1
|
Suh D, Lee SM. The Impact of Obesity on Readmission and Healthcare Costs in Patients with Skin and Subcutaneous Tissue Infections. Risk Manag Healthc Policy 2025; 18:1579-1590. [PMID: 40386074 PMCID: PMC12085134 DOI: 10.2147/rmhp.s516684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 05/08/2025] [Indexed: 05/20/2025] Open
Abstract
Purpose Obesity is a global public health issue linked to worsened skin and subcutaneous tissue infections (SSTIs), complicating clinical management and increasing healthcare costs. This study aimed to evaluate obesity's influence on hospitalization duration, readmission rates, and healthcare costs among patients with SSTIs, with an emphasis on sex-specific patterns. Patients and Methods This retrospective cohort study analyzed data from South Korea's national healthcare database. The study population comprised adults hospitalized with SSTIs between 2015 and 2020. Obesity measures included body mass index (BMI) and waist circumference (WC), categorized by standard thresholds. Statistical analyses included Cox proportional hazards models for hospitalization duration, while multivariable logistic regression evaluated readmission risks. Healthcare costs were analyzed using generalized linear models, with sex-stratified analysis to examine clinical and economic outcome disparities. Results Male patients demonstrated an inverse relationship between BMI and hospitalization duration and costs, with minimal WC influence. Conversely, female patients exhibited positive associations between both obesity measures and hospitalization outcomes. SSTI-related readmissions within two years increased with rising BMI and WC across both sexes (p < 0.001). Estimated readmission costs showed significant sex-specific variations, increasing 55% among males with WC ≥ 100 cm versus < 80 cm and 132% among females with WC ≥ 95 cm versus < 75 cm. Conclusion Obesity substantially impacts SSTI clinical severity and economic costs, with distinct sex-specific disparities. Implementing tailored antimicrobial regimens, weight management strategies, and sex-specific treatment protocols is essential for outcome optimization and cost reduction. Future research should prioritize sex-specific interventions and resource allocation strategies in SSTI management.
Collapse
Affiliation(s)
- David Suh
- School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Seung-Mi Lee
- College of Pharmacy, Daegu Catholic University, Gyeongsan, Republic of Korea
| |
Collapse
|
2
|
Roberts TD, Hutchinson DS, Wootten D, De Blasio MJ, Ritchie RH. Advances in incretin therapies for targeting cardiovascular disease in diabetes. J Mol Cell Cardiol 2025; 202:102-115. [PMID: 40086589 DOI: 10.1016/j.yjmcc.2025.03.007] [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: 11/01/2024] [Revised: 02/12/2025] [Accepted: 03/11/2025] [Indexed: 03/16/2025]
Abstract
The global prevalence of obesity is skyrocketing at an alarming rate, with recent data estimating that one-in-eight people are now living with the disease. Obesity is a chronic metabolic disorder that shares underlying pathophysiology with other metabolically-linked diseases such as type 2 diabetes mellitus, cardiovascular disease and diabetic cardiomyopathy. There is a distinct correlation between type 2 diabetes status and the likelihood of heart failure. Of note, there is an apparent sexual dimorphism, with women disproportionately affected with respect to the degree of severity of the cardiac phenotype of diabetic cardiomyopathy that results from diabetes. The current pharmacotherapies available for the attenuation of hyperglycaemia in type 2 diabetes are not always effective, and have varying degrees of efficacy in the setting of heart failure. Insulin can worsen heart failure prognosis whereas metformin, sodium-glucose cotransporter 2 inhibitors (SGLT2i) and more recently, glucagon-like peptide-1 receptor agonists (GLP-1RAs), have demonstrated cardioprotection with their administration. This review will highlight the advancement of incretin therapies for individuals with diabetes and heart failure and explore newly-reported evidence of the clinical usefulness of GLP-1R agonists in this distinct phenotype of heart failure.
Collapse
Affiliation(s)
- Timothy D Roberts
- Heart Failure Pharmacology Laboratory, Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, VIC, Australia
| | - Dana S Hutchinson
- Metabolic G Protein-Coupled Receptor Laboratory, Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, VIC, Australia
| | - Denise Wootten
- Metabolic G Protein-Coupled Receptor Laboratory, Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, VIC, Australia; ARC Centre for Cryo-Electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, Victoria, Australia
| | - Miles J De Blasio
- Heart Failure Pharmacology Laboratory, Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, VIC, Australia.
| | - Rebecca H Ritchie
- Heart Failure Pharmacology Laboratory, Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, VIC, Australia.
| |
Collapse
|
3
|
Santos GL, Dias Costa EF, Dalla Costa AP, Zanesco AM, Simoes MR, Rogério F, Demolin DMR, Navarro CDC, Velloso LA, Francisco A, Castilho RF. Influence of Mitochondrial NAD(P) + Transhydrogenase (NNT) on Hypothalamic Inflammation and Metabolic Dysfunction Induced by a High-Fat Diet in Mice. Horm Metab Res 2025; 57:199-207. [PMID: 39481390 DOI: 10.1055/a-2420-6549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/02/2024]
Abstract
The mitochondrial protein NAD(P)+ transhydrogenase (NNT) has been implicated in the metabolic derangements observed in obesity. Mice with the C57BL/6J genetic background bear a spontaneous mutation in the Nnt gene and are known to exhibit increased susceptibility to diet-induced metabolic disorders. Most of the studies on NNT in the context of diet-induced obesity have compared C57BL/6J mice with other mouse strains, where differences in genetic background can serve as confounding factors. Moreover, these studies have predominantly employed a high-fat diet (HFD) consisting of approximately 60% of calories from fat, which may not accurately mimic real-world fat-rich diets. In this study, we sought to examine the role of NNT in diet-induced hypothalamic inflammation and metabolic syndrome by using a congenic mice model lacking NNT, along with a HFD providing approximately 45% of calories from fat. Our findings indicate that mice lacking NNT were more protected from HFD-induced weight gain but presented a worse performance on glucose tolerance test, albeit not in insulin tolerance test. Interestingly, the brown adipose tissue of HFD-fed Nnt +/+ mice presented a greater mass and a higher whole-tissue ex-vivo oxygen consumption rate. Also, HFD increased the expression of the inflammatory markers Il1β, Tlr4 and Iba1 in the hypothalamus of Nnt -/- mice. In conclusion, our study highlights the importance of NNT in the context of diet-induced obesity and metabolic syndrome, indicating its contribution to mitigate hypothalamic inflammation and suggesting its role in the brown adipose tissue increased mass.
