
| |
Collapse
|
87
|
Anazco D, Acosta A. Precision medicine for obesity: current evidence and insights for personalization of obesity pharmacotherapy. Int J Obes (Lond) 2024:10.1038/s41366-024-01599-z. [PMID: 39127792 DOI: 10.1038/s41366-024-01599-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 06/17/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024]
Abstract
Obesity is a chronic and complex disease associated with increased morbidity, mortality, and financial burden. It is expected that by 2030 one of two people in the United States will have obesity. The backbone for obesity management continues to be lifestyle interventions, consisting of calorie deficit diets and increased physical activity levels, however, these interventions are often insufficient to achieve sufficient and maintained weight loss. As a result, multiple patients require additional interventions such as antiobesity medications or bariatric interventions in order to achieve clinically significant weight loss and improvement or resolution of obesity-associated comorbidities. Despite the recent advances in the field of obesity pharmacotherapy that have resulted in never-before-seen weight loss outcomes, comorbidity improvement, and even reduction in cardiovascular mortality, there is still a significant interindividual variability in terms of response to antiobesity medications, with a subset of patients not achieving a clinically significant weight loss. Currently, the trial-and-error paradigm for the selection of antiobesity medications results in increased costs and risks for developing side effects, while also reduces engagement in weight management programs for patients with obesity. The implementation of a precision medicine framework to the selection of antiobesity medications might help reduce heterogeneity and optimize weight loss outcomes by identifying unique subsets of patients, or phenotypes, that have a better response to a specific intervention. The detailed study of energy balance regulation holds promise, as actionable behavioral and physiologic traits could help guide antiobesity medication selection based on previous mechanistic studies. Moreover, the rapid advances in genotyping, multi-omics, and big data analysis might hold the key to discover additional signatures or phenotypes that might respond better to a certain intervention and might permit the widespread adoption of a precision medicine approach for obesity management.
Collapse
Affiliation(s)
- Diego Anazco
- 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
|
88
|
Qiu X, Kuang J, Huang Y, Wei C, Zheng X. The association between Weight-adjusted-Waist Index (WWI) and cognitive function in older adults: a cross-sectional NHANES 2011-2014 study. BMC Public Health 2024; 24:2152. [PMID: 39118100 PMCID: PMC11308487 DOI: 10.1186/s12889-024-19332-w] [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: 09/20/2023] [Accepted: 07/01/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND The impact of obesity on cognitive function has engendered considerable interest. Weight-adjusted waist index (WWI) has emerged as a novel and innovative marker of obesity that reflects weight-independent abdominal obesity. However, the association between WWI and cognitive function remains unclear. To address this gap, the present study aims to explore the relationship between weight-adjusted waist index (WWI) and cognitive performance in older adults. METHODS We conducted a cross-sectional investigation using datasets from the National Health and Nutrition Examination Survey (NHANES) 2011-2014. The study included 3,472 participants (48.59% male, 51.41% female) of various races (Mexican American, Other Hispanic, Non-Hispanic White, Non-Hispanic Black, and Other), with a mean age of 69.95 years (SD = 6.94). Multivariate regression and smoothing curve fitting were used to investigate the linear and nonlinear relationship between WWI and cognitive performance in the following domains: learning and memory, verbal fluency, and processing speed, as measured by Consortium to Establish a Registry for Alzheimer's Disease Word Learning subtest (CERAD-WL), Animal Fluency Test (AFT), and Digit Symbol Substitution Test (DSST), respectively. Subgroup analysis and interaction tests were conducted to examine the stability of this relationship across groups. Machine learning models based on random forests were used to analyze the predictive performance of WWI for cognitive function. RESULTS A total of 3,472 participants were included in the analysis. The results revealed significant negative associations between WWI and low scores on the CERAD-WL [-0.96 (-1.30, -0.62)], AFT [-0.77 (-1.05, -0.49)], and DSST [-3.67 (-4.55, -2.79)]. This relationship remained stable after converting WWI to a categorical variable. In addition, this significant negative association was more pronounced in men than women and diminished with advancing age. Non-linear threshold effects were observed, with correlations intensifying between WWI and CERAD-WL when WWI surpassed 12.25, AFT when WWI surpassed 11.54, and DSST when WWI surpassed 11.66. CONCLUSIONS A higher WWI, indicating increased abdominal obesity, was associated with deficits in learning, memory, verbal fluency, and processing speed among older adults. These findings suggest that abdominal obesity may play a crucial role in cognitive decline in this population. The stronger relationship observed between WWI and cognition in men highlights the need for gender-specific considerations in interventions targeting abdominal obesity. The results demonstrate the importance of interventions targeting abdominal obesity to preserve cognitive performance in older adults.
