1
|
Zhou H, Feng S, Cai J, Shao X, Zhu S, Zhou H, Cao Y, Wang R, Lin X, Wang J. Oestrogen suppresses the adipogenesis of fibro/adipogenic progenitors through reactivating the METTL3-ESR1-mediated loop in post-menopausal females. Clin Transl Med 2025; 15:e70206. [PMID: 39875775 PMCID: PMC11774659 DOI: 10.1002/ctm2.70206] [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: 09/30/2024] [Revised: 01/12/2025] [Accepted: 01/19/2025] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Post-menopausal women experience more severe muscular fatty infiltration, though the mechanisms remain unclear. The decline in estrogen levels is considered as a critical physiological alteration during post-menopause. Fibro/adipogenic progenitors (FAPs) are identified as major contributors to muscular fatty infiltration. This study aimed to investigate the detailed mechanism underlying the excessive muscular fatty infiltration in postmenopausal females. METHODS Supraspinatus muscle samples were collected from female patients with or without menopause, and from mice with or without ovariectomy (OVX), to evaluate muscular fatty infiltration and isolated FAPs. The expressions of (estrogen receptor 1) ESR1, methyltransferase-like 3 (METTL3), and adipogenesis ability in FAPs from post-menopausal women and OVX mice were investigated. RNA sequencing (RNA-Seq) was performed to explore the gene expression profiles and potential mechanisms in FAPs from Pdgfrα-CreERT2; Esr1 knockout (Esr1 KO) mice and Esr1 flox/flox (Esr1 f/f) mice. The interplay of the METTL3-ESR1 mediated loop and its role in regulating adipogenesis in FAPs were investigated using dual luciferase reporter assays, chromatin immunoprecipitation (ChIP), and protein and RNA stability assays. The effects of estrogen supplementation on muscular fatty infiltration and locomotor function in OVX mice were evaluated by immunofluorescent staining and functional analysis. RESULTS Decreased expression of ESR1/METTL3 and increased adipogenesis ability in FAPs was found in post-menopausal female. METTL3-mediated m6A methylation promoted ESR1 mRNA stability at the post-transcriptional level in FAPs. METTL3-mediated m6A modification promoted ESR1 expression by stabilizing ESR1 mRNA, while ESR1 acted as a transcription factor that enhanced METTL3 transcription in turn. ESR1 also suppressed the transcription of the adipogenic transcription factor peroxisome proliferator-activated receptor gamma (PPARγ), thereby inhibiting adipogenesis in FAPs. Reactivation of the METTL3-ESR1 mediated loop by estrogen alleviated excessive adipogenesis in FAPs from post-menopausal women, and it also reduced muscular fatty infiltration, and improved locomotor function in OVX mice. CONCLUSION Excessive muscular fatty infiltration in post-menopausal women arose from the disruption of the METTL3-ESR1 mediated loop of FAPs due to estrogen deficiency. Reactivation of the METTL3-ESR1 mediated loop by estrogen may serve as a novel intervention to inhibit excessive adipogenesis of post-menopausal female FAPs, thereby ameliorating muscular fatty infiltration and improving locomotor function in post-menopausal females. KEY POINTS Oestrogen insufficiency disrupted the METTL3ESR1 loop in post-menopausal FAPs, causing excessive muscular fatty infiltration. METTL3-mediated m6A modification stabilized ESR1 mRNA and enhanced ESR1 expression, while increased ESR1 further promoted METTL3 transcription. ESR1 inhibited the transcription of adipogenic factor PPARγ, ameliorating adipogenesis in FAPs. Reactivating the METTL3ESR1 loop via oestrogen in FAPs reduced muscular fatty infiltration and improved locomotor function.
Collapse
Affiliation(s)
- Hao Zhou
- Xinhua Hospital Affiliated to Shanghai Jiaotong University School of MedicineShanghaiChina
| | - Shujing Feng
- School of Exercise and HealthShanghai University of SportShanghaiChina
| | - Jinkui Cai
- Wuhan Third HospitalTongren Hospital of Wuhan UniversityWuhanChina
| | - Xiexiang Shao
- Xinhua Hospital Affiliated to Shanghai Jiaotong University School of MedicineShanghaiChina
| | - Siyuan Zhu
- Department of Hand SurgeryHuashan HospitalFudan UniversityShanghaiChina
| | - Han Zhou
- Xinhua Hospital Affiliated to Shanghai Jiaotong University School of MedicineShanghaiChina
| | - Yongmin Cao
- School of Exercise and HealthShanghai University of SportShanghaiChina
| | - Ru Wang
- School of Exercise and HealthShanghai University of SportShanghaiChina
| | | | - Jianhua Wang
- Xinhua Hospital Affiliated to Shanghai Jiaotong University School of MedicineShanghaiChina
| |
Collapse
|
2
|
Wang Y, Liu H, Zhou B, Yue W, Wang M, Hu K. Menopause and obstructive sleep apnea: revealing an independent mediating role of visceral fat beyond body mass index. BMC Endocr Disord 2025; 25:21. [PMID: 39863851 PMCID: PMC11765922 DOI: 10.1186/s12902-025-01850-2] [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: 10/18/2024] [Accepted: 01/20/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Menopause is a significant phase in women's health, in which the incidence of obstructive sleep apnea (OSA) is significantly increased. Body fat distribution changes with age and hormone levels in postmenopausal women, but the extent to which changes in body fat distribution affect the occurrence of OSA is unclear. METHODS This research performed a cross-sectional analysis utilizing data from the 2015-2016 National Health and Nutrition Examination Survey (NHANES). Body fat distribution was quantified using dual-energy X-ray absorptiometry in kilograms. Menopausal status and OSA symptoms were determined by questionnaire. Weighted multivariable regression analysis was utilized to investigate the correlation between menopausal status and OSA symptoms and body fat composition. We did a mediation analysis to assess how much of the effect of menopausal status on OSA symptoms was mediated through in body fat composition. RESULTS The analysis comprised 1459 individuals from NHANES, consisting of 1188 premenopausal and 271 postmenopausal women. In the weighted sample, 36.01% of premenopausal women and 53.39% of postmenopausal women had OSA symptoms. After adjusting for body mass index (BMI) and other potential confounders, menopausal status was correlated with a higher prevalence of OSA symptoms (OR = 1.57; 95% CI: 1.16,2.13), and increased visceral fat mass (β = 0.12; 95% CI: 0.07, 0.17). In addition, visceral fat mass exhibited a significant correlation with OSA symptoms (OR = 3.79; 95% CI: 1.61, 8.94). Mediation analysis showed that 29.76% of the effect of menopausal status on OSA symptoms was mediated through visceral fat. In age-matched analysis, postmenopausal women had higher visceral fat mass (0.63 kg vs. 0.52 kg, P = 0.02) and a higher prevalence of OSA symptoms (68.3% vs. 45.7%, P = 0.02) compared with premenopausal women; however, there was no significant difference in BMI (P > 0.05). CONCLUSION Our results suggest that menopausal status is associated with increased visceral fat accumulation and OSA symptoms prevalence. Visceral fat accumulation appears to play an important role in the development of OSA in postmenopausal women, independent of BMI; this highlights the importance of further studying this relationship.
Collapse
Affiliation(s)
- Yuhan Wang
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Hailing Liu
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Beini Zhou
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Wuriliga Yue
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Mengcan Wang
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ke Hu
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
3
|
McCormick JJ, Goulet N, King KE, Fujii N, Amano T, Kenny GP. The effect of high-intensity exercise in temperate and hot ambient conditions on autophagy and the cellular stress response in young and older females. Am J Physiol Regul Integr Comp Physiol 2025; 328:R90-R101. [PMID: 39601770 DOI: 10.1152/ajpregu.00178.2024] [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: 07/09/2024] [Revised: 09/23/2024] [Accepted: 10/08/2024] [Indexed: 11/29/2024]
Abstract
The process of autophagy is vital in maintaining normal cellular function, especially during exposure to elevated states of physiological stress associated with exercise and hot ambient temperatures. Although prior observations are primarily limited to responses in males, the autophagic response to acute physiological stress in females represents a considerable knowledge gap. Therefore, we assessed autophagy and related pathways of cellular stress in peripheral blood mononuclear cells (PBMCs) from 20 healthy young [n = 10, mean (SD): aged 23 yr (3)] and older [n = 10, aged 69 yr (3)] females in response to 30 min of semi-recumbent high-intensity cycling exercise (70% of predetermined maximal oxygen consumption) in temperate (25°C) and hot (40°C) ambient conditions (15% relative humidity). Mean body temperature (rectal and skin) was measured throughout, whereas cellular responses were evaluated before and after exercise, including up to 6 h of seated recovery. Proteins associated with autophagy and related pathways were assessed via Western blot. Mean body temperature was elevated after exercise in both conditions, with significant elevations observed after exercise in the heat (all, P ≤ 0.05). Although young females displayed signs of elevated autophagic activity [elevations in microtubule-associated light chain 3B (LC3)-II and beclin-2] in response to exercise performed in both temperate and hot ambient conditions (all, P ≤ 0.05), responses were attenuated in older females. This was accompanied by elevations in chaperone-mediated autophagy in young but not in older females in response to exercise independent of ambient temperature. Our findings indicate exercise, with and without ambient heat exposure may stimulate the autophagic response in young but not in older females.NEW & NOTEWORTHY We show for the first time that an acute bout (30 min) of high-intensity intensity exercise stimulates autophagy in young females irrespective of ambient heat exposure. However, older females did not display the same increase in autophagy as their younger counterparts when high-intensity exercise was performed in temperate or hot ambient conditions. Consequently, older females may be at an elevated risk of heat-induced cellular damage during exertional heat stress.