Collapse
Affiliation(s)
| | | | | | - Ariane Maria Zanesco
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, State University of Campinas (UNICAMP), Campinas, Brazil
| | - Marcela Reymond Simoes
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, State University of Campinas (UNICAMP), Campinas, Brazil
| | - Fábio Rogério
- Department of Pathology, State University of Campinas (UNICAMP), Campinas, Brazil
| | - Daniele Masselli Rodrigues Demolin
- Multidisciplinary Center for Biological Investigation on Laboratory Animals Science, State University of Campinas (UNICAMP), Campinas, Brazil
| | | | - Lício Augusto Velloso
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, State University of Campinas (UNICAMP), Campinas, Brazil
| | - Annelise Francisco
- Department of Experimental Medical Science, Lunds Universitet, Lund, Sweden
- Department of Pathology, State University of Campinas (UNICAMP), Campinas, Brazil
| | | |
Collapse
|
4
|
Fang Q, Ye L, Han L, Yao S, Cheng Q, Wei X, Zhang Y, Huang J, Ning G, Wang J, Zhang Y, Zhang Z. LGR4 is a key regulator of hepatic gluconeogenesis. Free Radic Biol Med 2025; 229:183-194. [PMID: 39826817 DOI: 10.1016/j.freeradbiomed.2025.01.025] [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: 11/26/2024] [Revised: 01/08/2025] [Accepted: 01/13/2025] [Indexed: 01/22/2025]
Abstract
AIMS/HYPOTHESIS Emerging evidence underscored the significance of leucine-rich repeat-containing G protein-coupled receptor (LGR) 4 in endocrine and metabolic disorders. Despite this, its role in LGR4 in hepatic glucose metabolism remains poorly understood. In this study we set out to test whether LGR4 regulates glucose production in liver through a specific signaling pathway. METHODS Hepatic glucose production and gluconeogenic gene expressions were detected after silence of LGR4 in three obese mice models. Then, whole-body LGR4-deficient (LGR4 KO) mice, liver-specific LGR4 knockout (LGR4LKO) mice, and liver-specific LGR4 overexpression (LGR4LOV) mice were generated, in which we analyzed the effects of LGR4 on hepatic glucose metabolism upon HFD feeding, among which live imaging and quantitative analysis of hepatic phosphoenolpyruvate carboxykinase (PEPCK)-luciferase activity were conducted. RESULTS LGR4 expression was significantly upregulated in the liver of three obese mouse models, and presented dynamic expression patterns in response to nutritional fluxes. We utilized global and liver-specific LGR4 knockouts (LGR4LKO), along with adenoviral-mediated LGR4 knockdown in mice, to show improved glucose tolerance and decreased hepatic gluconeogenesis. Specifically, the expression of rate-limiting gluconeogenic enzymes, PEPCK was significantly downregulated. Conversely, mouse model with adenovirus-mediated LGR4 overexpression (LGR4LOV) exhibited elevated gluconeogenesis and PEPCK expression and reversed the suppression observed in LGR4 knockout models. Notably, neither RANKL nor PKA signaling pathways, which were reported to take part in LGR4's function, were involved in the process of LGR4 regulating PEPCK. Instead, TopFlash reporter system and inhibitors application suggested that LGR4's influence on hepatic gluconeogenesis operates through the canonical Wnt/β-catenin/TCF7L2 signaling pathway. CONCLUSIONS/INTERPRETATION Overall, these findings underscore a novel mechanism by which LGR4 regulates hepatic gluconeogenesis, presenting a potential therapeutic target for diabetes management.
Collapse
Affiliation(s)
- Qianhua Fang
- 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, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Linmin Ye
- 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, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Luyu Han
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Shuangshuang Yao
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qianyun Cheng
- 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, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xing Wei
- 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, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan 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, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Juelin Huang
- 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, 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, 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, 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, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Zhiguo 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, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
5
|
Gans IM, Chepurko E, Chepurko V, Mangoba GA, Asson-Batres MA, Vary CP, Sawyer DB. Vitamin A deficiency and male-specific effects on heart function in mice. Biochem Biophys Res Commun 2025; 748:151300. [PMID: 39818189 PMCID: PMC11801225 DOI: 10.1016/j.bbrc.2025.151300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 12/30/2024] [Accepted: 01/08/2025] [Indexed: 01/18/2025]
Abstract
Hepatic stores of Vitamin A (retinol) are mobilized and metabolized in the heart following myocardial infarction. The physiological consequences of this mobilization are poorly understood. Here we used dietary depletion in a lecithin retinol acyltransferase mutant mouse line to induce Vitamin A deficiency and investigate the effects on cardiac function and recovery from myocardial infarction. We found that uninjured Vitamin A-depleted hearts had decreased contractile function but, paradoxically, improved recovery after injury. These effects on cardiac function were specific to male mice, which experienced more rapid and severe depletion of circulating Vitamin A. Following injury, Vitamin A deficiency also caused hepatic hypolipidemia and gene expression changes in heart and liver suggesting altered metabolism contributed to cardiac phenotypes.
Collapse
Affiliation(s)
- Ian M Gans
- Center for Molecular Medicine, Maine Health Institute for Research, 81 Research Drive, Scarborough, ME, USA.
| | - Elena Chepurko
- Center for Molecular Medicine, Maine Health Institute for Research, 81 Research Drive, Scarborough, ME, USA
| | - Vadim Chepurko
- Center for Molecular Medicine, Maine Health Institute for Research, 81 Research Drive, Scarborough, ME, USA
| | - Grazina A Mangoba
- Center for Molecular Medicine, Maine Health Institute for Research, 81 Research Drive, Scarborough, ME, USA
| | - Mary Ann Asson-Batres
- Center for Molecular Medicine, Maine Health Institute for Research, 81 Research Drive, Scarborough, ME, USA; Tennessee State University, Nashville, TN, USA
| | - Calvin P Vary
- Center for Molecular Medicine, Maine Health Institute for Research, 81 Research Drive, Scarborough, ME, USA; Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, USA
| | - Douglas B Sawyer
- Center for Molecular Medicine, Maine Health Institute for Research, 81 Research Drive, Scarborough, ME, USA
| |
Collapse
|
6
|
Song M, Bai Y, Song F. High-fat diet and neuroinflammation: The role of mitochondria. Pharmacol Res 2025; 212:107615. [PMID: 39842474 DOI: 10.1016/j.phrs.2025.107615] [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: 08/15/2024] [Revised: 12/28/2024] [Accepted: 01/17/2025] [Indexed: 01/24/2025]
Abstract
In recent years, increasing evidence has supported that high-fat diet (HFD) can induce the chronic, low-grade neuroinflammation in the brain, which is closely associated with the impairment of cognitive function. As the key organelles responsible for energy metabolism in the cell, mitochondria are believed to involved in the pathogenesis of a variety of neurological disorders. This review summarizes the current progress in the field of the relationship between HFD exposure and neurodegenerative diseases, and outline the major routines of HFD induced neuroinflammation and its pathological significance in the pathogenesis of neurodegenerative diseases. Furthermore, the article highlights the pivotal role of mitochondrial dysfunction in driving the neuroinflammation in the setting of HFD. Danger-associated molecular patterns (DAMPs) from damaged mitochondria can activate innate immune signaling pathways, while mitochondrial dysfunction itself can lead to metabolic remodeling of inflammatory cells, thus inducing neuroinflammation. More importantly, mitochondrial damage, neuroinflammation, and insulin resistance caused by HFD form a mutually reinforcing vicious cycle, ultimately leading to the death of neurons and promoting the progression of neurodegenerative diseases. Thus, in-depth elucidation of the role and underlying mechanisms of mitochondrial dysfunction in HFD-induced metabolic disorders may not only expand our understanding of the mechanistic linkages between HFD and etiology of neurodegenerative diseases, but also help develop the specific strategies for the prevention and treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Mingxue Song
- Department of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Road, Jinan, Shandong 250012, China.