Collapse
Affiliation(s)
- Xichenhui Qiu
- Health Science Center, Shenzhen University, No. 1066, Xueyuan Avenue, Nanshan District, Shenzhen, Guangdong Province, 518060, People's Republic of China
| | - Jiahao Kuang
- Health Science Center, Shenzhen University, No. 1066, Xueyuan Avenue, Nanshan District, Shenzhen, Guangdong Province, 518060, People's Republic of China
| | - Yiqing Huang
- Health Science Center, Shenzhen University, No. 1066, Xueyuan Avenue, Nanshan District, Shenzhen, Guangdong Province, 518060, People's Republic of China
| | - Changning Wei
- School of Tech X Academy, Shenzhen Polytechnic University, No. 7098, Liuxian Avenue Nanshan District, Shenzhen, Guangdong Province, 518118, People's Republic of China
| | - Xujuan Zheng
- Health Science Center, Shenzhen University, No. 1066, Xueyuan Avenue, Nanshan District, Shenzhen, Guangdong Province, 518060, People's Republic of China.
| |
Collapse
|
89
|
Chen J, Jia S, Guo C, Fan Z, Yan W, Dong K. Research Progress on the Effect and Mechanism of Exercise Intervention on Sarcopenia Obesity. Clin Interv Aging 2024; 19:1407-1422. [PMID: 39139211 PMCID: PMC11319865 DOI: 10.2147/cia.s473083] [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: 05/09/2024] [Accepted: 08/02/2024] [Indexed: 08/15/2024] Open
Abstract
With the increasingly severe situation of obesity and population aging, there is growing concern about sarcopenia obesity (SO). SO refers to the coexistence of obesity and sarcopenia, which imposes a heavier burden on individuals and society compared to obesity or sarcopenia alone. Therefore, comprehending the pathogenesis of SO and implementing effective clinical interventions are vital for its prevention and treatment. This review uses a comprehensive literature search and analysis of PubMed, Web of Science, and CNKI databases, with search terms including "Sarcopenic obesity", "exercise", "cytokines", "inflammation", "mitochondrial quality control", and "microRNA", covering relevant studies published up to July 2024. The results indicate that the pathogenesis of SO is complex, involving mechanisms like age-related changes in body composition, hormonal alterations, inflammation, mitochondrial dysfunction, and genetic and epigenetic factors. Regarding exercise interventions for SO, aerobic exercise can reduce fat mass, resistance exercise can increase skeletal muscle mass and strength, and combined exercise can achieve both, making it the optimal intervention for SO. The potential mechanisms by which exercise may prevent and treat SO include regulating cytokine secretion, inhibiting inflammatory pathways, improving mitochondrial quality, and mediating microRNA expression. This review emphasizes the effectiveness of exercise interventions in mitigating sarcopenic obesity through comprehensive analysis of its multifactorial pathogenesis and the mechanistic insights into exercise's therapeutic effects. Understanding these mechanisms informs targeted therapeutic strategies aimed at alleviating the societal and individual burdens associated with SO.