Collapse
Affiliation(s)
- James J McCormick
- Human and Environmental Physiology Research Unit, School of Human Kinetics, University of Ottawa, Ottawa, Ontario, Canada
| | - Nicholas Goulet
- Human and Environmental Physiology Research Unit, School of Human Kinetics, University of Ottawa, Ottawa, Ontario, Canada
| | - Kelli E King
- Human and Environmental Physiology Research Unit, School of Human Kinetics, University of Ottawa, Ottawa, Ontario, Canada
| | - Naoto Fujii
- Institute of Health and Sport Sciences, University of Tsukuba, Tsukuba, Japan
| | - Tatsuro Amano
- Laboratory for Exercise and Environmental Physiology, Faculty of Education, Niigata University, Niigata, Japan
| | - Glen P Kenny
- Human and Environmental Physiology Research Unit, School of Human Kinetics, University of Ottawa, Ottawa, Ontario, Canada
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| |
Collapse
|
4
|
Zhao Y, Klionsky DJ, Wang X, Huang Q, Deng Z, Xiang J. The Estrogen-Autophagy Axis: Insights into Cytoprotection and Therapeutic Potential in Cancer and Infection. Int J Mol Sci 2024; 25:12576. [PMID: 39684286 DOI: 10.3390/ijms252312576] [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: 10/23/2024] [Revised: 11/14/2024] [Accepted: 11/16/2024] [Indexed: 12/18/2024] Open
Abstract
Macroautophagy, commonly referred to as autophagy, is an essential cytoprotective mechanism that plays a significant role in cellular homeostasis. It has emerged as a promising target for drug development aimed at treating various cancers and infectious diseases. However, the scientific community has yet to reach a consensus on the most effective approach to manipulating autophagy, with ongoing debates about whether its inhibition or stimulation is preferable for managing these complex conditions. One critical factor contributing to the variability in treatment responses for both cancers and infectious diseases is estrogen, a hormone known for its diverse biological effects. Given the strong correlations observed between estrogen signaling and autophagy, this review seeks to summarize the intricate molecular mechanisms that underlie the dual cytoprotective effects of estrogen signaling in conjunction with autophagy. We highlight recent findings from studies that involve various ligands, disease contexts, and cell types, including immune cells. Furthermore, we discuss several factors that regulate autophagy in the context of estrogen's influence. Ultimately, we propose a hypothetical model to elucidate the regulatory effects of the estrogen-autophagy axis on cell fate. Understanding these interactions is crucial for advancing our knowledge of related diseases and facilitating the development of innovative treatment strategies.
Collapse
Affiliation(s)
- Ying Zhao
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Daniel J Klionsky
- Life Sciences Institute, University of Michigan, Mary Sue Coleman Hall, 210 Washtenaw Avenue, Ann Arbor, MI 48109-2216, USA
| | - Xin Wang
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Qiaoying Huang
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Zixin Deng
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Jin Xiang
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| |
Collapse
|
5
|
Zhou H, Lin X, Feng S, Zhu S, Zhou H, Chen H, Youwu H, Wang Z, Wang R, Shao X, Wang J. Metformin mitigates adipogenesis of fibro-adipogenic progenitors after rotator cuff tears via activating mTOR/ULK1-mediated autophagy. Am J Physiol Cell Physiol 2024; 326:C1590-C1603. [PMID: 38586878 PMCID: PMC11371331 DOI: 10.1152/ajpcell.00034.2024] [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: 01/16/2024] [Revised: 03/28/2024] [Accepted: 03/29/2024] [Indexed: 04/09/2024]
Abstract
Muscular fatty infiltration is a common issue after rotator cuff tears (RCTs), which impair shoulder function. Females suffer a higher prevalence and a more severe degree of muscular fatty infiltration after RCT when compared with males, with the underlying mechanisms remaining unclear. Fibro-adipogenic progenitors (FAPs) are the primary source of muscular fatty infiltration following RCT. Our findings disclose that gender-specific disparities in muscular fatty infiltration are linked to mTOR/ULK1-mediated autophagy of FAPs. Decreased autophagic activity contributes to adipogenic differentiation in female FAPs after RCT. Furthermore, metformin could enhance mTOR/ULK1-mediated autophagic processes of FAPs, thereby alleviating fatty infiltration and improving shoulder functionality after RCT. Together, our study reveals that gender differences in muscular fatty infiltration arise from distinct autophagic activities. Metformin could be a promising noninvasive intervention to ameliorate muscular fatty infiltration of RCT.NEW & NOTEWORTHY The current study demonstrated that gender-specific disparities in muscular fatty infiltration are attributed to mTOR/ULK1-mediated autophagy of FAPs. Decreased autophagic activity contributes to adipogenic differentiation in female FAPs after RCT. Moreover, metformin could enhance mTOR/ULK1-mediated autophagic processes of FAPs, thereby alleviating fatty infiltration and improving shoulder functionality after RCT. Therefore, metformin could be a promising noninvasive intervention to ameliorate muscular fatty infiltration of RCT.
Collapse
Affiliation(s)
- Hao Zhou
- Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Xingzuan Lin
- Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Shujing Feng
- Department of Sports Medicine, School of Exercise and Health, Shanghai University of Sport, Shanghai, People's Republic of China
| | - Siyuan Zhu
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Han Zhou
- Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Huifang Chen
- Department of Sports Medicine, School of Exercise and Health, Shanghai University of Sport, Shanghai, People's Republic of China
| | - He Youwu
- Department of Hand Plastic Surgery, The First People's Hospital of Linping District, Hangzhou, People's Republic of China
| | - Zekai Wang
- Department of Life Science, University of Toronto, Toronto, Ontario, Canada
| | - Ru Wang
- Department of Sports Medicine, School of Exercise and Health, Shanghai University of Sport, Shanghai, People's Republic of China
| | - Xiexiang Shao
- Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Jianhua Wang
- Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
6
|
Shen F, Guo C, Zhang D, Liu Y, Zhang P. Visceral adiposity index as a predictor of type 2 diabetes mellitus risk: A systematic review and dose-response meta-analysis. Nutr Metab Cardiovasc Dis 2024; 34:811-822. [PMID: 38326187 DOI: 10.1016/j.numecd.2023.04.009] [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: 11/25/2022] [Revised: 04/02/2023] [Accepted: 04/07/2023] [Indexed: 02/09/2024]
Abstract
AIMS Considering the positive association between visceral adiposity index (VAI) and type 2 diabetes mellitus (T2DM), no comprehensive assessment on the summarized and dose-response relationship between VAI and T2DM has yet been reported. Therefore, we performed a meta-analysis, including dose-response analysis, to quantitively elucidate this association. DATA SYNTHESIS MEDLINE via PubMed and Embase databases were searched for relevant articles up to December 14, 2021. Random-effects generalized least squares regression models were used to assess the quantitative association between VAI and T2DM risk across studies. Restricted cubic splines were used to model the dose-response association. A total of 9 prospective cohort studies and 5 cross sectional studies were included in our review. Based on the meta-analysis, the pooled RR of T2DM was 2.05 (95% CI 1.74-2.41) for the highest versus reference VAI category. We found that the risk of T2DM was increased by 44% (RR, 1.44; 95% CI, 1.23-1.68) with each 1-unit increment of VAI. While, we found no evidence of a nonlinear dose-response association of VAI and T2DM (Pnon-linearity = 0.428). With the linear cubic spline model, when compared to population with VAI at 0.6, for those with VAI at 2.0, the risk of T2DM was increased by 81% (RR, 1.81; 95% CI 1.55-2.12). CONCLUSIONS Our meta-analysis provides quantitative data suggesting that VAI is associated with an increased risk of T2DM. Public health strategies focusing on weight loss among obesity, especially the people characterized by the thin-on-the-outside--fat-on-the-inside phenotype could possibly reduce a substantial risk of T2DM. SYSTEMATIC REVIEW REGISTRATION PROSPERO CRD42022372666.
Collapse
Affiliation(s)
- Fang Shen
- Department of Clinical Nutrition, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China.
| | - Changman Guo
- Department of Prevention and Treatment of infectious disease, Center for Disease Control and Prevention of Xihu District, Hangzhou, Zhejiang, 310000, China.
| | - Dongdong Zhang
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China.
| | | | - Pianhong Zhang
- Department of Clinical Nutrition, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China.
| |
Collapse
|
7
|
Kuang M, Qiu J, Li D, Hu C, Zhang S, Sheng G, Zou Y. The newly proposed Metabolic Score for Visceral Fat is a reliable tool for identifying non-alcoholic fatty liver disease, requiring attention to age-specific effects in both sexes. Front Endocrinol (Lausanne) 2023; 14:1281524. [PMID: 38089634 PMCID: PMC10711077 DOI: 10.3389/fendo.2023.1281524] [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: 08/24/2023] [Accepted: 11/10/2023] [Indexed: 12/18/2023] Open
Abstract
Objective The newly proposed Metabolic Visceral Fat Score (METS-VF) is considered a more effective measure for visceral adipose tissue (VAT) than other obesity indicators. This study aimed to reveal the association between METS-VF and non-alcoholic fatty liver disease (NAFLD), and its variations across age groups within both sexes. Methods Data from 14,251 medical examiners in the NAGALA project were employed in this study. 3D fitted surface plots were constructed based on multivariate logistic regression models to visualize the isolated and combined effects of aging and METS-VF on NAFLD. Receiver operating characteristic curve (ROC) analysis was conducted to compare the diagnostic performance of METS-VF with other VAT surrogate markers in predicting NAFLD. Results The results of multivariate logistic regression analysis showed that each unit increase in METS-VF was independently associated with a 333% and 312% increase in the odds of NAFLD in males and females, respectively. Additionally, the 3D fitted surface plot showed that age significantly influenced the association between METS-VF and the odds of NAFLD in both sexes, as follows: (i) In males, when METS-VF was less than 6.2, the METS-VF-related odds of NAFLD increased gradually with age in the 20-45 age group, reached a plateau in the 45-65 age group, and then decreased in the group above 65 years old; however, when male METS-VF exceeded 6.2, aging and METS-VF combined to further increase the odds of NAFLD in all age groups, particularly in the 45-65 age group. (ii) In females, aging seemed to reduce METS-VF-related odds of NAFLD in the 18-40 age group, but significantly increased it in the 40-60 age group, particularly for those with higher METS-VF levels. Further ROC analysis revealed that compared to other VAT surrogate markers, METS-VF showed the highest diagnostic accuracy for NAFLD in females, especially in those under 45 years of age [area under the curve (AUC) = 0.9256]. Conclusions This study firstly revealed a significant positive correlation between METS-VF and the odds of NAFLD, with METS-VF surpassing other VAT surrogate markers in NAFLD diagnosis. Moreover, age significantly influenced the METS-VF-related odds of NAFLD and METS-VF's diagnostic efficacy for NAFLD in both sexes.