| | - Yao Bai
- NHC Key Laboratory of Food Safety Risk Assessment, China National Center for Food Safety Risk Assessment, Beijing 100021, China.
| | - Fuyong Song
- Department of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Road, Jinan, Shandong 250012, China.
| |
Collapse
|
7
|
Dolatshahi M, Commean PK, Rahmani F, Xu Y, Liu J, Hosseinzadeh Kassani S, Naghashzadeh M, Lloyd L, Nguyen C, McBee Kemper A, Hantler N, Ly M, Yu G, Flores S, Ippolito JE, Song SK, Sirlin CB, Dai W, Mittendorfer B, Morris JC, Benzinger TLS, Raji CA. Relationships between abdominal adipose tissue and neuroinflammation with diffusion basis spectrum imaging in midlife obesity. Obesity (Silver Spring) 2025; 33:41-53. [PMID: 39517107 DOI: 10.1002/oby.24188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 11/16/2024]
Abstract
OBJECTIVE This study investigated how obesity, BMI ≥ 30 kg/m2, abdominal adiposity, and systemic inflammation relate to neuroinflammation using diffusion basis spectrum imaging. METHODS We analyzed data from 98 cognitively normal midlife participants (mean age: 49.4 [SD 6.2] years; 34 males [34.7%]; 56 with obesity [57.1%]). Participants underwent brain and abdominal magnetic resonance imaging (MRI), blood tests, and amyloid positron emission tomography (PET) imaging. Abdominal visceral and subcutaneous adipose tissue (VAT and SAT, respectively) was segmented, and Centiloids were calculated. Diffusion basis spectrum imaging parameter maps were created using an in-house script, and tract-based spatial statistics assessed white matter differences in high versus low BMI values, VAT, SAT, insulin resistance, systemic inflammation, and Centiloids, with age and sex as covariates. RESULTS Obesity, high VAT, and high SAT were linked to lower axial diffusivity, reduced fiber fraction, and increased restricted fraction in white matter. Obesity was additionally associated with higher hindered fraction and lower fractional anisotropy. Also, individuals with high C-reactive protein showed lower axial diffusivity. Higher restricted fraction correlated with continuous BMI and SAT particularly in male individuals, whereas VAT effects were similar in male and female individuals. CONCLUSIONS The findings suggest that, at midlife, obesity and abdominal fat are associated with reduced brain axonal density and increased inflammation, with visceral fat playing a significant role in both sexes.
Collapse
Affiliation(s)
- Mahsa Dolatshahi
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Paul K Commean
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Farzaneh Rahmani
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Yifei Xu
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jingxia Liu
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | - Mahshid Naghashzadeh
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - LaKisha Lloyd
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Caitlyn Nguyen
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Abby McBee Kemper
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Nancy Hantler
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Maria Ly
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Gary Yu
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Shaney Flores
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Joseph E Ippolito
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Sheng-Kwei Song
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Claude B Sirlin
- Liver Imaging Group, Department of Radiology, University of California, Los Angeles, California, USA
| | - Weiying Dai
- Department of Computer Science, State University of New York at Binghamton, Binghamton, New York, USA
| | - Bettina Mittendorfer
- Departments of Medicine and Nutrition & Exercise Physiology, University of Missouri School of Medicine, Columbia, Missouri, USA
| | - John C Morris
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Tammie L S Benzinger
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Cyrus A Raji
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri, USA
| |
Collapse
|
8
|
Schuetz LT, Duran G, Baeten P, Lintsen D, Hermans D, Chenine S, Verreycken J, Vanmierlo T, Wouters K, Broux B. Sex differentially affects pro-inflammatory cell subsets in adipose tissue depots in a diet induced obesity model. Biol Sex Differ 2024; 15:105. [PMID: 39696610 DOI: 10.1186/s13293-024-00677-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 11/21/2024] [Indexed: 12/20/2024] Open
Abstract
Obesity is a growing pandemic that increases the risk for cardiovascular diseases, type 2 diabetes, and particularly in women also the risk of cancer and neurodegenerative disorders such as dementia and multiple sclerosis. Preclinical studies on obesity focus on male mice as they gain bodyweight faster and show a clear pro-inflammatory phenotype. Here, using male and female mice, we induced obesity by feeding a high fat diet (HFD), and compared adipose tissue (AT) inflammation at the same adiposity stage (% AT/bodyweight) between both sexes. Doing so, we identified that female mice show an increase in the number of pro-inflammatory immune cells in the visceral AT at a lower adiposity stage than male mice, but the effect of HFD is diminished with higher adiposity. Interestingly, only female mice showed an increase in immune cells in the subcutaneous AT after HFD feeding. Nonetheless, we found that pro-inflammatory cytokines in blood plasma mirror the inflammatory stage of the visceral AT in both male and female mice. Uniquely in male mice, myeloid cells in the visceral AT showed a higher inflammasome activation upon HFD. In summary, we showed that adiposity differentially affects immune cells in fat depots based on sex.
Collapse
Affiliation(s)
- Lisa T Schuetz
- Neuro-Immune Connections and Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- CARIM-School of Cardiovascular Diseases, Maastricht University, Maastricht, Netherlands
- Internal Medicine, Maastricht University, Maastricht, Netherlands
- University MS Center, Campus Diepenbeek, Diepenbeek, Belgium
- MHeNs-Mental Health and Neuroscience Institute, Maastricht University, Maastricht, Netherlands
| | - Gayel Duran
- Neuro-Immune Connections and Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- University MS Center, Campus Diepenbeek, Diepenbeek, Belgium
| | - Paulien Baeten
- Neuro-Immune Connections and Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- University MS Center, Campus Diepenbeek, Diepenbeek, Belgium
| | - Daphne Lintsen
- Neuro-Immune Connections and Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- University MS Center, Campus Diepenbeek, Diepenbeek, Belgium
| | - Doryssa Hermans
- Neuro-Immune Connections and Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- University MS Center, Campus Diepenbeek, Diepenbeek, Belgium
| | - Sarah Chenine
- Neuro-Immune Connections and Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- University MS Center, Campus Diepenbeek, Diepenbeek, Belgium
- MHeNs-Mental Health and Neuroscience Institute, Maastricht University, Maastricht, Netherlands
| | - Janne Verreycken
- Neuro-Immune Connections and Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- University MS Center, Campus Diepenbeek, Diepenbeek, Belgium
| | - Tim Vanmierlo
- Neuro-Immune Connections and Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- University MS Center, Campus Diepenbeek, Diepenbeek, Belgium
- MHeNs-Mental Health and Neuroscience Institute, Maastricht University, Maastricht, Netherlands
| | - Kristiaan Wouters
- CARIM-School of Cardiovascular Diseases, Maastricht University, Maastricht, Netherlands.