Collapse
Affiliation(s)
- Jun Chen
- School of Graduate, Wuhan Sport University, Wuhan, 430079, People’s Republic of China
| | - Shaohui Jia
- School of Sports Medicine, Wuhan Sport University, Wuhan, 430079, People’s Republic of China
| | - Chenggen Guo
- School of Sports Training, Wuhan Sport University, Wuhan, 430079, People’s Republic of China
| | - Zhiwei Fan
- School of Graduate, Wuhan Sport University, Wuhan, 430079, People’s Republic of China
| | - Weiyi Yan
- School of Graduate, Wuhan Sport University, Wuhan, 430079, People’s Republic of China
| | - Kunwei Dong
- School of Arts, Wuhan Sport University, Wuhan, 430079, People’s Republic of China
| |
Collapse
|
90
|
Adolph TE, Tilg H. Western diets and chronic diseases. Nat Med 2024; 30:2133-2147. [PMID: 39085420 DOI: 10.1038/s41591-024-03165-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 06/28/2024] [Indexed: 08/02/2024]
Abstract
'Westernization', which incorporates industrial, cultural and dietary trends, has paralleled the rise of noncommunicable diseases across the globe. Today, the Western-style diet emerges as a key stimulus for gut microbial vulnerability, chronic inflammation and chronic diseases, affecting mainly the cardiovascular system, systemic metabolism and the gut. Here we review the diet of modern times and evaluate the threat it poses for human health by summarizing recent epidemiological, translational and clinical studies. We discuss the links between diet and disease in the context of obesity and type 2 diabetes, cardiovascular diseases, gut and liver diseases and solid malignancies. We collectively interpret the evidence and its limitations and discuss future challenges and strategies to overcome these. We argue that healthcare professionals and societies must react today to the detrimental effects of the Western diet to bring about sustainable change and improved outcomes in the future.
Collapse
Affiliation(s)
- Timon E Adolph
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria.
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
91
|
Liu T, Woodruff PG, Zhou X. Advances in non-type 2 severe asthma: from molecular insights to novel treatment strategies. Eur Respir J 2024; 64:2300826. [PMID: 38697650 PMCID: PMC11325267 DOI: 10.1183/13993003.00826-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 04/18/2024] [Indexed: 05/05/2024]
Abstract
Asthma is a prevalent pulmonary disease that affects more than 300 million people worldwide and imposes a substantial economic burden. While medication can effectively control symptoms in some patients, severe asthma attacks, driven by airway inflammation induced by environmental and infectious exposures, continue to be a major cause of asthma-related mortality. Heterogeneous phenotypes of asthma include type 2 (T2) and non-T2 asthma. Non-T2 asthma is often observed in patients with severe and/or steroid-resistant asthma. This review covers the molecular mechanisms, clinical phenotypes, causes and promising treatments of non-T2 severe asthma. Specifically, we discuss the signalling pathways for non-T2 asthma including the activation of inflammasomes, interferon responses and interleukin-17 pathways, and their contributions to the subtypes, progression and severity of non-T2 asthma. Understanding the molecular mechanisms and genetic determinants underlying non-T2 asthma could form the basis for precision medicine in severe asthma treatment.
Collapse
Affiliation(s)
- Tao Liu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine and Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- Department of Biochemistry and Molecular Biology, School of Medicine, Southeast University, Nanjing, China
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Prescott G Woodruff
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine and Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
| | - Xiaobo Zhou
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
92
|
Tahiri I, Llana SR, Fos-Domènech J, Milà-Guash M, Toledo M, Haddad-Tóvolli R, Claret M, Obri A. Paternal obesity induces changes in sperm chromatin accessibility and has a mild effect on offspring metabolic health. Heliyon 2024; 10:e34043. [PMID: 39100496 PMCID: PMC11296027 DOI: 10.1016/j.heliyon.2024.e34043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 06/28/2024] [Accepted: 07/02/2024] [Indexed: 08/06/2024] Open
Abstract
The increasing global burden of metabolic disorders including obesity and diabetes necessitates a comprehensive understanding of their etiology, which not only encompasses genetic and environmental factors but also parental influence. Recent evidence has unveiled paternal obesity as a contributing factor to offspring's metabolic health via sperm epigenetic modifications. In this study, we investigated the impact of a Western diet-induced obesity in C57BL/6 male mice on sperm chromatin accessibility and the subsequent metabolic health of their progeny. Utilizing Assay for Transposase-Accessible Chromatin with sequencing, we discovered 450 regions with differential accessibility in sperm from obese fathers, implicating key developmental and metabolic pathways. Contrary to expectations, these epigenetic alterations in sperm were not predictive of long-term metabolic disorders in offspring, who exhibited only mild transient metabolic changes early in life. Both male and female F1 progeny showed no enduring predisposition to obesity or diabetes. These results underscore the biological resilience of offspring to paternal epigenetic inheritance, suggesting a complex interplay between inherited epigenetic modifications and the offspring's own developmental compensatory mechanisms. This study calls for further research into the biological processes that confer this resilience, which could inform interventional strategies to combat the heritability of metabolic diseases.