Collapse
Affiliation(s)
- Maobin Kuang
- Department of Internal Medicine, Medical College of Nanchang University, Jiangxi Provincial People’s Hospital, Nanchang, Jiangxi, China
- Jiangxi Cardiovascular Research Institute, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
- Jiangxi Provincial Geriatric Hospital, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Jiajun Qiu
- Department of Internal Medicine, Medical College of Nanchang University, Jiangxi Provincial People’s Hospital, Nanchang, Jiangxi, China
- Jiangxi Cardiovascular Research Institute, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
- Jiangxi Provincial Geriatric Hospital, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Dongdong Li
- Department of Internal Medicine, Medical College of Nanchang University, Jiangxi Provincial People’s Hospital, Nanchang, Jiangxi, China
- Jiangxi Cardiovascular Research Institute, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
- Jiangxi Provincial Geriatric Hospital, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
- Department of Pulmonary and Critical Care Medicine, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Chong Hu
- Department of Gastroenterology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Shuhua Zhang
- Jiangxi Cardiovascular Research Institute, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Guotai Sheng
- Jiangxi Provincial Geriatric Hospital, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Yang Zou
- Jiangxi Cardiovascular Research Institute, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| |
Collapse
|
8
|
Saito Y, Kobayashi M, Tamaki S, Nakamura K, Hirate D, Takahashi K, Takekuma Y, Sakakibara-Konishi J, Shimizu Y, Kinoshita I, Sugawara M. Risk factor analysis for cisplatin-induced nephrotoxicity with the short hydration method in diabetic patients. Sci Rep 2023; 13:17126. [PMID: 37816823 PMCID: PMC10564853 DOI: 10.1038/s41598-023-44477-w] [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: 06/22/2023] [Accepted: 10/09/2023] [Indexed: 10/12/2023] Open
Abstract
The occurrence of cisplatin (CDDP)-induced nephrotoxicity (CIN) has decreased with advancements in supportive care. In contrast, we reported that baseline diabetes mellitus (DM) complications significantly worsen CIN. This study aimed to determine further risk factors associated with CIN development in DM patients. Patients with thoracic cancer requiring DM pharmacotherapy, who received CDDP (≥ 60 mg/m2)-containing regimens using the short hydration method (n = 140), were enrolled in this retrospective multicenter observational study. The primary endpoint of the present study was the elucidation of risk factors (patient factors, DM medication influence, and treatment-related factors) associated with CIN development in patients with DM. Cisplatin-induced nephrotoxicity occurred in 22.1% of patients with DM. The median worst variation of serum creatinine levels and creatinine clearance (worst level - baseline level) was 0.16 mg/dL (range: - 0.12-1.41 mg/dL) and - 15.9 mL/min (- 85.5-24.3 mL/min), respectively. Multivariate logistic regression analyses identified female sex as the singular risk factor for CIN development in the DM population (adjusted odds ratio; 2.87, 95% confidence interval; 1.08-7.67, P = 0.04). Diabetes mellitus medication and treatment-related factors did not affect CIN development. In conclusion, our study revealed that female sex is significantly associated with CIN development in patients with DM and thoracic cancer.
Collapse
Affiliation(s)
- Yoshitaka Saito
- Department of Clinical Pharmaceutics & Therapeutics, Faculty of Pharmaceutical Sciences, Hokkaido University of Science, 4-1, Maeda 7-Jo 15-Chome, Teine-Ku, Sapporo, 006-8585, Japan.
- Department of Pharmacy, Hokkaido University Hospital, Kita 14-Jo, Nishi 5-Chome, Kita-Ku, Sapporo, 060-8648, Japan.
| | - Masaki Kobayashi
- Laboratory of Clinical Pharmaceutics & Therapeutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita 12-Jo, Nishi 6-Chome, Kita-Ku, Sapporo, 060-0812, Japan
| | - Shinya Tamaki
- Department of Pharmacy, KKR Sapporo Medical Center, 3-40, Hiragishi 1-Jo 6-Chome, Toyohira-Ku, Sapporo, 062-0931, Japan
| | - Katsuyuki Nakamura
- Division of Hospital Pharmacy, Sapporo Medical University, 291, Minami 1-Jo, Nishi 16-Chome, Chuo-Ku, Sapporo, 060-8543, Japan
| | - Daisuke Hirate
- Department of Pharmacy, Teine Keijinkai Hospital, 1-40, Maeda 1-Jo 12-Chome, Teine-Ku, Sapporo, 006-8555, Japan
| | - Kenta Takahashi
- Department of Pharmacy, NTT Medical Center Sapporo, Minami 1-Jo, Nishi 15-Chome, Chuo-Ku, Sapporo, 060-0061, Japan
| | - Yoh Takekuma
- Department of Pharmacy, Hokkaido University Hospital, Kita 14-Jo, Nishi 5-Chome, Kita-Ku, Sapporo, 060-8648, Japan
| | - Jun Sakakibara-Konishi
- Department of Respiratory Medicine, Faculty of Medicine, Hokkaido University, Kita 15-Jo, Nishi 7-Chome, Kita-Ku, Sapporo, 060-8638, Japan
| | - Yasushi Shimizu
- Department of Medical Oncology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita 15-Jo, Nishi 7-Chome, Kita-Ku, Sapporo, 060-8638, Japan
| | - Ichiro Kinoshita
- Department of Medical Oncology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita 15-Jo, Nishi 7-Chome, Kita-Ku, Sapporo, 060-8638, Japan
| | - Mitsuru Sugawara
- Department of Pharmacy, Hokkaido University Hospital, Kita 14-Jo, Nishi 5-Chome, Kita-Ku, Sapporo, 060-8648, Japan
- Laboratory of Pharmacokinetics, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita 12-Jo, Nishi 6-Chome, Kita-Ku, Sapporo, 060-0812, Japan
| |
Collapse
|
9
|
Peng W, Han M, Xu G. The association between visceral adiposity index and chronic kidney disease in the elderly: A cross-sectional analysis of NHANES 2011-2018. Prev Med Rep 2023; 35:102306. [PMID: 37449005 PMCID: PMC10336672 DOI: 10.1016/j.pmedr.2023.102306] [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: 01/02/2023] [Revised: 05/23/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
Visceral adiposity index (VAI) is a new metric for evaluating visceral adiposity dysfunction. The aim of the present study was to explore the association between VAI and incident chronic kidney disease (CKD) in the American elderly population. We included 6085 participants aged 60 years or older with available data on renal function and VAI from the National Health and Nutrition Examination Survey (NHANES) from 2011 to 2018 and divided them into four groups according to the VAI quartiles. VAI values were ln-transformed to reduce skewness. We conducted multivariable logistic regression analyses and spline smoothing plot analyses to evaluate the association between VAI and CKD. After fully adjusting for confounding factors, higher lnVAI was associated with a higher risk of CKD (OR, 1.23; 95 %CI, 1.02, 1.48). Compared to the Q1 group (VAI < 1.1), The third and fourth VAI quartiles were associated with a higher prevalence of CKD (Q3: VAI, 1.8-3.0; Q4: VAI > 3.0). The adjusted ORs (95% CI) in Q3 and Q4 were 1.74 (1.24, 2.42) and 1.59 (1.08, 2.33). Adjusted smoothed plots suggested a nonlinear relationship between lnVAI and CKD. The development of CKD increased with increasing VAI among participants with a lnVAI between -0.6 and 1.6 (VAI: 0.5-5.0). In conclusion, VAI is significantly associated with the risk of CKD in the US elderly population aged 60 years or older.
Collapse
Affiliation(s)
| | - Min Han
- Corresponding author at: Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, China.
| | | |
Collapse
|
10
|
Zhang QH, Chen LH, An Q, Pi P, Dong YF, Zhao Y, Wang N, Fang X, Pu RW, Song QW, Lin LJ, Liu JH, Liu AL. Quantification of the renal sinus fat and exploration of its relationship with ectopic fat deposition in normal subjects using MRI fat fraction mapping. Front Endocrinol (Lausanne) 2023; 14:1187781. [PMID: 37621645 PMCID: PMC10446762 DOI: 10.3389/fendo.2023.1187781] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 05/29/2023] [Indexed: 08/26/2023] Open
Abstract
Purpose To determine the renal sinus fat (RSF) volume and fat fraction (FF) in normal Chinese subjects using MRI fat fraction mapping and to explore their associations with age, gender, body mass index (BMI) and ectopic fat deposition. Methods A total of 126 subjects were included in the analysis. RSF volume and FF, visceral adipose tissue (VAT) and subcutaneous adipose tissue (SAT) area, and hepatic and pancreatic FFs were measured for each subject. The comparisons in gender were determined using two-tailed t-tests or the nonparametric Mann-Whitney U-test for normally or non-normally distributed data for continuous variables and the chi-square test for categorical variables. Comparisons of RFS volume and FF between right and left kidneys were determined using paired sample t-tests. Multivariable logistic models were performed to confirm whether RSF differences between men and women are independent of VAT or SAT area. When parameters were normally distributed, the Pearson correlation coefficient was used; otherwise, the Spearman correlation coefficient was applied. Results The RSF volumes (cm3) of both kidneys in men (26.86 ± 8.81 for right and 31.62 ± 10.32 for left kidneys) were significantly bigger than those of women (21.47 ± 6.90 for right and 26.03 ± 8.55 for left kidneys) (P < 0.05). The RSF FFs (%) of both kidneys in men (28.33 ± 6.73 for right and 31.21 ± 6.29 for left kidneys) were significantly higher than those of the women (23.82 ± 7.74 for right and 27.92 ± 8.15 for left kidneys) (P < 0.05). The RSF differences between men and women are independent of SAT area and dependent of VAT area (except for right RSF volume). In addition, the RSF volumes and FFs in both kidneys in the overall subjects show significant correlations with age, BMI, VAT area, hepatic fat fraction and pancreatic fat fraction (P < 0.05). However, the patterns of these correlations varied by gender. The RSF volume and FF of left kidney were significantly larger than those of the right kidney (P < 0.05). Conclusion The association between renal sinus fat and ectopic fat deposition explored in this study may help establish a consensus on the normal values of RSF volume and FF for the Chinese population. This will facilitate the identification of clinicopathological changes and aid in the investigation of whether RSF volume and FF can serve as early biomarkers for metabolic diseases and renal dysfunction in future studies.