- Internal Medicine, Maastricht University, Maastricht, Netherlands.
| | - Bieke Broux
- Neuro-Immune Connections and Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- University MS Center, Campus Diepenbeek, Diepenbeek, Belgium
| |
Collapse
|
9
|
Lu W, Duan Y, Li K, Cheng Z, Qiu J. Effect of abdominal adipose tissue glucose uptake on brain aging. Alzheimers Dement 2024; 20:7104-7112. [PMID: 39136090 PMCID: PMC11485312 DOI: 10.1002/alz.14193] [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: 02/29/2024] [Revised: 06/27/2024] [Accepted: 07/24/2024] [Indexed: 10/18/2024]
Abstract
INTRODUCTION Abdominal adipose tissue (AT) mass has adverse effects on the brain. This study aimed to investigate the effect of glucose uptake by abdominal AT on brain aging. METHODS Three-hundred twenty-five participants underwent total-body positron emission tomography scan. Brain age was estimated in an independent test set (n = 98) using a support vector regression model that was built using a training set (n = 227). Effects of abdominal subcutaneous and visceral AT (SAT/VAT) glucose uptake on brain age delta were evaluated using linear regression. RESULTS Higher VAT glucose uptake was linked to negative brain age delta across all subgroups. Higher SAT glucose uptake was associated with negative brain age delta in lean individuals. In contrast, increased SAT glucose uptake demonstrated positive trends with brain age delta in female and overweight/obese participants. DISCUSSION Increased glucose uptake of the abdominal VAT has positive influences on the brain, while SAT may not have such influences, except for lean individuals. HIGHLIGHTS Higher glucose uptake of the visceral adipose tissue was linked to decelerated brain aging. Higher glucose uptake of the subcutaneous adipose tissue (SAT) was associated with negative brain age delta in lean individuals. Faster brain aging was associated with increased glucose uptake of the SAT in female and overweight and obese individuals.
Collapse
Affiliation(s)
- Weizhao Lu
- School of RadiologyShandong First Medical University & Shandong Academy of Medical SciencesTaianChina
| | - Yanhua Duan
- Department of Nuclear Medicinethe First Affiliated Hospital of Shandong First Medical University, Shandong Provincial Qianfoshan Hospital Affiliated to Shandong UniversityJinanChina
| | - Kun Li
- Department of Nuclear Medicinethe First Affiliated Hospital of Shandong First Medical University, Shandong Provincial Qianfoshan Hospital Affiliated to Shandong UniversityJinanChina
| | - Zhaoping Cheng
- Department of Nuclear Medicinethe First Affiliated Hospital of Shandong First Medical University, Shandong Provincial Qianfoshan Hospital Affiliated to Shandong UniversityJinanChina
| | - Jianfeng Qiu
- School of RadiologyShandong First Medical University & Shandong Academy of Medical SciencesTaianChina
| |
Collapse
|
10
|
Dolatshahi M, Commean PK, Rahmani F, Liu J, Lloyd L, Nguyen C, Hantler N, Ly M, Yu G, Ippolito JE, Sirlin C, Morris JC, Benzinger TL, Raji CA. Alzheimer Disease Pathology and Neurodegeneration in Midlife Obesity: A Pilot Study. Aging Dis 2024; 15:1843-1854. [PMID: 37548931 PMCID: PMC11272197 DOI: 10.14336/ad.2023.0707] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 07/07/2023] [Indexed: 08/08/2023] Open
Abstract
Obesity and excess adiposity at midlife are risk factors for Alzheimer disease (AD). Visceral fat is known to be associated with insulin resistance and a pro-inflammatory state, the two mechanisms involved in AD pathology. We assessed the association of obesity, MRI-determined abdominal adipose tissue volumes, and insulin resistance with PET-determined amyloid and tau uptake in default mode network areas, and MRI-determined brain volume and cortical thickness in AD cortical signature in the cognitively normal midlife population. Thirty-two middle-aged (age: 51.27±6.12 years, 15 males, body mass index (BMI): 32.28±6.39 kg/m2) cognitively normal participants, underwent bloodwork, brain and abdominal MRI, and amyloid and tau PET scan. Visceral and subcutaneous adipose tissue (VAT, SAT) were semi-automatically segmented using VOXel Analysis Suite (Voxa). FreeSurfer was used to automatically segment brain regions using a probabilistic atlas. PET scans were acquired using [11C]PiB and AV-1451 tracers and were analyzed using PET unified pipeline. The association of brain volumes, cortical thicknesses, and PiB and AV-1451 standardized uptake value ratios (SUVRs) with BMI, VAT/SAT ratio, and insulin resistance were assessed using Spearman's partial correlation. VAT/SAT ratio was associated significantly with PiB SUVRs in the right precuneus cortex (p=0.034) overall, controlling for sex. This association was significant only in males (p=0.044), not females (p=0.166). Higher VAT/SAT ratio and PiB SUVRs in the right precuneus cortex were associated with lower cortical thickness in AD-signature areas predominantly including bilateral temporal cortices, parahippocampal, medial orbitofrontal, and cingulate cortices, with age and sex as covariates. Also, higher BMI and insulin resistance were associated with lower cortical thickness in bilateral temporal poles. In midlife cognitively normal adults, we demonstrated higher amyloid pathology in the right precuneus cortex in individuals with a higher VAT/SAT ratio, a marker of visceral obesity, along with a lower cortical thickness in AD-signature areas associated with higher visceral obesity, insulin resistance, and amyloid pathology.
Collapse
Affiliation(s)
- Mahsa Dolatshahi
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA.
| | - Paul K Commean
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA.
| | - Farzaneh Rahmani
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA.
| | - Jingxia Liu
- Washington University School of Medicine, Division of Public Health Sciences, Department of Surgery, St. Louis, Missouri, USA.
| | - LaKisha Lloyd
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA.
| | - Caitlyn Nguyen
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA.
| | - Nancy Hantler
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA.
| | - Maria Ly
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA.
| | - Gary Yu
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA.
| | - Joseph E Ippolito
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA.
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri, USA.
| | - Claude Sirlin
- Liver Imaging Group, Department of Radiology, University of California, San Diego, La Jolla, California, USA.
| | - John C Morris
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri, USA.
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, Missouri, USA.
| | - Tammie L.S Benzinger
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA.
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, Missouri, USA.
- Department of Neurosurgery, Washington University School of Medicine, St Louis, Missouri, USA.
| | - Cyrus A Raji
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA.
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri, USA.