Collapse
Affiliation(s)
- Iasim Tahiri
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Sergio R. Llana
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Júlia Fos-Domènech
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Maria Milà-Guash
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Miriam Toledo
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Roberta Haddad-Tóvolli
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Marc Claret
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
- School of Medicine, Universitat de Barcelona, Barcelona, Spain
| | - Arnaud Obri
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| |
Collapse
|
93
|
Nicze M, Dec A, Borówka M, Krzyżak D, Bołdys A, Bułdak Ł, Okopień B. Molecular Mechanisms behind Obesity and Their Potential Exploitation in Current and Future Therapy. Int J Mol Sci 2024; 25:8202. [PMID: 39125772 PMCID: PMC11311839 DOI: 10.3390/ijms25158202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/22/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
Obesity is a chronic disease caused primarily by the imbalance between the amount of calories supplied to the body and energy expenditure. Not only does it deteriorate the quality of life, but most importantly it increases the risk of cardiovascular diseases and the development of type 2 diabetes mellitus, leading to reduced life expectancy. In this review, we would like to present the molecular pathomechanisms underlying obesity, which constitute the target points for the action of anti-obesity medications. These include the central nervous system, brain-gut-microbiome axis, gastrointestinal motility, and energy expenditure. A significant part of this article is dedicated to incretin-based drugs such as GLP-1 receptor agonists (e.g., liraglutide and semaglutide), as well as the brand new dual GLP-1 and GIP receptor agonist tirzepatide, all of which have become "block-buster" drugs due to their effectiveness in reducing body weight and beneficial effects on the patient's metabolic profile. Finally, this review article highlights newly designed molecules with the potential for future obesity management that are the subject of ongoing clinical trials.
Collapse
Affiliation(s)
- Michał Nicze
- Department of Internal Medicine and Clinical Pharmacology, Faculty of Medical Sciences, Medical University of Silesia in Katowice, Medyków 18, 40-752 Katowice, Poland (A.B.); (B.O.)
| | | | | | | | | | - Łukasz Bułdak
- Department of Internal Medicine and Clinical Pharmacology, Faculty of Medical Sciences, Medical University of Silesia in Katowice, Medyków 18, 40-752 Katowice, Poland (A.B.); (B.O.)
| | | |
Collapse
|
94
|
Hao J, Jin R, Yi Y, Jiang X, Yu J, Xu Z, Schnicker NJ, Chimenti MS, Sugg SL, Li B. Development of a humanized anti-FABP4 monoclonal antibody for potential treatment of breast cancer. Breast Cancer Res 2024; 26:119. [PMID: 39054536 PMCID: PMC11270797 DOI: 10.1186/s13058-024-01873-y] [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: 05/09/2024] [Accepted: 07/18/2024] [Indexed: 07/27/2024] Open
Abstract
BACKGROUND Breast cancer is the most common cancer in women diagnosed in the U.S. and worldwide. Obesity increases breast cancer risk without clear underlying molecular mechanisms. Our studies demonstrate that circulating adipose fatty acid binding protein (A-FABP, or FABP4) links obesity-induced dysregulated lipid metabolism and breast cancer risk, thus potentially offering a new target for breast cancer treatment. METHODS We immunized FABP4 knockout mice with recombinant human FABP4 and screened hybridoma clones with specific binding to FABP4. The potential effects of antibodies on breast cancer cells in vitro were evaluated using migration, invasion, and limiting dilution assays. Tumor progression in vivo was evaluated in various types of tumorigenesis models including C57BL/6 mice, Balb/c mice, and SCID mice. The phenotype and function of immune cells in tumor microenvironment were characterized with multi-color flow cytometry. Tumor stemness was detected by ALDH assays. To characterize antigen-antibody binding capacity, we determined the dissociation constant of selected anti-FABP4 antibodies via surface plasmon resonance. Further analyses in tumor tissue were performed using 10X Genomics Visium spatial single cell technology. RESULTS Herein, we report the generation of humanized monoclonal antibodies blocking FABP4 activity for breast cancer treatment in mouse models. One clone, named 12G2, which significantly reduced circulating levels of FABP4 and inhibited mammary tumor growth, was selected for further characterization. After confirming the therapeutic efficacy of the chimeric 12G2 monoclonal antibody consisting of mouse variable regions and human IgG1 constant regions, 16 humanized 12G2 monoclonal antibody variants were generated by grafting its complementary determining regions to selected human germline sequences. Humanized V9 monoclonal antibody showed consistent results in inhibiting mammary tumor growth and metastasis by affecting tumor cell mitochondrial metabolism. CONCLUSIONS Our current evidence suggests that targeting FABP4 with humanized monoclonal antibodies may represent a novel strategy for the treatment of breast cancer and possibly other obesity- associated diseases.