Collapse
Affiliation(s)
- Qin-He Zhang
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Li-Hua Chen
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Qi An
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Peng Pi
- Department of Medical Imaging, Dalian Medical University, Dalian, China
| | - Yi-Fan Dong
- Department of Medical Imaging, Dalian Medical University, Dalian, China
| | - Ying Zhao
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Nan Wang
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xin Fang
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Ren-Wang Pu
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Qing-Wei Song
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Liang-Jie Lin
- Clinical & Technical Solutions, Philips Healthcare, Beijing, China
| | - Jing-Hong Liu
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Ai-Lian Liu
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
11
|
Liu S, Xu S, Liu S, Chen H. Importance of DJ-1 in autophagy regulation and disease. Arch Biochem Biophys 2023:109672. [PMID: 37336341 DOI: 10.1016/j.abb.2023.109672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/28/2023] [Accepted: 06/16/2023] [Indexed: 06/21/2023]
Abstract
Autophagy is a highly conserved biological process that has evolved across evolution. It can be activated by various external stimuli including oxidative stress, amino acid starvation, infection, and hypoxia. Autophagy is the primary mechanism for preserving cellular homeostasis and is implicated in the regulation of metabolism, cell differentiation, tolerance to starvation conditions, and resistance to aging. As a multifunctional protein, DJ-1 is commonly expressed in vivo and is associated with a variety of biological processes. Its most widely studied role is its function as an oxidative stress sensor that inhibits the production of excessive reactive oxygen species (ROS) in the mitochondria and subsequently the cellular damage caused by oxidative stress. In recent years, many studies have identified DJ-1 as another important factor regulating autophagy; it regulates autophagy in various ways, most commonly by regulating the oxidative stress response. In particular, DJ-1-regulated autophagy is involved in cancer progression and plays a key role in alleviating neurodegenerative diseases(NDS) and defective reperfusion diseases. It could serve as a potential target for the regulation of autophagy and participate in disease treatment as a meaningful modality. Therefore, exploring DJ-1-regulated autophagy could provide new avenues for future disease treatment.
Collapse
Affiliation(s)
- Shiyi Liu
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, 330006, PR China; Second Clinical Medical College, Nanchang University, Nanchang, 330006, PR China
| | - Sheng Xu
- Second Clinical Medical College, Nanchang University, Nanchang, 330006, PR China
| | - Song Liu
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, 330006, PR China
| | - Heping Chen
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, 330006, PR China.
| |
Collapse
|
12
|
Shi L, Tao Z, Zheng L, Yang J, Hu X, Scott K, de Kloet A, Krause E, Collins JF, Cheng Z. FoxO1 regulates adipose transdifferentiation and iron influx by mediating Tgfβ1 signaling pathway. Redox Biol 2023; 63:102727. [PMID: 37156218 DOI: 10.1016/j.redox.2023.102727] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 04/24/2023] [Accepted: 04/30/2023] [Indexed: 05/10/2023] Open
Abstract
Adipose plasticity is critical for metabolic homeostasis. Adipocyte transdifferentiation plays an important role in adipose plasticity, but the molecular mechanism of transdifferentiation remains incompletely understood. Here we show that the transcription factor FoxO1 regulates adipose transdifferentiation by mediating Tgfβ1 signaling pathway. Tgfβ1 treatment induced whitening phenotype in beige adipocytes, reducing UCP1 and mitochondrial capacity and enlarging lipid droplets. Deletion of adipose FoxO1 (adO1KO) dampened Tgfβ1 signaling by downregulating Tgfbr2 and Smad3 and induced browning of adipose tissue in mice, increasing UCP1 and mitochondrial content and activating metabolic pathways. Silencing FoxO1 also abolished the whitening effect of Tgfβ1 on beige adipocytes. The adO1KO mice exhibited a significantly higher energy expenditure, lower fat mass, and smaller adipocytes than the control mice. The browning phenotype in adO1KO mice was associated with an increased iron content in adipose tissue, concurrent with upregulation of proteins that facilitate iron uptake (DMT1 and TfR1) and iron import into mitochondria (Mfrn1). Analysis of hepatic and serum iron along with hepatic iron-regulatory proteins (ferritin and ferroportin) in the adO1KO mice revealed an adipose tissue-liver crosstalk that meets the increased iron requirement for adipose browning. The FoxO1-Tgfβ1 signaling cascade also underlay adipose browning induced by β3-AR agonist CL316243. Our study provides the first evidence of a FoxO1-Tgfβ1 axis in the regulation of adipose browning-whitening transdifferentiation and iron influx, which sheds light on the compromised adipose plasticity in conditions of dysregulated FoxO1 and Tgfβ1 signaling.
Collapse
Affiliation(s)
- Limin Shi
- Food Science and Human Nutrition Department, University of Florida, Gainesville, FL, 32611, USA; Interdisciplinary Nutritional Sciences Doctoral Program, Center for Nutritional Sciences, University of Florida, Gainesville, FL, 32611, USA; Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, FL, 32610, USA
| | - Zhipeng Tao
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, VA, 24061, USA; Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Louise Zheng
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Jinying Yang
- Food Science and Human Nutrition Department, University of Florida, Gainesville, FL, 32611, USA; Interdisciplinary Nutritional Sciences Doctoral Program, Center for Nutritional Sciences, University of Florida, Gainesville, FL, 32611, USA
| | - Xinran Hu
- Food Science and Human Nutrition Department, University of Florida, Gainesville, FL, 32611, USA
| | - Karen Scott
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, FL, 32610, USA; Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL32610, USA
| | - Annette de Kloet
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, FL, 32610, USA; Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL, 32610, USA
| | - Eric Krause
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, FL, 32610, USA; Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL32610, USA
| | - James F Collins
- Food Science and Human Nutrition Department, University of Florida, Gainesville, FL, 32611, USA; Interdisciplinary Nutritional Sciences Doctoral Program, Center for Nutritional Sciences, University of Florida, Gainesville, FL, 32611, USA
| | - Zhiyong Cheng
- Food Science and Human Nutrition Department, University of Florida, Gainesville, FL, 32611, USA; Interdisciplinary Nutritional Sciences Doctoral Program, Center for Nutritional Sciences, University of Florida, Gainesville, FL, 32611, USA; Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, FL, 32610, USA; Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, VA, 24061, USA.
| |
Collapse
|
13
|
Tao Z, Cheng Z. Hormonal regulation of metabolism-recent lessons learned from insulin and estrogen. Clin Sci (Lond) 2023; 137:415-434. [PMID: 36942499 PMCID: PMC10031253 DOI: 10.1042/cs20210519] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 02/24/2023] [Accepted: 03/03/2023] [Indexed: 03/23/2023]
Abstract
Hormonal signaling plays key roles in tissue and metabolic homeostasis. Accumulated evidence has revealed a great deal of insulin and estrogen signaling pathways and their interplays in the regulation of mitochondrial, cellular remodeling, and macronutrient metabolism. Insulin signaling regulates nutrient and mitochondrial metabolism by targeting the IRS-PI3K-Akt-FoxOs signaling cascade and PGC1α. Estrogen signaling fine-tunes protein turnover and mitochondrial metabolism through its receptors (ERα, ERβ, and GPER). Insulin and estrogen signaling converge on Sirt1, mTOR, and PI3K in the joint regulation of autophagy and mitochondrial metabolism. Dysregulated insulin and estrogen signaling lead to metabolic diseases. This article reviews the up-to-date evidence that depicts the pathways of insulin signaling and estrogen-ER signaling in the regulation of metabolism. In addition, we discuss the cross-talk between estrogen signaling and insulin signaling via Sirt1, mTOR, and PI3K, as well as new therapeutic options such as agonists of GLP1 receptor, GIP receptor, and β3-AR. Mapping the molecular pathways of insulin signaling, estrogen signaling, and their interplays advances our understanding of metabolism and discovery of new therapeutic options for metabolic disorders.
Collapse
Affiliation(s)
- Zhipeng Tao
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, U.S.A
| | - Zhiyong Cheng
- Department of Food Science and Human Nutrition, University of Florida, Gainesville, Florida, U.S.A
| |
Collapse
|
14
|
Kuang M, Yang R, Xie Q, Peng N, Lu S, Xie G, Zhang S, Zou Y. The role of predicted lean body mass and fat mass in non-alcoholic fatty liver disease in both sexes: Results from a secondary analysis of the NAGALA study. Front Nutr 2023; 10:1103665. [PMID: 36742435 PMCID: PMC9894318 DOI: 10.3389/fnut.2023.1103665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 01/03/2023] [Indexed: 01/20/2023] Open
Abstract
Objective High body mass index (BMI) is an important risk factor for non-alcoholic fatty liver disease (NAFLD). However, the association of body composition such as fat mass (FM) and lean body mass (LBM) with NAFLD has not been adequately studied. The purpose of this study was to clarify the contribution of body composition FM and LBM to NAFLD. Methods We analyzed data from 7,411 men and 6,840 women in the NAGALA cohort study. LBM and FM were estimated for all subjects using validated anthropometric prediction equations previously developed from the National Health and Nutrition Examination Survey (NHANES). Using multiple logistic regression and restricted cubic spline (RCS) to analyze the association and the dose-response curve of predicted LBM and FM with NAFLD in both sexes. Results The prevalence of NAFLD in man and woman subjects was 27.37 and 6.99%, respectively. Predicted FM was positively and linearly associated with NAFLD in both sexes, with each 1 kg increase in predicted FM associated with a 27 and 40% increased risk of NAFLD in men and women, respectively. In contrast, predicted LBM was negatively associated with NAFLD in both sexes, with each 1 kg increase in predicted LBM reducing the risk of NAFLD by 4 and 19% in men and women, respectively. In addition, according to the RCS curve, the risk of NAFLD did not change in men when the predicted LBM was between 47 and 52 kg, and there seemed to be a saturation effect; further, the threshold value of the saturation effect was calculated to be about 52.08 kg by two-piecewise logistic regression, and the protective effect on NAFLD would be significantly enhanced when the man predicted LBM was greater than 52.08 kg. Conclusion The current findings suggested that body composition LBM and FM had opposite associations with NAFLD in both sexes, with higher LBM associated with a lower risk of NAFLD and higher FM increasing the risk of NAFLD, especially in women.