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, Missouri, USA.
| |
Collapse
|
11
|
Ferrario CR, Münzberg-Gruening H, Rinaman L, Betley JN, Borgland SL, Dus M, Fadool DA, Medler KF, Morton GJ, Sandoval DA, de La Serre CB, Stanley SA, Townsend KL, Watts AG, Maruvada P, Cummings D, Cooke BM. Obesity- and diet-induced plasticity in systems that control eating and energy balance. Obesity (Silver Spring) 2024; 32:1425-1440. [PMID: 39010249 PMCID: PMC11269035 DOI: 10.1002/oby.24060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 07/17/2024]
Abstract
In April 2023, the National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), in partnership with the National Institute of Child Health and Human Development, the National Institute on Aging, and the Office of Behavioral and Social Sciences Research, hosted a 2-day online workshop to discuss neural plasticity in energy homeostasis and obesity. The goal was to provide a broad view of current knowledge while identifying research questions and challenges regarding neural systems that control food intake and energy balance. This review includes highlights from the meeting and is intended both to introduce unfamiliar audiences with concepts central to energy homeostasis, feeding, and obesity and to highlight up-and-coming research in these areas that may be of special interest to those with a background in these fields. The overarching theme of this review addresses plasticity within the central and peripheral nervous systems that regulates and influences eating, emphasizing distinctions between healthy and disease states. This is by no means a comprehensive review because this is a broad and rapidly developing area. However, we have pointed out relevant reviews and primary articles throughout, as well as gaps in current understanding and opportunities for developments in the field.
Collapse
Grants
- P30 DK048520 NIDDK NIH HHS
- NSF1949989 National Science Foundation
- T32 DC000044 NIDCD NIH HHS
- R01 DK089056 NIDDK NIH HHS
- R01 DK124801 NIDDK NIH HHS
- R01 DK100685 NIDDK NIH HHS
- R01 DK130875 NIDDK NIH HHS
- R01 DK133464 NIDDK NIH HHS
- R01 DK125890 NIDDK NIH HHS
- Z99 DK999999 Intramural NIH HHS
- R01 DK124461 NIDDK NIH HHS
- K26 DK138368 NIDDK NIH HHS
- R01 DK121995 NIDDK NIH HHS
- R01 DK121531 NIDDK NIH HHS
- P30 DK089503 NIDDK NIH HHS
- P01 DK119130 NIDDK NIH HHS
- R01 DK118910 NIDDK NIH HHS
- R01 AT011683 NCCIH NIH HHS
- Reported research was supported by DK130246, DK092587, AT011683, MH059911, DK100685, DK119130, DK124801, DK133399, AG079877, DK133464, T32DC000044, F31DC016817, NSF1949989, DK089056, DK124238, DK138368, DK121995, DK125890, DK118910, DK121531, DK124461, DK130875; Canada Research Chair: 950-232211, CIHRFDN148473, CIHRPJT185886; USDA Predoctoral Fellowship; Endowment from the Robinson Family and Tallahassee Memorial Hospital; Department of Defense W81XWH-20-1-0345 and HT9425-23-1-0244; American Diabetes Association #1-17-ACE-31; W.M. Keck Foundation Award; National Science Foundation CAREER 1941822
- R01 DK133399 NIDDK NIH HHS
- HT9425-23-1-0244 Department of Defense
- R01 DK092587 NIDDK NIH HHS
- W81XWH-20-1-0345 Department of Defense
- 1941822 National Science Foundation
- R01 MH059911 NIMH NIH HHS
- F31 DC016817 NIDCD NIH HHS
- R01 AG079877 NIA NIH HHS
- R01 DK130246 NIDDK NIH HHS
- P30 DK017047 NIDDK NIH HHS
- R01 DK124238 NIDDK NIH HHS
Collapse
Affiliation(s)
- Carrie R Ferrario
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan, USA
| | - Heike Münzberg-Gruening
- Laboratory of Central Leptin Signaling, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - Linda Rinaman
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, Florida, USA
| | - J Nicholas Betley
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Stephanie L Borgland
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Monica Dus
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Debra A Fadool
- Department of Biological Science, Program in Neuroscience, Institute of Molecular Biophysics, Florida State University, Tallahassee, Florida, USA
| | - Kathryn F Medler
- School of Animal Sciences, Virginia Tech, Blacksburg, Virginia, USA
| | - Gregory J Morton
- Department of Medicine, University of Washington Medicine Diabetes Institute at South Lake Union, Seattle, Washington, USA
| | - Darleen A Sandoval
- Department of Pediatrics, Section of Nutrition, University of Colorado-Anschutz Medical Campus, Aurora, Colorado, USA
| | - Claire B de La Serre
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Sarah A Stanley
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kristy L Townsend
- Department of Neurological Surgery, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Alan G Watts
- Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California, USA
| | - Padma Maruvada
- National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, USA
| | - Diana Cummings
- National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, USA
| | - Bradley M Cooke
- National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, USA
| |
Collapse
|
12
|
Valentine Y, Nikolajczyk BS. T cells in obesity-associated inflammation: The devil is in the details. Immunol Rev 2024; 324:25-41. [PMID: 38767210 PMCID: PMC11694249 DOI: 10.1111/imr.13354] [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: 05/22/2024]
Abstract
Obesity presents a significant health challenge, affecting 41% of adults and 19.7% of children in the United States. One of the associated health challenges of obesity is chronic low-grade inflammation. In both mice and humans, T cells in circulation and in the adipose tissue play a pivotal role in obesity-associated inflammation. Changes in the numbers and frequency of specific CD4+ Th subsets and their contribution to inflammation through cytokine production indicate declining metabolic health, that is, insulin resistance and T2D. While some Th subset alterations are consistent between mice and humans with obesity, some changes mainly characterize male mice, whereas female mice often resist obesity and inflammation. However, protection from obesity and inflammation is not observed in human females, who can develop obesity-related T-cell inflammation akin to males. The decline in female sex hormones after menopause is also implicated in promoting obesity and inflammation. Age is a second underappreciated factor for defining and regulating obesity-associated inflammation toward translating basic science findings to the clinic. Weight loss in mice and humans, in parallel with these other factors, does not resolve obesity-associated inflammation. Instead, inflammation persists amid modest changes in CD4+ T cell frequencies, highlighting the need for further research into resolving changes in T-cell function after weight loss. How lingering inflammation after weight loss affecting the common struggle to maintain lower weight is unknown. Semaglutide, a newly popular pharmaceutical used for treating T2D and reversing obesity, holds promise for alleviating obesity-associated health complications, yet its impact on T-cell-mediated inflammation remains unexplored. Further work in this area could significantly contribute to the scientific understanding of the impacts of weight loss and sex/hormones in obesity and obesity-associated metabolic decline.