Collapse
Affiliation(s)
- Jiaqing Hao
- Department of Pathology, University of Iowa, 431 Newton Road, Iowa City, IA, 52242, USA
| | - Rong Jin
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yanmei Yi
- School of Basic Medical Sciences, Guangdong Medical University, Zhanjiang, China
| | - Xingshan Jiang
- Department of Pathology, University of Iowa, 431 Newton Road, Iowa City, IA, 52242, USA
| | - Jianyu Yu
- Department of Pathology, University of Iowa, 431 Newton Road, Iowa City, IA, 52242, USA
| | - Zhen Xu
- Protein and Crystallography Facility, University of Iowa, Iowa City, IA, USA
| | | | - Michael S Chimenti
- Iowa Institute of Human Genetics, University of Iowa, Iowa City, IA, USA
| | - Sonia L Sugg
- Department of Surgery, University of Iowa, Iowa City, IA, USA
| | - Bing Li
- Department of Pathology, University of Iowa, 431 Newton Road, Iowa City, IA, 52242, USA.
| |
Collapse
|
95
|
Abu-Rub LI, Al-Barazenji T, Abiib S, Hammad AS, Abbas A, Hussain K, Al-Shafai M. Identification of KSR2 Variants in Pediatric Patients with Severe Early-Onset Obesity from Qatar. Genes (Basel) 2024; 15:966. [PMID: 39202327 PMCID: PMC11353872 DOI: 10.3390/genes15080966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 07/14/2024] [Accepted: 07/18/2024] [Indexed: 09/03/2024] Open
Abstract
The kinase suppressor of Ras 2 (KSR2) gene is associated with monogenic obesity, and loss-of-function variants in KSR2 have been identified in individuals with severe early-onset obesity. This study investigated KSR2 variants in 9 pediatric patients with severe early-onset obesity in Qatar using whole genome sequencing among a cohort of 240 individuals. We focused on KSR2 variants with a minor allele frequency (MAF) below 1% and a Combined Annotation Dependent Depletion (CADD) score above 13 to identify potential causative variants. Our analysis identified four KSR2 variants: one intronic (c.1765-8G>A) and three missense variants (c.1057G>A, c.1673G>A, and c.923T>C) in nine patients. The intronic variant c.1765-8G>A was the most frequent (seen in six individuals) and had a CADD score of 21.10, suggesting possible pathogenicity. This variant showed a significantly higher allele frequency in the Qatari population compared to the Genome Aggregation Database (gnomAD), indicating a possible founder effect. Molecular modeling of the missense variants revealed structural changes in the protein structure. The study concludes that these four KSR2 variants are associated with monogenic obesity, with an autosomal dominant inheritance pattern. The c.1765-8G>A variant's prevalence in Qatar underscores its importance in genetic screening for severe obesity. This research advances the understanding of genetic factors in severe early-onset obesity and may inform better management strategies.
Collapse
Affiliation(s)
- Lubna I. Abu-Rub
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha 2713, Qatar; (L.I.A.-R.); (T.A.-B.); (S.A.); (A.S.H.); (A.A.)
| | - Tara Al-Barazenji
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha 2713, Qatar; (L.I.A.-R.); (T.A.-B.); (S.A.); (A.S.H.); (A.A.)
| | - Sumaya Abiib
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha 2713, Qatar; (L.I.A.-R.); (T.A.-B.); (S.A.); (A.S.H.); (A.A.)
| | - Ayat S Hammad
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha 2713, Qatar; (L.I.A.-R.); (T.A.-B.); (S.A.); (A.S.H.); (A.A.)
| | - Alaa Abbas
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha 2713, Qatar; (L.I.A.-R.); (T.A.-B.); (S.A.); (A.S.H.); (A.A.)
| | - Khalid Hussain
- Division of Endocrinology, Department of Pediatric Medicine, Sidra Medicine, Doha P.O. Box 26999, Qatar
| | - Mashael Al-Shafai
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha 2713, Qatar; (L.I.A.-R.); (T.A.-B.); (S.A.); (A.S.H.); (A.A.)