Collapse
Affiliation(s)
- Maobin Kuang
- Department of Cardiology, Jiangxi Provincial People’s Hospital, Medical College of Nanchang University, Nanchang, Jiangxi, China,Jiangxi Cardiovascular Research Institute, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Ruijuan Yang
- Department of Cardiology, Jiangxi Provincial People’s Hospital, Medical College of Nanchang University, Nanchang, Jiangxi, China,Department of Endocrinology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Qiyang Xie
- Department of Cardiology, Jiangxi Provincial People’s Hospital, Medical College of Nanchang University, Nanchang, Jiangxi, China,Jiangxi Cardiovascular Research Institute, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Nan Peng
- Department of Cardiology, Jiangxi Provincial People’s Hospital, Medical College of Nanchang University, Nanchang, Jiangxi, China
| | - Song Lu
- Department of Cardiology, Jiangxi Provincial People’s Hospital, Medical College of Nanchang University, Nanchang, Jiangxi, China
| | - Guobo Xie
- Department of Cardiology, Jiangxi Provincial People’s Hospital, Medical College of Nanchang University, Nanchang, Jiangxi, China
| | - Shuhua Zhang
- Jiangxi Cardiovascular Research Institute, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China,*Correspondence: Shuhua Zhang,
| | - Yang Zou
- Jiangxi Cardiovascular Research Institute, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China,Yang Zou,
| |
Collapse
|
15
|
Wu R, Feng S, Li F, Shu G, Wang L, Gao P, Zhu X, Zhu C, Wang S, Jiang Q. Transcriptional and post-transcriptional control of autophagy and adipogenesis by YBX1. Cell Death Dis 2023; 14:29. [PMID: 36642732 PMCID: PMC9841012 DOI: 10.1038/s41419-023-05564-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 01/03/2023] [Accepted: 01/06/2023] [Indexed: 01/17/2023]
Abstract
Obesity is strongly associated with metabolic diseases, which have become a global health problem. Exploring the underlying mechanism of adipogenesis is crucial for the treatment of excess white fat. Oncogene YBX1 is a multifunctional DNA- and RNA-binding protein that regulates brown adipogenesis. However, the role of YBX1 in white adipogenesis and adipose tissue expansion remains unknown. Here, we showed that YBX1 deficiency inhibited murine and porcine adipocyte differentiation. YBX1 positively regulated adipogenesis through promoting ULK1- and ULK2-mediated autophagy. Mechanistically, we identified YBX1 serves as a 5-methylcytosine (m5C)-binding protein directly targeting m5C-containing Ulk1 mRNA by using RNA immunoprecipitation. RNA decay assay further proved that YBX1 upregulated ULK1 expression though stabilizing its mRNA. Meanwhile, YBX1 promoted Ulk2 transcription and expression as a transcription factor, thereby enhancing autophagy and adipogenesis. Importantly, YBX1 overexpression in white fat enhanced ULK1/ULK2-mediated autophagy and promoted adipose tissue expansion in mice. Collectively, these findings unveil the post-transcriptional and transcriptional mechanism and functional importance of YBX1 in autophagy and adipogenesis regulation, providing an attractive molecular target for therapies of obesity and metabolic diseases.
Collapse
Affiliation(s)
- Ruifan Wu
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Shengchun Feng
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Fan Li
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Gang Shu
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Lina Wang
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Ping Gao
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Xiaotong Zhu
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Canjun Zhu
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Songbo Wang
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Qingyan Jiang
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
16
|
Abstract
The transcription factor FoxO1 (forkhead box O1) regulates genes that are involved in development, metabolism, cellular innovation, longevity, and stress responses. Assessment of FoxO1 activity is therefore critical to understand the regulatory network of this transcription factor. FoxO1 transactivation activity relies on its ability to bind to the promoters of target genes, which is controlled by posttranslational modifications (e.g., dephosphorylation or phosphorylation) that may promote nuclear translocation or exclusion of FoxO1. In this chapter we describe the protocols for FoxO1 activity assessment using Western blotting analysis of the posttranslational modification of FoxO1 in whole cell lysates and ELISA of DNA binding activity of FoxO1 in nuclear extracts.
Collapse
Affiliation(s)
- Limin Shi
- Food Science and Human Nutrition Department, University of Florida, Gainesville, FL, USA
| | - Zhipeng Tao
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, VA, USA
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Zhiyong Cheng
- Food Science and Human Nutrition Department, University of Florida, Gainesville, FL, USA.
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, VA, USA.
| |
Collapse
|
17
|
Role of FOXO3a Transcription Factor in the Regulation of Liver Oxidative Injury. Antioxidants (Basel) 2022; 11:antiox11122478. [PMID: 36552685 PMCID: PMC9774119 DOI: 10.3390/antiox11122478] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 12/08/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Oxidative stress has been identified as a key mechanism in liver damage caused by various chemicals. The transcription factor FOXO3a has emerged as a critical regulator of redox imbalance. Multiple post-translational changes and epigenetic processes closely regulate the activity of FOXO3a, resulting in synergistic or competing impacts on its subcellular localization, stability, protein-protein interactions, DNA binding affinity, and transcriptional programs. Depending on the chemical nature and subcellular context, the oxidative-stress-mediated activation of FOXO3a can induce multiple transcriptional programs that play crucial roles in oxidative injury to the liver by chemicals. Here, we mainly review the role of FOXO3a in coordinating programs of genes that are essential for cellular homeostasis, with an emphasis on exploring the regulatory mechanisms and potential application of FOXO3a as a therapeutic target to prevent and treat liver oxidative injury.
Collapse
|
18
|
He Q, Liu S, Feng Z, Li T, Chu J, Hu W, Chen X, Han Q, Sun N, Sun H, Shen Y. Association between the visceral adiposity index and risks of all-cause and cause-specific mortalities in a large cohort: Findings from the UK biobank. Nutr Metab Cardiovasc Dis 2022; 32:2204-2215. [PMID: 35843793 DOI: 10.1016/j.numecd.2022.05.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 05/10/2022] [Accepted: 05/25/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND AND AIMS The visceral adiposity index (VAI) has been recently established as a measure of visceral fat distribution and is shown to be associated with a wide range of adverse health events. However, the precise associations between the VAI score and all-cause and cause-specific mortalities in the general population remain undetermined. METHODS AND RESULTS In this large-scale prospective epidemiological study, 357,457 participants (aged 38-73 years) were selected from the UK Biobank. We used Cox competing risk regression models to estimate the association between the VAI score and all-cause, cardiovascular disease (CVD), cancer, and other mortalities. The VAI score was significantly correlated with an increased risk of all-cause mortality (hazard ratio [HR], 1.200; 95% confidence interval [CI], 1.148-1.255; P < 0.0001), cancer mortality (HR, 1.224; 95% CI, 1.150-1.303; P < 0.0001), CVD mortality (HR, 1.459; 95% CI, 1.148-1.255; P < 0.0001), and other mortalities (HR, 1.200; 95% CI, 1.148-1.255; P < 0.0001) after adjusting for a series of confounders. In addition, the subgroup analyses showed that HRs were significantly higher in participants who were male, aged below 65 years, and body mass index less than 25. CONCLUSION In summary, VAI was positively associated with an increased risk of all-cause and cause-specific mortalities in a nationwide, well-characterised population identified in a UK Biobank. The VAI score might be a complementary traditional predictive indicator for evaluating the risk of adverse health events in the population of Western adults aged 38 years and older.
Collapse
Affiliation(s)
- Qida He
- Department of Epidemiology and Biostatistics, School of Public Health, Medical College of Soochow University, 199 Renai Road, Suzhou, 215123, PR China
| | - Siyuan Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Medical College of Soochow University, 199 Renai Road, Suzhou, 215123, PR China
| | - Zhaolong Feng
- Department of Epidemiology and Biostatistics, School of Public Health, Medical College of Soochow University, 199 Renai Road, Suzhou, 215123, PR China
| | - Tongxing Li
- Department of Epidemiology and Biostatistics, School of Public Health, Medical College of Soochow University, 199 Renai Road, Suzhou, 215123, PR China
| | - Jiadong Chu
- Department of Epidemiology and Biostatistics, School of Public Health, Medical College of Soochow University, 199 Renai Road, Suzhou, 215123, PR China
| | - Wei Hu
- Department of Epidemiology and Biostatistics, School of Public Health, Medical College of Soochow University, 199 Renai Road, Suzhou, 215123, PR China
| | - Xuanli Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Medical College of Soochow University, 199 Renai Road, Suzhou, 215123, PR China
| | - Qiang Han
- Department of Epidemiology and Biostatistics, School of Public Health, Medical College of Soochow University, 199 Renai Road, Suzhou, 215123, PR China
| | - Na Sun
- Department of Epidemiology and Biostatistics, School of Public Health, Medical College of Soochow University, 199 Renai Road, Suzhou, 215123, PR China
| | - Hongpeng Sun
- Department of Epidemiology and Biostatistics, School of Public Health, Medical College of Soochow University, 199 Renai Road, Suzhou, 215123, PR China.
| | - Yueping Shen
- Department of Epidemiology and Biostatistics, School of Public Health, Medical College of Soochow University, 199 Renai Road, Suzhou, 215123, PR China.
| |
Collapse
|
19
|
FoxO transcription factors in mitochondrial homeostasis. Biochem J 2022; 479:525-536. [PMID: 35195252 PMCID: PMC8883485 DOI: 10.1042/bcj20210777] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 02/10/2022] [Accepted: 02/14/2022] [Indexed: 12/24/2022]
Abstract
Mitochondria play essential roles in cellular energetics, biosynthesis, and signaling transduction. Dysfunctional mitochondria have been implicated in different diseases such as obesity, diabetes, cardiovascular disease, nonalcoholic fatty liver disease, neurodegenerative disease, and cancer. Mitochondrial homeostasis is controlled by a triad of mitochondrial biogenesis, dynamics (fusion and fission), and autophagy (mitophagy). Studies have underscored FoxO transcription factors as key mitochondrial regulators. Specifically, FoxOs regulate mitochondrial biogenesis by dampening NRF1-Tfam and c-Myc-Tfam cascades directly, and inhibiting NAD-Sirt1-Pgc1α cascade indirectly by inducing Hmox1 or repressing Fxn and Urod. In addition, FoxOs mediate mitochondrial fusion (via Mfn1 and Mfn2) and fission (via Drp1, Fis1, and MIEF2), during which FoxOs elicit regulatory mechanisms at transcriptional, posttranscriptional (e.g. via miR-484/Fis1), and posttranslational (e.g. via Bnip3-calcineurin mediated Drp1 dephosphorylation) levels. Furthermore, FoxOs control mitochondrial autophagy in the stages of autophagosome formation and maturation (e.g. initiation, nucleation, and elongation), mitochondria connected to and engulfed by autophagosome (e.g. via PINK1 and Bnip3 pathways), and autophagosome-lysosome fusion to form autolysosome for cargo degradation (e.g. via Tfeb and cathepsin proteins). This article provides an up-to-date view of FoxOs regulating mitochondrial homeostasis and discusses the potential of targeting FoxOs for therapeutics.