Collapse
Affiliation(s)
- Yolander Valentine
- Department of Pharmacology and Nutritional Science, University of Kentucky, Lexington, Kentucky, USA
| | - Barbara S. Nikolajczyk
- Department of Pharmacology and Nutritional Science, University of Kentucky, Lexington, Kentucky, USA
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky, USA
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, Kentucky, USA
- Barnstable Brown Diabetes and Obesity Research Center, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
13
|
Sal-Sarria S, López-Taboada I, González-Pardo H, Conejo NM. A shift to a standard diet after exposure to a high-fat, high-sucrose diet from gestation to weaning restores brain metabolism and behavioral flexibility in adult rats. Behav Brain Res 2024; 467:115020. [PMID: 38679144 DOI: 10.1016/j.bbr.2024.115020] [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: 02/07/2024] [Revised: 04/14/2024] [Accepted: 04/25/2024] [Indexed: 05/01/2024]
Abstract
Prolonged consumption of diets high in saturated fat and sugar has been related to obesity and overweight, which in turn are linked to cognitive impairment in both humans and rodents. This has become a current issue, especially in children and adolescents, because these stages are crucial to neurodevelopmental processes and programming of adult behavior. To evaluate the effects of gestational and early exposure to an obesogenic diet, three groups with different dietary patterns were established: high-fat and high-sucrose diet (HFS), standard diet (SD), and a dietary shift from a high-fat, high-sucrose diet to a standard diet after weaning (R). Spatial learning and behavioral flexibility in adult male and female Wistar rats were evaluated using the Morris water maze (MWM) at PND 60. Furthermore, regional brain oxidative metabolism was assessed in the prefrontal cortex and the hippocampus. Contrary to our hypothesis, the HFS diet groups showed similar performance on the spatial learning task as the other groups, although they showed impaired cognitive flexibility. The HFS group had increased brain metabolic capacity compared to that of animals fed the standard diet. Shifting from the HFS diet to the SD diet after weaning restored the brain metabolic capacity in both sexes to levels similar to those observed in animals fed the SD diet. In addition, animals in the R group performed similarly to those fed the SD diet in the Morris water maze in both tasks. However, dietary shift from HFS diet to standard diet after weaning had only moderate sex-dependent effects on body weight and fat distribution. In conclusion, switching from an HFS diet to a balanced diet after weaning would have beneficial effects on behavioral flexibility and brain metabolism, without significant sex differences.
Collapse
Affiliation(s)
- Saúl Sal-Sarria
- Laboratory of Neuroscience, Department of Psychology, University of Oviedo, Oviedo, Spain; Institute of Neurosciences of the Principality of Asturias (INEUROPA), Oviedo, Spain; Health Research Institute of the Principality of Asturias (ISPA), Oviedo, Spain
| | - Isabel López-Taboada
- Laboratory of Neuroscience, Department of Psychology, University of Oviedo, Oviedo, Spain; Institute of Neurosciences of the Principality of Asturias (INEUROPA), Oviedo, Spain; Health Research Institute of the Principality of Asturias (ISPA), Oviedo, Spain
| | - Héctor González-Pardo
- Laboratory of Neuroscience, Department of Psychology, University of Oviedo, Oviedo, Spain; Institute of Neurosciences of the Principality of Asturias (INEUROPA), Oviedo, Spain; Health Research Institute of the Principality of Asturias (ISPA), Oviedo, Spain
| | - Nélida M Conejo
- Laboratory of Neuroscience, Department of Psychology, University of Oviedo, Oviedo, Spain; Institute of Neurosciences of the Principality of Asturias (INEUROPA), Oviedo, Spain; Health Research Institute of the Principality of Asturias (ISPA), Oviedo, Spain.
| |
Collapse
|
14
|
Vick LV, Rosario S, Riess JW, Canter RJ, Mukherjee S, Monjazeb AM, Murphy WJ. Potential roles of sex-linked differences in obesity and cancer immunotherapy: revisiting the obesity paradox. NPJ METABOLIC HEALTH AND DISEASE 2024; 2:5. [PMID: 38800540 PMCID: PMC11116109 DOI: 10.1038/s44324-024-00007-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 04/08/2024] [Indexed: 05/29/2024]
Abstract
Obesity, a condition of excess adiposity usually defined by a BMI > 30, can have profound effects on both metabolism and immunity, connecting the condition with a broad range of diseases, including cancer and negative outcomes. Obesity and cancer have been associated with increased incidence, progression, and poorer outcomes of multiple cancer types in part due to the pro-inflammatory state that arises. Surprisingly, obesity has also recently been demonstrated in both preclinical models and clinical outcomes to be associated with improved response to immune checkpoint inhibition (ICI). These observations have laid the foundation for what has been termed the "obesity paradox". The mechanisms underlying these augmented immunotherapy responses are still unclear given the pleiotropic effects obesity exerts on cells and tissues. Other important variables such as age and sex are being examined as further affecting the obesity effect. Sex-linked factors exert significant influences on obesity biology, metabolism as well as differential effects of different immune cell-types. Age can be another confounding factor contributing to the effects on both sex-linked changes, immune status, and obesity. This review aims to revisit the current body of literature describing the immune and metabolic changes mediated by obesity, the role of obesity on cancer immunotherapy, and to highlight questions on how sex-linked differences may influence obesity and immunotherapy outcome.
Collapse
Affiliation(s)
- Logan V. Vick
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA USA
| | - Spencer Rosario
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY USA
- Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY USA
| | - Jonathan W. Riess
- Department of Medicine, Division of Hematology/Oncology, UC Davis Comprehensive Cancer Center, Sacramento, CA USA
| | - Robert J. Canter
- Department of Surgery, Division of Surgical Oncology, University of California Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA USA
| | - Sarbajit Mukherjee
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY USA
| | - Arta M. Monjazeb
- Department of Radiation Oncology, University of California Davis Comprehensive Cancer Center, University of California School of Medicine, Sacramento, CA USA
| | - William J. Murphy
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA USA
- Department of Internal Medicine, Division of Malignant Hematology, Cellular Therapy and Transplantation, University of California Davis School of Medicine, Sacramento, CA USA
| |
Collapse
|
15
|
Zhang J, Hu B, Deng X, Sun R, Zhang R, Chen K, Guo W. Multiomics analysis investigating the impact of a high-fat diet in female Sprague-Dawley rats: alterations in plasma, intestinal metabolism, and microbial composition. Front Nutr 2024; 11:1359989. [PMID: 38646105 PMCID: PMC11026666 DOI: 10.3389/fnut.2024.1359989] [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: 12/22/2023] [Accepted: 03/20/2024] [Indexed: 04/23/2024] Open
Abstract
Introduction With improvements in living conditions, modern individuals exhibit a pronounced inclination towards a high-fat diet, largely because of its distinctive gustatory appeal. However, the association between high-fat diets and metabolic complications has largely been ignored, and metabolic diseases such as obesity and non-alcoholic fatty liver disease now constitute a major public health concern. Because high-fat diets increase the risk of metabolic diseases, a thorough investigation into the impact of high-fat diets on gut microbiota and metabolism is required. Methods We utilize 16S rRNA sequencing and untargeted metabolomics analysis to demonstrate that SD rats fed a high-fat diet exhibited marked alterations in gut microbiota and plasma, intestinal metabolism. Results Changes in gut microbiota included a decreased abundance at phylum level for Verrucomicrobiota, and a decreased abundance at genus level for Akkermansia, Ralstonia, Bacteroides, and Faecalibacterium. Additionally, significant changes were observed in both intestinal and plasma metabolite levels, including an upregulation of bile acid metabolism, an upregulation of glucose-lipid metabolism, and increased levels of metabolites such as norlithocholic acid, cholic acid, D-fructose, D-mannose, fructose lactate, and glycerophosphocholine. We also investigated the correlations between microbial communities and metabolites, revealing a significant negative correlation between Akkermansia bacteria and cholic acid. Discussion Overall, our findings shed light on the relationship between symbiotic bacteria associated with high-fat diets and metabolic biomarkers, and they provide insights for identifying novel therapeutic approaches to mitigate disease risks associated with a high-fat diet.