- Biomedical Research Center, Qatar University, Doha P.O. Box 2713, Qatar
| |
Collapse
|
96
|
Liang Y, Kaushal D, Wilson RB. Cellular Senescence and Extracellular Vesicles in the Pathogenesis and Treatment of Obesity-A Narrative Review. Int J Mol Sci 2024; 25:7943. [PMID: 39063184 PMCID: PMC11276987 DOI: 10.3390/ijms25147943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/04/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
This narrative review explores the pathophysiology of obesity, cellular senescence, and exosome release. When exposed to excessive nutrients, adipocytes develop mitochondrial dysfunction and generate reactive oxygen species with DNA damage. This triggers adipocyte hypertrophy and hypoxia, inhibition of adiponectin secretion and adipogenesis, increased endoplasmic reticulum stress and maladaptive unfolded protein response, metaflammation, and polarization of macrophages. Such feed-forward cycles are not resolved by antioxidant systems, heat shock response pathways, or DNA repair mechanisms, resulting in transmissible cellular senescence via autocrine, paracrine, and endocrine signaling. Senescence can thus affect preadipocytes, mature adipocytes, tissue macrophages and lymphocytes, hepatocytes, vascular endothelium, pancreatic β cells, myocytes, hypothalamic nuclei, and renal podocytes. The senescence-associated secretory phenotype is closely related to visceral adipose tissue expansion and metaflammation; inhibition of SIRT-1, adiponectin, and autophagy; and increased release of exosomes, exosomal micro-RNAs, pro-inflammatory adipokines, and saturated free fatty acids. The resulting hypernefemia, insulin resistance, and diminished fatty acid β-oxidation lead to lipotoxicity and progressive obesity, metabolic syndrome, and physical and cognitive functional decline. Weight cycling is related to continuing immunosenescence and exposure to palmitate. Cellular senescence, exosome release, and the transmissible senescence-associated secretory phenotype contribute to obesity and metabolic syndrome. Targeted therapies have interrelated and synergistic effects on cellular senescence, obesity, and premature aging.
Collapse
Affiliation(s)
- Yicong Liang
- Bankstown Hospital, University of New South Wales, Sydney, NSW 2560, Australia;
| | - Devesh Kaushal
- Campbelltown Hospital, Western Sydney University, Sydney, NSW 2560, Australia;
| | - Robert Beaumont Wilson
- School of Clinical Medicine, University of New South Wales, High St., Kensington, Sydney, NSW 2052, Australia
| |
Collapse
|
97
|
Yang K, Yu X, Guo Z, Fang Z, Zhang H, Zhang W, Liu C, Ji Y, Dong Z, Gu Q, Yao J, Liu C. PIM1 alleviated liver oxidative stress and NAFLD by regulating the NRF2/HO-1/NQO1 pathway. Life Sci 2024; 349:122714. [PMID: 38735366 DOI: 10.1016/j.lfs.2024.122714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/06/2024] [Accepted: 05/09/2024] [Indexed: 05/14/2024]
Abstract
AIMS Non-alcoholic fatty liver disease (NAFLD) has risen as a significant global public health issue, for which vertical sleeve gastrectomy (VSG) has become an effective treatment method. The study sought to elucidate the processes through which PIM1 mitigates the advancement of NAFLD. The Pro-viral integration site for Moloney murine leukemia virus 1 (PIM1) functions as a serine/threonine kinase. Bioinformatics analysis revealed that reduced PIM1 expression in NAFLD. METHODS To further prove the role of PIM1 in NAFLD, an in-depth in vivo experiment was performed, in which male C57BL/6 mice were randomly grouped to receive a normal or high-fat diet for 24 weeks. They were operated or delivered the loaded adeno-associated virus which the PIM1 was overexpressed (AAV-PIM1). In an in vitro experiment, AML12 cells were treated with palmitic acid to induce hepatic steatosis. KEY FINDINGS The results revealed that the VSG surgery and virus delivery of mice alleviated oxidative stress, and apoptosis in vivo. For AML12 cells, the levels of oxidative stress, apoptosis, and lipid metabolism were reduced via PIM1 upregulation. Moreover, ML385 treatment resulted in the downregulation of the NRF2/HO-1/NQO1 signaling cascade, indicating that PIM1 mitigates NAFLD by targeting this pathway. SIGNIFICANCE PIM1 alleviated mice liver oxidative stress and NAFLD induced by high-fat diet by regulating the NRF2/HO-1/NQO1 signaling Pathway.