Collapse
|
20
|
Mechanisms of autophagic responses to altered nutritional status. J Nutr Biochem 2022; 103:108955. [PMID: 35134508 DOI: 10.1016/j.jnutbio.2022.108955] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 10/09/2021] [Accepted: 01/05/2022] [Indexed: 01/18/2023]
|
21
|
Wang P, Huang Y, Zhao Y, Zhou Y, Wang H, Zhang L, Shi H, Sze-Yin Leung K, Zhang Y. Organic UV filters mixture exposure and childhood adiposity: A prospective follow-up study in China. ENVIRONMENT INTERNATIONAL 2022; 158:106912. [PMID: 34619533 DOI: 10.1016/j.envint.2021.106912] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND UV filters are emerging contaminants with endocrine disrupting effects, but little is known about their health effects, especially for children. OBJECTIVE To assess the association between multiple organic UV filters exposure and adiposity measures and by gender in peripubertal children. METHODS This prospective follow-up study included 327 children aged 7-15 years old. Urinary organic UV filters including benzophenone derivatives (BP-2, BP-3), octyl dimethyl para-aminobenzoic acid (OD-PABA), ethylhexyl methoxycinnamate (EHMC) and its metabolite (4-MCA and 4'-MAP) were quantified. Six adiposity biometrics including height, weight, waist and hip circumferences, and triceps and subscapular skinfold thickness were measured with 1.5-year duration. The Bayesian kernel machine regression method was used to estimate the associations of UV filters mixture with adiposity measurements, and longitudinal analyses were then considered to further evaluate the associations between individual UV filters and trajectories of growth development using linear mixed models or generalized linear mixed models. RESULTS Exposure to mixture of UV filters was negatively associated with most adiposity measurements, with a reduction of 1.399 kg/m2 (95% CI: -2.246 to -0.551 kg/m2) in BMI, 0.674 (95% CI: -1.045 to -0.304) in BMI z-score, 0.033 BF% (95% CI: -0.053 to -0.013), and 2.301 mm (95% CI: -3.823 to -0.78) in subscapular skinfold thickness at baseline, comparing the 75th percentile to the 25th level of UV filters mixture exposure. Consistent associations were found at follow-up. Both baseline and follow-up results suggested that EHMC was identified as the most important contributor to lower adiposity measurements, which was also confirmed by linear mixed models in longitudinal analyses. No significant effects were found in girls. CONCLUSION This study found that childhood organic UV filters exposure was negatively associated with adiposity measures in peripubertal boys, but not girls.
Collapse
Affiliation(s)
- Pengpeng Wang
- Key Lab of Health Technology Assessment, National Health Commission of the People's Republic of China (Fudan University), China; Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai 200032, China
| | - Yanran Huang
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tong, Hong Kong Special Administrative Region
| | - Yingya Zhao
- Key Lab of Health Technology Assessment, National Health Commission of the People's Republic of China (Fudan University), China; Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai 200032, China
| | - Yuhan Zhou
- Key Lab of Health Technology Assessment, National Health Commission of the People's Republic of China (Fudan University), China; Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai 200032, China
| | - Hang Wang
- Key Lab of Health Technology Assessment, National Health Commission of the People's Republic of China (Fudan University), China; Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai 200032, China
| | - Liyi Zhang
- Key Lab of Health Technology Assessment, National Health Commission of the People's Republic of China (Fudan University), China; Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai 200032, China
| | - Huijing Shi
- Key Lab of Health Technology Assessment, National Health Commission of the People's Republic of China (Fudan University), China; Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai 200032, China
| | - Kelvin Sze-Yin Leung
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tong, Hong Kong Special Administrative Region; Institute of Research and Continuing Education, Shenzhen Virtual University Park, Shenzhen, China
| | - Yunhui Zhang
- Key Lab of Health Technology Assessment, National Health Commission of the People's Republic of China (Fudan University), China; Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai 200032, China.
| |
Collapse
|
22
|
Bjune JI, Strømland PP, Jersin RÅ, Mellgren G, Dankel SN. Metabolic and Epigenetic Regulation by Estrogen in Adipocytes. Front Endocrinol (Lausanne) 2022; 13:828780. [PMID: 35273571 PMCID: PMC8901598 DOI: 10.3389/fendo.2022.828780] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 01/24/2022] [Indexed: 12/12/2022] Open
Abstract
Sex hormones contribute to differences between males and females in body fat distribution and associated disease risk. Higher concentrations of estrogens are associated with a more gynoid body shape and with more fat storage on hips and thighs rather than in visceral depots. Estrogen-mediated protection against visceral adiposity is shown in post-menopausal women with lower levels of estrogens and the reduction in central body fat observed after treatment with hormone-replacement therapy. Estrogen exerts its physiological effects via the estrogen receptors (ERα, ERβ and GPR30) in target cells, including adipocytes. Studies in mice indicate that estrogen protects against adipose inflammation and fibrosis also before the onset of obesity. The mechanisms involved in estrogen-dependent body fat distribution are incompletely understood, but involve, e.g., increased mTOR signaling and suppression of autophagy and adipogenesis/lipid storage. Estrogen plays a key role in epigenetic regulation of adipogenic genes by interacting with enzymes that remodel DNA methylation and histone tail post-translational modifications. However, more studies are needed to map the differential epigenetic effects of ER in different adipocyte subtypes, including those in subcutaneous and visceral adipose tissues. We here review recent discoveries of ER-mediated transcriptional and epigenetic regulation in adipocytes, which may explain sexual dimorphisms in body fat distribution and obesity-related disease risk.
Collapse
Affiliation(s)
- Jan-Inge Bjune
- Hormone Laboratory, Department of Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Pouda Panahandeh Strømland
- Hormone Laboratory, Department of Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
| | - Regine Åsen Jersin
- Hormone Laboratory, Department of Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Gunnar Mellgren
- Hormone Laboratory, Department of Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Simon Nitter Dankel
- Hormone Laboratory, Department of Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
- *Correspondence: Simon Nitter Dankel,
| |
Collapse
|
23
|
Kim JY, Park EJ, Kim SM, Lee HJ. Optimization of adipogenic differentiation conditions for canine adipose-derived stem cells. J Vet Sci 2021; 22:e53. [PMID: 34170094 PMCID: PMC8318799 DOI: 10.4142/jvs.2021.22.e53] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/24/2021] [Accepted: 06/04/2021] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Canine adipose-derived stem cells (cADSCs) exhibit various differentiation properties and are isolated from the canine subcutaneous fat. Although cADSCs are valuable as tools for research on adipogenic differentiation, studies focusing on adipogenic differentiation methods and the underlying mechanisms are still lacking. OBJECTIVES In this study, we aimed to establish an optimal method for adipogenic differentiation conditions of cADSCs and evaluate the role of peroxisome proliferator-activated receptor gamma (PPARγ) and estrogen receptor (ER) signaling in the adipogenic differentiation. METHODS To induce adipogenic differentiation of cADSCs, 3 different adipogenic medium conditions, MDI, DRI, and MDRI, using 3-isobutyl-1-methylxanthine (M), dexamethasone (D), insulin (I), and rosiglitazone (R) were tested. RESULTS MDRI, addition of PPARγ agonist rosiglitazone to MDI, was the most significantly facilitated cADSC into adipocyte. GW9662, an antagonist of PPARγ, significantly reduced adipogenic differentiation induced by rosiglitazone. Adipogenic differentiation was also stimulated when 17β-estradiol was added to MDI and DRI, and this stimulation was inhibited by the ER antagonist ICI182,780. CONCLUSIONS Taken together, our results suggest that PPARγ and ER signaling are related to the adipogenic differentiation of cADSCs. This study could provide basic information for future research on obesity or anti-obesity mechanisms in dogs.
Collapse
Affiliation(s)
- Jong Yeon Kim
- Department of Food and Nutrition, College of BioNano Technology, Gachon University, Seongnam 13120, Korea
| | - Eun Jung Park
- Department of Food and Nutrition, College of BioNano Technology, Gachon University, Seongnam 13120, Korea.,Institute for Aging and Clinical Nutrition Research, Gachon University, Seongnam 13120, Korea
| | - Sung Min Kim
- Department of Food and Nutrition, College of BioNano Technology, Gachon University, Seongnam 13120, Korea.,Institute for Aging and Clinical Nutrition Research, Gachon University, Seongnam 13120, Korea
| | - Hae Jeung Lee
- Department of Food and Nutrition, College of BioNano Technology, Gachon University, Seongnam 13120, Korea.,Institute for Aging and Clinical Nutrition Research, Gachon University, Seongnam 13120, Korea.
| |
Collapse
|
24
|
Bisphenols and the Development of Type 2 Diabetes: The Role of the Skeletal Muscle and Adipose Tissue. ENVIRONMENTS 2021. [DOI: 10.3390/environments8040035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Bisphenol A (BPA) and bisphenol S (BPS) are environmental contaminants that have been associated with the development of insulin resistance and type 2 diabetes (T2D). Two organs that are often implicated in the development of insulin resistance are the skeletal muscle and the adipose tissue, however, seldom studies have investigated the effects of bisphenols on their metabolism. In this review we discuss metabolic perturbations that occur in both the skeletal muscle and adipose tissue affected with insulin resistance, and how exposure to BPA or BPS has been linked to these changes. Furthermore, we highlight the possible effects of BPA on the cross-talk between the skeletal muscle and adipose tissue.
Collapse
|
25
|
Sirt1 coordinates with ERα to regulate autophagy and adiposity. Cell Death Discov 2021; 7:53. [PMID: 33723227 PMCID: PMC7960718 DOI: 10.1038/s41420-021-00438-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/25/2021] [Accepted: 02/14/2021] [Indexed: 12/15/2022] Open
Abstract
Sex difference in adiposity has long been recognized but the mechanism remains incompletely understood. Previous studies suggested that adiposity was regulated by autophagy in response to energy status change. Here, we show that the energy sensor Sirt1 mediates sex difference in adiposity by regulating autophagy and adipogenesis in partnership with estrogen receptor α (ERα). Autophagy and adipogenesis were suppressed by Sirt1 activation or overexpression, which was associated with reduced sex difference in adiposity. Mechanistically, Sirt1 deacetylated and activated AKT and STAT3, resulting in suppression of autophagy and adipogenesis via mTOR-ULK1 and p55 cascades. ERα induced Sirt1 expression and inhibited autophagy in adipocytes, while silencing Sirt1 reversed the effects of ERα on autophagy and promoted adipogenesis. Moreover, Sirt1 deacetylated ERα, which constituted a positive feedback loop in the regulation of autophagy and adiposity. Our results revealed a new mechanism of Sirt1 regulating autophagy in adipocytes and shed light on sex difference in adiposity.