Collapse
Affiliation(s)
- Jiacheng Zhang
- Department of Hepatobiliary, Pancreatic and Liver Transplantation Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou, China
- Open and Key Laboratory of Hepatobiliary and Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, Zhengzhou, China
| | - Binhong Hu
- College of Chemistry and Life Sciences, Chengdu Normal University, Chengdu, China
| | - Xin Deng
- College of Chemistry and Life Sciences, Chengdu Normal University, Chengdu, China
| | - Rong Sun
- College of Chemistry and Life Sciences, Chengdu Normal University, Chengdu, China
| | - Rong Zhang
- School of Physical Science and Technology, ShanghaiTech University, Shanghai, China
| | - Kuo Chen
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenzhi Guo
- Department of Hepatobiliary, Pancreatic and Liver Transplantation Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Open and Key Laboratory of Hepatobiliary and Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, Zhengzhou, China
- Henan Research Centre for Organ Transplantation, Zhengzhou, China
| |
Collapse
|
16
|
Stranahan AM, Tabet A, Anikeeva P. Region-specific targeting of microglia in vivo using direct delivery of tamoxifen metabolites via microfluidic polymer fibers. Brain Behav Immun 2024; 115:131-142. [PMID: 37820974 PMCID: PMC10842189 DOI: 10.1016/j.bbi.2023.09.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 09/07/2023] [Accepted: 09/30/2023] [Indexed: 10/13/2023] Open
Abstract
Region-specific genetic manipulation of glial cells remains challenging due to the lack of anatomically selective transgenic models. Although local transduction is achievable with viral vectors, uniform recombination can be challenging in larger brain regions. We investigated the efficacy of intraparenchymal delivery of the tamoxifen metabolite endoxifen using inducible cre reporter mice. After observing localized reporter induction following stereotaxic injections of endoxifen in CX3CR1creERT2 mice, we carried out chronic delivery via osmotic pumps attached to bilateral cannulas made of stainless steel or microfluidic polymer fibers. Analysis of reporter expression in sections or iDISCO-cleared brains from TMEM119creERT2 mice revealed widespread induction following chronic infusion. Neuronal damage and gliosis were more prevalent around steel cannulas than polymer fibers, and glial reactivity was further attenuated when devices were implanted two months before drug delivery. In summary, region-specific recombination is achievable in glia with minimal tissue damage after endoxifen delivery via microfluidic polymer implants.
Collapse
Affiliation(s)
- Alexis M Stranahan
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th St, Augusta, GA 30912, USA.
| | - Anthony Tabet
- Research Laboratory of Electronics and McGovern Institute for Brain Research, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139-4307, USA
| | - Polina Anikeeva
- Research Laboratory of Electronics and McGovern Institute for Brain Research, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139-4307, USA; Departments of Materials Science & Engineering and Brain & Cognitive Sciences, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139-4307, USA
| |
Collapse
|
17
|
Yu Z, Liu H, Liu X, Tao Y, Zhang X, Zhao X, Chang H, Huang J, Zhao Y, Zhang H, Huang C. Dynamic changes in ambient PM 2.5 and body mass index among old adults: a nationwide cohort study. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:115929-115937. [PMID: 37897584 DOI: 10.1007/s11356-023-30620-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 10/18/2023] [Indexed: 10/30/2023]
Abstract
Outdoor air pollution has been considered as a severe environmental health issue that almost affecting everyone in the world, and intensive actions were launched. However, little is known about the association between dynamic changes in ambient fine particulate matter (PM2.5) exposure and body mass index (BMI) among old adults. To investigate the dynamic changes in ambient PM2.5 and body mass index among the elderly, we included a total of 7204 participants from 28 provinces of China during 2011-2015 in the China Health and Retirement Longitudinal Study (CHARLS). Ambient fine particle matter (PM2.5) was estimated using a well-validated space-time extremely randomized trees model. Change in PM2.5 and BMI (ΔPM2.5 and ΔBMI) were calculated as the value at a follow-up visit minus value at baseline. Linear mixed-effects models were applied to quantify the associations, controlling for sociodemographic factors. We found that per 1 μg/m3 increase in PM2.5 exposure was associated with a 0.031-0.044 kg/m2 increase in BMI among the elderly. We observed an approximate linear concentration-response relationship of PM2.5 and BMI in each visit. Each 1 μg/m3 increase in ΔPM2.5 exposure was associated with an increase in ΔBMI (β = 0.040, 95% CI 0.030, 0.049), while per 1 μg/m3 decrease in the ΔPM2.5 exposure level was associated with a decrease in ΔBMI (β = -0.016, 95% CI -0.027, -0.004). Our findings suggest that dynamic changes in ambient PM2.5 was positively associated with changes in BMI among old Chinese population.
Collapse
Affiliation(s)
- Zengli Yu
- School of Public Health, Zhengzhou University, 100 Science Avenue, Zhengzhou, 450001, China
| | - Hongyan Liu
- Department of Medical Genetics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaozhuan Liu
- Department of Medical Genetics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuchang Tao
- Department of Medical Genetics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaoan Zhang
- The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xin Zhao
- The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hui Chang
- The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jia Huang
- Department of Medical Genetics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuanfang Zhao
- School of Public Health, Zhengzhou University, 100 Science Avenue, Zhengzhou, 450001, China
| | - Huanhuan Zhang
- Department of Medical Genetics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China.