Collapse
Affiliation(s)
- Kai Yang
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaoxiao Yu
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zihao Guo
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhihao Fang
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hongyu Zhang
- Department of Neurosurgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wanyangchuan Zhang
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Changxu Liu
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yanchao Ji
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhichao Dong
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qiang Gu
- Department of Neurosurgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiahao Yao
- Department of Neurosurgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chang Liu
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
98
|
Venkatesh SS, Ganjgahi H, Palmer DS, Coley K, Linchangco GV, Hui Q, Wilson P, Ho YL, Cho K, Arumäe K, Wittemans LBL, Nellåker C, Vainik U, Sun YV, Holmes C, Lindgren CM, Nicholson G. Characterising the genetic architecture of changes in adiposity during adulthood using electronic health records. Nat Commun 2024; 15:5801. [PMID: 38987242 PMCID: PMC11237142 DOI: 10.1038/s41467-024-49998-0] [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: 01/25/2023] [Accepted: 06/25/2024] [Indexed: 07/12/2024] Open
Abstract
Obesity is a heritable disease, characterised by excess adiposity that is measured by body mass index (BMI). While over 1,000 genetic loci are associated with BMI, less is known about the genetic contribution to adiposity trajectories over adulthood. We derive adiposity-change phenotypes from 24.5 million primary-care health records in over 740,000 individuals in the UK Biobank, Million Veteran Program USA, and Estonian Biobank, to discover and validate the genetic architecture of adiposity trajectories. Using multiple BMI measurements over time increases power to identify genetic factors affecting baseline BMI by 14%. In the largest reported genome-wide study of adiposity-change in adulthood, we identify novel associations with BMI-change at six independent loci, including rs429358 (APOE missense variant). The SNP-based heritability of BMI-change (1.98%) is 9-fold lower than that of BMI. The modest genetic correlation between BMI-change and BMI (45.2%) indicates that genetic studies of longitudinal trajectories could uncover novel biology of quantitative traits in adulthood.
Collapse
Affiliation(s)
- Samvida S Venkatesh
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK.
| | - Habib Ganjgahi
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
- Department of Statistics, University of Oxford, Oxford, UK
| | - Duncan S Palmer
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
- Nuffield Department of Population Health, Medical Sciences Division, University of Oxford, Oxford, UK
| | - Kayesha Coley
- Department of Population Health Sciences, University of Leicester, Leicester, UK
| | - Gregorio V Linchangco
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA, USA
- Atlanta VA Health Care System, Decatur, GA, USA
| | - Qin Hui
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA, USA
- Atlanta VA Health Care System, Decatur, GA, USA
| | - Peter Wilson
- Atlanta VA Health Care System, Decatur, GA, USA
- Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Yuk-Lam Ho
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), Veterans Affairs Boston Healthcare System, Boston, MA, USA
| | - Kelly Cho
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), Veterans Affairs Boston Healthcare System, Boston, MA, USA
- Division of Aging, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kadri Arumäe
- Institute of Psychology, Faculty of Social Sciences, University of Tartu, Tartu, Estonia
| | - Laura B L Wittemans
- Novo Nordisk Research Centre Oxford, Oxford, UK
- Nuffield Department of Women's and Reproductive Health, Medical Sciences Division, University of Oxford, Oxford, UK
| | - Christoffer Nellåker
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
- Nuffield Department of Women's and Reproductive Health, Medical Sciences Division, University of Oxford, Oxford, UK
| | - Uku Vainik
- Institute of Psychology, Faculty of Social Sciences, University of Tartu, Tartu, Estonia
- Estonian Genome Centre, Institute of Genomics, Faculty of Science and Technology, University of Tartu, Tartu, Estonia
- Department of Neurology and Neurosurgery, Faculty of Medicine and Health Sciences, University of McGill, Montreal, Canada
| | - Yan V Sun
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA, USA
- Atlanta VA Health Care System, Decatur, GA, USA
| | - Chris Holmes
- Department of Statistics, University of Oxford, Oxford, UK
- Nuffield Department of Medicine, Medical Sciences Division, University of Oxford, Oxford, UK
- The Alan Turing Institute, London, UK
| | - Cecilia M Lindgren
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK.