Collapse
|
26
|
Gender difference in the association of chronic kidney disease with visceral adiposity index and lipid accumulation product index in Korean adults: Korean National Health and Nutrition Examination Survey. Int Urol Nephrol 2021; 53:1417-1425. [PMID: 33389514 DOI: 10.1007/s11255-020-02735-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Accepted: 12/02/2020] [Indexed: 10/22/2022]
Abstract
BACKGROUND This study was conducted to assess gender difference in the association of chronic kidney disease (CKD) with visceral adiposity index (VAI) and lipid accumulation product index (LAP) in Korean adults. MATERIALS AND METHODS The study was performed using data from the 2015 Korean National Health and Nutrition Examination Survey and included 4947 adults aged 20 years or older. RESULTS There were several key findings in the present study. First, overall, after adjusting for related variables (including age), CKD (estimated glomerular filtration rate < 60 ml/min/1.73 m2) was positively associated with the quartiles of VAI and LAP. Second, when analyzed separately for gender, the results in men and women were different. After adjusting for the related variables (except age), CKD was positively associated with the quartiles of VAI and LAP in both men and women. After further adjustment for age, CKD was still positively associated with the quartiles of VAI and LAP in men. However, in women, the relationships between CKD and the quartiles of VAI and LAP were no longer significant. CONCLUSIONS The VAI and LAP index were positively associated with CKD in Korean men but not in women.
Collapse
|
27
|
Ohlsson C, Gustafsson KL, Farman HH, Henning P, Lionikaite V, Movérare-Skrtic S, Sjögren K, Törnqvist AE, Andersson A, Islander U, Bernardi AI, Poutanen M, Chambon P, Lagerquist MK. Phosphorylation site S122 in estrogen receptor α has a tissue-dependent role in female mice. FASEB J 2020; 34:15991-16002. [PMID: 33067917 DOI: 10.1096/fj.201901376rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 09/10/2020] [Accepted: 09/21/2020] [Indexed: 12/11/2022]
Abstract
Estrogen treatment increases bone mass and reduces fat mass but is associated with adverse effects in postmenopausal women. Knowledge regarding tissue-specific estrogen signaling is important to aid the development of new tissue-specific treatments. We hypothesized that the posttranslational modification phosphorylation in estrogen receptor alpha (ERα) may modulate ERα activity in a tissue-dependent manner. Phosphorylation of site S122 in ERα has been shown in vitro to affect ERα activity, but the tissue-specific role in vivo is unknown. We herein developed and phenotyped a novel mouse model with a point mutation at the phosphorylation site 122 in ERα (S122A). Female S122A mice had increased fat mass and serum insulin levels but unchanged serum sex steroid levels, uterus weight, bone mass, thymus weight, and lymphocyte maturation compared to WT mice. In conclusion, phosphorylation site S122 in ERα has a tissue-dependent role with an impact specifically on fat mass in female mice. This study is the first to demonstrate in vivo that a phosphorylation site in a transactivation domain in a nuclear steroid receptor modulates the receptor activity in a tissue-dependent manner.
Collapse
Affiliation(s)
- Claes Ohlsson
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Karin L Gustafsson
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Helen H Farman
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Petra Henning
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Vikte Lionikaite
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Sofia Movérare-Skrtic
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Klara Sjögren
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Anna E Törnqvist
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Annica Andersson
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Ulrika Islander
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Angelina I Bernardi
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Matti Poutanen
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.,Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Pierre Chambon
- Institut de Génétique et de Biologie Moléculaire et Cellulaire Centre National de la Recherche Scientifique, National de la Sante et de la Recherche Medicale, ULP, Collège de France, Illkirch-Strasbourg, France
| | - Marie K Lagerquist
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
28
|
Bertocchi I, Oberto A, Longo A, Palanza P, Eva C. Conditional inactivation of Npy1r gene in mice induces sex-related differences of metabolic and behavioral functions. Horm Behav 2020; 125:104824. [PMID: 32755609 DOI: 10.1016/j.yhbeh.2020.104824] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 07/15/2020] [Accepted: 07/23/2020] [Indexed: 02/07/2023]
Abstract
Sex hormone-driven differences in gene expression have been identified in experimental animals, highlighting brain neuronal populations implicated in dimorphism of metabolic and behavioral functions. Neuropeptide Y-Y1 receptor (NPY-Y1R) system is sexually dimorphic and sensitive to gonadal steroids. In the present study we compared the phenotype of male and female conditional knockout mice (Npy1rrfb mice), carrying the inactivation of Npy1r gene in excitatory neurons of the brain limbic system. Compared to their male control (Npy1r2lox) littermates, male Npy1rrfb mice exhibited hyperactivation of the hypothalamic-pituitary-adrenal (HPA) axis that is associated with anxiety and executive dysfunction, reduced body weight growth, after-fasting refeeding, white adipose tissue (WAT) mass and plasma leptin levels. Conversely, female Npy1rrfb mice displayed an anxious-like behavior but no differences in HPA axis activity, executive function and body weight, compared to control females. Moreover, conditional inactivation of Npy1r gene induced an increase of subcutaneous and gonadal WAT weight and plasma leptin levels and a compensatory decrease of Agouti-related protein immunoreactivity in the hypothalamic arcuate (ARC) nucleus in females, compared to their respective control littermates. Interestingly, Npy1r mRNA expression was reduced in the ARC and in the paraventricular hypothalamic nuclei of female, but not male mice. These results demonstrated that female mice are resilient to hormonal and metabolic effects of limbic Npy1r gene inactivation, suggesting the existence of an estrogen-dependent relay necessary to ensure the maintenance of the homeostasis, that can be mediated by hypothalamic Y1R.
Collapse
Affiliation(s)
- Ilaria Bertocchi
- Neuroscience Institute of the Cavalieri-Ottolenghi Foundation, 10043 Orbassano, Turin, Italy; Department of Neuroscience, University of Turin, 10126 Turin, Italy; Neuroscience Institute of Turin, Italy
| | - Alessandra Oberto
- Neuroscience Institute of the Cavalieri-Ottolenghi Foundation, 10043 Orbassano, Turin, Italy; Department of Neuroscience, University of Turin, 10126 Turin, Italy; Neuroscience Institute of Turin, Italy
| | - Angela Longo
- Neuroscience Institute of the Cavalieri-Ottolenghi Foundation, 10043 Orbassano, Turin, Italy
| | - Paola Palanza
- Department of Medicine and Surgery, University of Parma, 43100 Parma, Italy
| | - Carola Eva
- Neuroscience Institute of the Cavalieri-Ottolenghi Foundation, 10043 Orbassano, Turin, Italy; Department of Neuroscience, University of Turin, 10126 Turin, Italy; Neuroscience Institute of Turin, Italy.
| |
Collapse
|
29
|
De Martinis M, Sirufo MM, Suppa M, Di Silvestre D, Ginaldi L. Sex and Gender Aspects for Patient Stratification in Allergy Prevention and Treatment. Int J Mol Sci 2020; 21:E1535. [PMID: 32102344 PMCID: PMC7073150 DOI: 10.3390/ijms21041535] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 02/20/2020] [Indexed: 12/11/2022] Open
Abstract
Allergies are rapidly worsening in recent decades, representing the most common immunological diseases. The mechanism of disorders such as asthma, rhinocongiuntivitis, urticaria, atopic dermatitis, food and drug allergies, and anaphylaxis still remain unclear and consequently treatments is mostly still symptomatic and aspecific while developments of new therapies are limited. A growing amount of data in the literature shows us how the prevalence of allergic diseases is different in both sexes and its changes over the course of life. Genes, hormones, environmental and immunological factors affect sex disparities associated with the development and control of allergic diseases, while they more rarely are considered and reported regarding their differences related to social, psychological, cultural, economic, and employment aspects. This review describes the available knowledge on the role of sex and gender in allergies in an attempt to improve the indispensable gender perspective whose potential is still underestimated while it represents a significant turning point in research and the clinic. It will offer insights to stimulate exploration of the many aspects still unknown in this relationship that could ameliorate the preventive, diagnostic, and therapeutic strategies in allergic diseases.
Collapse
Affiliation(s)
- Massimo De Martinis
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (M.M.S.); (D.D.S.); (L.G.)
- Allergy and Clinical Immunology Unit, Center for the diagnosis and treatment of Osteoporosis, AUSL 04 Teramo, Italy
| | - Maria Maddalena Sirufo
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (M.M.S.); (D.D.S.); (L.G.)
- Allergy and Clinical Immunology Unit, Center for the diagnosis and treatment of Osteoporosis, AUSL 04 Teramo, Italy
| | - Mariano Suppa
- Department of Dermatology, Hôpital Erasme, Université Libre de Bruxelles, 1070 Brussels, Belgium;
| | - Daniela Di Silvestre
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (M.M.S.); (D.D.S.); (L.G.)
- Allergy and Clinical Immunology Unit, Center for the diagnosis and treatment of Osteoporosis, AUSL 04 Teramo, Italy
| | - Lia Ginaldi
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (M.M.S.); (D.D.S.); (L.G.)