- School of Public Health, Zhengzhou University, 100 Science Avenue, Zhengzhou, 450001, China.
| | - Cunrui Huang
- Vanke School of Public Health, Tsinghua University, Beijing, China
| |
Collapse
|
18
|
Rouskas K, Katsareli EA, Amerikanou C, Dimopoulos AC, Glentis S, Kalantzi A, Skoulakis A, Panousis N, Ongen H, Bielser D, Planchon A, Romano L, Harokopos V, Reczko M, Moulos P, Griniatsos I, Diamantis T, Dermitzakis ET, Ragoussis J, Dedoussis G, Dimas AS. Identifying novel regulatory effects for clinically relevant genes through the study of the Greek population. BMC Genomics 2023; 24:442. [PMID: 37543566 PMCID: PMC10403965 DOI: 10.1186/s12864-023-09532-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: 03/02/2023] [Accepted: 07/25/2023] [Indexed: 08/07/2023] Open
Abstract
BACKGROUND Expression quantitative trait loci (eQTL) studies provide insights into regulatory mechanisms underlying disease risk. Expanding studies of gene regulation to underexplored populations and to medically relevant tissues offers potential to reveal yet unknown regulatory variants and to better understand disease mechanisms. Here, we performed eQTL mapping in subcutaneous (S) and visceral (V) adipose tissue from 106 Greek individuals (Greek Metabolic study, GM) and compared our findings to those from the Genotype-Tissue Expression (GTEx) resource. RESULTS We identified 1,930 and 1,515 eGenes in S and V respectively, over 13% of which are not observed in GTEx adipose tissue, and that do not arise due to different ancestry. We report additional context-specific regulatory effects in genes of clinical interest (e.g. oncogene ST7) and in genes regulating responses to environmental stimuli (e.g. MIR21, SNX33). We suggest that a fraction of the reported differences across populations is due to environmental effects on gene expression, driving context-specific eQTLs, and suggest that environmental effects can determine the penetrance of disease variants thus shaping disease risk. We report that over half of GM eQTLs colocalize with GWAS SNPs and of these colocalizations 41% are not detected in GTEx. We also highlight the clinical relevance of S adipose tissue by revealing that inflammatory processes are upregulated in individuals with obesity, not only in V, but also in S tissue. CONCLUSIONS By focusing on an understudied population, our results provide further candidate genes for investigation regarding their role in adipose tissue biology and their contribution to disease risk and pathogenesis.
Collapse
Affiliation(s)
- Konstantinos Rouskas
- Institute for Bioinnovation, Biomedical Sciences Research Center 'Alexander Fleming', Vari, Greece
- Institute of Applied Biosciences, Centre for Research & Technology Hellas, Thessaloniki, Greece
| | - Efthymia A Katsareli
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, Athens, Greece
| | - Charalampia Amerikanou
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, Athens, Greece
| | - Alexandros C Dimopoulos
- Institute for Fundamental Biomedical Science, Biomedical Sciences Research Center 'Alexander Fleming', Vari, Greece
- Hellenic Naval Academy, Hatzikyriakou Avenue, Pireaus, Greece
| | - Stavros Glentis
- Institute for Bioinnovation, Biomedical Sciences Research Center 'Alexander Fleming', Vari, Greece
- Pediatric Hematology/Oncology Unit (POHemU), First Department of Pediatrics, University of Athens, Aghia Sophia Children's Hospital, Athens, Greece
| | - Alexandra Kalantzi
- Institute for Bioinnovation, Biomedical Sciences Research Center 'Alexander Fleming', Vari, Greece
| | - Anargyros Skoulakis
- Institute for Bioinnovation, Biomedical Sciences Research Center 'Alexander Fleming', Vari, Greece
| | | | - Halit Ongen
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
- Swiss Institute of Bioinformatics, University of Geneva, Geneva, Switzerland
- Institute of Genetics and Genomics in Geneva, University of Geneva, Geneva, Switzerland
| | - Deborah Bielser
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
| | - Alexandra Planchon
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
| | - Luciana Romano
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
| | - Vaggelis Harokopos
- Institute for Bioinnovation, Biomedical Sciences Research Center 'Alexander Fleming', Vari, Greece
| | - Martin Reczko
- Institute for Fundamental Biomedical Science, Biomedical Sciences Research Center 'Alexander Fleming', Vari, Greece
| | - Panagiotis Moulos
- Institute for Fundamental Biomedical Science, Biomedical Sciences Research Center 'Alexander Fleming', Vari, Greece
- Center of New Biotechnologies & Precision Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioannis Griniatsos
- First Department of Surgery, National and Kapodistrian University of Athens, Medical School, Laiko Hospital, Athens, Greece
| | - Theodoros Diamantis
- First Department of Surgery, National and Kapodistrian University of Athens, Medical School, Laiko Hospital, Athens, Greece
| | - Emmanouil T Dermitzakis
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
| | - Jiannis Ragoussis
- Department of Human Genetics, McGill University Genome Centre, McGill University, Montréal, QC, Canada
- Department of Bioengineering, McGill University, Montréal, QC, Canada
| | - George Dedoussis
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, Athens, Greece
| | - Antigone S Dimas
- Institute for Bioinnovation, Biomedical Sciences Research Center 'Alexander Fleming', Vari, Greece.
| |
Collapse
|
19
|
Wang X, Sundermann EE, Buckley RF, Reas ET, McEvoy LK, Banks SJ. Sex Differences in the Associations of Obesity with Tau, Amyloid PET, and Cognitive Outcomes in Preclinical Alzheimer's Disease: Cross-Sectional A4 Study. J Alzheimers Dis 2023; 95:615-624. [PMID: 37574737 PMCID: PMC11827697 DOI: 10.3233/jad-230466] [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: 08/15/2023]
Abstract
BACKGROUND The association between obesity and Alzheimer's disease (AD) is complex. Recent studies indicated the relationships between obesity and AD may differ by sex, and women may benefit from being overweight in terms of AD risk. OBJECTIVE We investigated whether sex modifies the associations of obesity with tau positron emission tomography (PET), amyloid PET, and cognition in preclinical AD. METHODS We included 387 cognitively-unimpaired amyloid-positive participants (221 women, 166 men, 87.6% non-Hispanic White) with available 18F-flortaucipir PET, 18F-florbetapir PET, and completed the Preclinical Alzheimer Cognitive Composite (PACC) tests from the Anti-Amyloid Treatment in Asymptomatic Alzheimer's Disease (A4) study. Participants were categorized based on body mass index (BMI: kg/m2): normal-weight (BMI: 18.5-25), overweight (BMI: 25-30), and obese (BMI≥30). RESULTS Significant sex by BMI category interactions on PACC and its components: Mini-Mental State Examination (MMSE) and Reminding Test-Free+Total Recall (FCSRT96) revealed that overweight and obese women outperformed normal-weight women on FCSRT96, while obese men showed poorer MMSE performance than normal-weight men. These interactions were independent of APOE4. There were no significant interactions of sex by BMI category on tau and amyloid PET. However, sex-stratified analyses observed obesity was associated with less regional tau and mean cortical amyloid in women, not in men. CONCLUSION This study found that in preclinical AD, overweight and obesity were associated with better verbal memory in women, whereas obesity was associated with worse global cognition among men. Future studies focusing on the mechanism for this relationship may inform sex-specific interventions for AD prevention.
Collapse
Affiliation(s)
- Xin Wang
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Erin E. Sundermann
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Rachel F. Buckley
- Department of Neurology, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | - Emilie T. Reas
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Linda K. McEvoy
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego, La Jolla, CA, USA
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA
| | - Sarah J. Banks
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|