- Nuffield Department of Women's and Reproductive Health, Medical Sciences Division, University of Oxford, Oxford, UK.
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| | | |
Collapse
|
99
|
Sivakumar S, Lama D, Rabhi N. Childhood obesity from the genes to the epigenome. Front Endocrinol (Lausanne) 2024; 15:1393250. [PMID: 39045266 PMCID: PMC11263020 DOI: 10.3389/fendo.2024.1393250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 06/25/2024] [Indexed: 07/25/2024] Open
Abstract
The prevalence of obesity and its associated comorbidities has surged dramatically in recent decades. Especially concerning is the increased rate of childhood obesity, resulting in diseases traditionally associated only with adulthood. While obesity fundamentally arises from energy imbalance, emerging evidence over the past decade has revealed the involvement of additional factors. Epidemiological and murine studies have provided extensive evidence linking parental obesity to increased offspring weight and subsequent cardiometabolic complications in adulthood. Offspring exposed to an obese environment during conception, pregnancy, and/or lactation often exhibit increased body weight and long-term metabolic health issues, suggesting a transgenerational inheritance of disease susceptibility through epigenetic mechanisms rather than solely classic genetic mutations. In this review, we explore the current understanding of the mechanisms mediating transgenerational and intergenerational transmission of obesity. We delve into recent findings regarding both paternal and maternal obesity, shedding light on the underlying mechanisms and potential sex differences in offspring outcomes. A deeper understanding of the mechanisms behind obesity inheritance holds promise for enhancing clinical management strategies in offspring and breaking the cycle of increased metabolic risk across generations.
Collapse
Affiliation(s)
| | | | - Nabil Rabhi
- Department of Biochemistry and Cell Biology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States
| |
Collapse
|
100
|
Onay T, Beyazıt U, Uçar A, Bütün Ayhan A. Obesity in childhood: associations with parental neglect, nutritional habits, and obesity awareness. Front Nutr 2024; 11:1430418. [PMID: 39015536 PMCID: PMC11250508 DOI: 10.3389/fnut.2024.1430418] [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: 05/09/2024] [Accepted: 06/17/2024] [Indexed: 07/18/2024] Open
Abstract
Background The relationships underlying the dynamic between obesity and parental neglect in terms of nutritional habits and obesity awareness are unclear. Parental neglect remains a significant subject of concern that needs to be examined in the context of obesity. Methods The aim was to examine the relationships between childhood obesity, parental neglect, children's eating habits and obesity. The study group consisted of 404 children and their parents from Ankara, Turkiye. As data collection tools, an Individual Information Form, Obesity Awareness Scale, the Parents Form of the Multidimensional Neglectful Behaviors Scale were administered. In addition, information on the children's body mass indexes was obtained by anthropometric measurements and the findings were recorded on the questionnaires of each child. Results It was found that 98 (24.3%) of the children included in the study were overweight and 63 (15.6%) were obese. The results of the multinomial logistic regression analysis indicated that in the underweight and overweight group, the parents' perception of their child's weight predicted body mass index in children, and in the obese group, along with the parents' perception of their child's weight, the age and gender of the child, eating fast, obesity in the family and parental neglect were also predictors. Conclusion Practitioners such as nurses, dietitians and child developmentalists working in schools should consider weight problems in children as one of the indicators of parental neglect and should implement interventive efforts to enhance parental supervision of children at risk.
Collapse
Affiliation(s)
- Tuba Onay
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Bandirma Onyedi Eylul University, Balıkesir, Türkiye
| | - Utku Beyazıt
- Child Development Department, Kumluca Health Sciences Faculty, Akdeniz University, Antalya, Türkiye
| | - Aslı Uçar
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Ankara University, Ankara, Türkiye
| | - Aynur Bütün Ayhan
- Child Development Department, Faculty of Health Sciences, Ankara University, Ankara, Türkiye
| |
Collapse
|