- Allergy and Clinical Immunology Unit, Center for the diagnosis and treatment of Osteoporosis, AUSL 04 Teramo, Italy
| |
Collapse
|
30
|
Berry A, Marconi M, Musillo C, Chiarotti F, Bellisario V, Matarrese P, Gambardella L, Vona R, Lombardi M, Foglieni C, Cirulli F. Trehalose administration in C57BL/6N old mice affects healthspan improving motor learning and brain anti-oxidant defences in a sex-dependent fashion: a pilot study. Exp Gerontol 2020; 129:110755. [DOI: 10.1016/j.exger.2019.110755] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 09/04/2019] [Accepted: 10/14/2019] [Indexed: 01/31/2023]
|
31
|
Cheng Z. The FoxO-Autophagy Axis in Health and Disease. Trends Endocrinol Metab 2019; 30:658-671. [PMID: 31443842 DOI: 10.1016/j.tem.2019.07.009] [Citation(s) in RCA: 151] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 07/02/2019] [Accepted: 07/08/2019] [Indexed: 12/21/2022]
Abstract
Autophagy controls cellular remodeling and quality control. Dysregulated autophagy has been implicated in several human diseases including obesity, diabetes, cardiovascular disease, neurodegenerative diseases, and cancer. Current evidence has revealed that FoxO (forkhead box class O) transcription factors have a multifaceted role in autophagy regulation and dysregulation. Nuclear FoxOs transactivate genes that control the formation of autophagosomes and their fusion with lysosomes. Independently of transactivation, cytosolic FoxO proteins induce autophagy by directly interacting with autophagy proteins. Autophagy is also controlled by FoxOs through epigenetic mechanisms. Moreover, FoxO proteins can be degraded directly or indirectly by autophagy. Cutting-edge evidence is reviewed that the FoxO-autophagy axis plays a crucial role in health and disease.
Collapse
Affiliation(s)
- Zhiyong Cheng
- Food Science and Human Nutrition Department, The University of Florida, Gainesville, FL 32611, USA.
| |
Collapse
|
32
|
Wei Y, Huang J. Role of estrogen and its receptors mediated-autophagy in cell fate and human diseases. J Steroid Biochem Mol Biol 2019; 191:105380. [PMID: 31078693 DOI: 10.1016/j.jsbmb.2019.105380] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 05/08/2019] [Accepted: 05/09/2019] [Indexed: 12/11/2022]
Abstract
Studies have shown that morbidity of several diseases varies between males and females. This difference likely arises due to sex-related hormones. Estrogen, a primary female sex steroid hormone, plays a critical role in mediating many of the physiological functions like growth, differentiation, metabolism, and cell death. Recently, it has been demonstrated that estrogen mediates autophagy through its receptors (ERs) namely ERα, ERβ, and G-protein coupled estrogen receptor (GPER). However, the specific role of estrogen and its receptors mediated-autophagy in cell fate and human diseases such as cancers, cardiovascular disease and nervous system disease remains unclear. In this review, we comprehensively summarize the complex role of estrogen and its receptors-mediated autophagy in different cell lines and human diseases. In addition, we further discuss the key signaling molecules governing the role of ERs in autophagy. This review will serve as the basis for a proposed model of autophagy constituting a new frontier in estrogen-related human diseases. Here, we discuss the dual role of ERα in classical and non-classical autophagy through B-cell lymphoma 2 (BCL2)-associated athanogene 3 (BAG3). Next, we review the role of ERβ in pro-survival pathways through the promotion of autophagy under stress conditions. We further discuss activation of GPER via estrogen often mediates autophagy or mitophagy suppression, respectively. In summary, we believe that understanding the relationship between estrogen and its receptors mediated-autophagy on cell fate and human diseases will provide insightful knowledge for future therapeutic implications.
Collapse
Affiliation(s)
- Yong Wei
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, Hubei, PR China
| | - Jian Huang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, Hubei, PR China.
| |
Collapse
|
33
|
Garratt M, Leander D, Pifer K, Bower B, Herrera JJ, Day SM, Fiehn O, Brooks SV, Miller RA. 17-α estradiol ameliorates age-associated sarcopenia and improves late-life physical function in male mice but not in females or castrated males. Aging Cell 2019; 18:e12920. [PMID: 30740872 PMCID: PMC6413653 DOI: 10.1111/acel.12920] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 11/28/2018] [Accepted: 12/26/2018] [Indexed: 12/26/2022] Open
Abstract
Pharmacological treatments can extend mouse lifespan, but lifespan effects often differ between sexes. 17-α estradiol (17aE2), a less feminizing structural isomer of 17-β estradiol, produces lifespan extension only in male mice, suggesting a sexually dimorphic mechanism of lifespan regulation. We tested whether these anti-aging effects extend to anatomical and functional aging-important in late-life health-and whether gonadally derived hormones control aging responses to 17aE2 in either sex. While 17aE2 started at 4 months of age diminishes body weight in both sexes during adulthood, in late-life 17aE2-treated mice better maintain body weight. In 17aE2-treated male mice, the higher body weight is associated with heavier skeletal muscles and larger muscle fibers compared with untreated mice during aging, while treated females have heavier subcutaneous fat. Maintenance of skeletal muscle in male mice is associated with improved grip strength and rotarod capacity at 25 months, in addition to higher levels of most amino acids in quadriceps muscle. We further show that sex-specific responses to 17aE2-metabolomic, structural, and functional-are regulated by gonadal hormones in male mice. Castrated males have heavier quadriceps than intact males at 25 months, but do not respond to 17aE2, suggesting 17aE2 promotes an anti-aging skeletal muscle phenotype similar to castration. Finally, 17aE2 treatment benefits can be recapitulated in mice when treatment is started at 16 months, suggesting that 17aE2 may be able to improve aspects of late-life function even when started after middle age.
Collapse
Affiliation(s)
- Michael Garratt
- Department of Pathology; University of Michigan Medical School; Ann Arbor Michigan
| | - Danielle Leander
- Department of Pathology; University of Michigan Medical School; Ann Arbor Michigan
| | - Kaitlyn Pifer
- Department of Pathology; University of Michigan Medical School; Ann Arbor Michigan
| | - Brian Bower
- Department of Pathology; University of Michigan Medical School; Ann Arbor Michigan
| | - Jonathan J. Herrera
- Molecular and Integrative Physiology; University of Michigan; Ann Arbor Michigan
- Internal Medicine; University of Michigan; Ann Arbor Michigan
| | - Sharlene M. Day
- Molecular and Integrative Physiology; University of Michigan; Ann Arbor Michigan
- Internal Medicine; University of Michigan; Ann Arbor Michigan
| | - Oliver Fiehn
- Genome Center; University of California Davis; Davis California
| | - Susan V. Brooks
- Molecular and Integrative Physiology; University of Michigan; Ann Arbor Michigan
- Department of Biomedical Engineering; University of Michigan; Ann Arbor Michigan
| | - Richard A. Miller
- Department of Pathology; University of Michigan Medical School; Ann Arbor Michigan
- University of Michigan Geriatrics Center; Ann Arbor Michigan
| |
Collapse
|
34
|
Frank AP, de Souza Santos R, Palmer BF, Clegg DJ. Determinants of body fat distribution in humans may provide insight about obesity-related health risks. J Lipid Res 2018; 60:1710-1719. [PMID: 30097511 DOI: 10.1194/jlr.r086975] [Citation(s) in RCA: 142] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 08/07/2018] [Indexed: 12/24/2022] Open
Abstract
Obesity increases the risks of developing cardiovascular and metabolic diseases and degrades quality of life, ultimately increasing the risk of death. However, not all forms of obesity are equally dangerous: some individuals, despite higher percentages of body fat, are at less risk for certain chronic obesity-related complications. Many open questions remain about why this occurs. Data suggest that the physical location of fat and the overall health of fat dramatically influence disease risk; for example, higher concentrations of visceral relative to subcutaneous adipose tissue are associated with greater metabolic risks. As such, understanding the determinants of the location and health of adipose tissue can provide insight about the pathological consequences of obesity and can begin to outline targets for novel therapeutic approaches to combat the obesity epidemic. Although age and sex hormones clearly play roles in fat distribution and location, much remains unknown about gene regulation at the level of adipose tissue or how genetic variants regulate fat distribution. In this review, we discuss what is known about the determinants of body fat distribution, and we highlight the important roles of sex hormones, aging, and genetic variation in the determination of body fat distribution and its contribution to obesity-related comorbidities.
Collapse
Affiliation(s)
- Aaron P Frank
- Diabetes, Obesity, and Wellness Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Roberta de Souza Santos
- Diabetes, Obesity, and Wellness Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Biff F Palmer
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX
| | - Deborah J Clegg
- Diabetes, Obesity, and Wellness Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| |
Collapse
|
35
|
Skubic C, Drakulić Ž, Rozman D. Personalized therapy when tackling nonalcoholic fatty liver disease: a focus on sex, genes, and drugs. Expert Opin Drug Metab Toxicol 2018; 14:831-841. [PMID: 29969922 DOI: 10.1080/17425255.2018.1492552] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Nonalcoholic fatty liver disease (NAFLD) is the most frequent liver disease in the world. It describes a term for a group of hepatic diseases including steatosis, fibrosis, and cirrhosis that can finally lead to hepatocellular carcinoma. There are many factors influencing NAFLD initiation and progression, such as obesity, dyslipidemia, insulin resistance, genetic factors, and hormonal changes. However, there is also lean-NAFLD which is not associated with obesity. NAFLD is considered to be a sexually dimorphic disease. In most cases, men have a higher prevalence for the disease compared to premenopausal women. Areas covered: In this review, we first summarize the NAFLD disease epidemiology, pathology, and diagnosis. We describe NAFLD progression with the focus on sexual and genetic differences for disease development and pharmacological treatment. Personalized treatment for multifactorial NAFLD is discussed in consideration of different factors, including genetics, gender and sex. Expert opinion: The livers of female and male NAFLD patients have different metabolic capacities which influence the metabolism of all drugs applied to such patients. This aspect is not yet sufficiently taken into account. The liver computational models might quicken the pace toward assessing personalized disease progression and treatment options.
Collapse
Affiliation(s)
- Cene Skubic
- a Centre for Functional Genomic and Biochips, Institute of Biochemistry, Faculty of Medicine , University of Ljubljana , Ljubljana , Slovenia
| | - Živa Drakulić
- a Centre for Functional Genomic and Biochips, Institute of Biochemistry, Faculty of Medicine , University of Ljubljana , Ljubljana , Slovenia
| | - Damjana Rozman
- a Centre for Functional Genomic and Biochips, Institute of Biochemistry, Faculty of Medicine , University of Ljubljana , Ljubljana , Slovenia
| |
Collapse
|
36
|
Congdon EE. Sex Differences in Autophagy Contribute to Female Vulnerability in Alzheimer's Disease. Front Neurosci 2018; 12:372. [PMID: 29988365 PMCID: PMC6023994 DOI: 10.3389/fnins.2018.00372] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 05/14/2018] [Indexed: 12/11/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia, with over 5. 4 million cases in the US alone (Alzheimer's Association, 2016). Clinically, AD is defined by the presence of plaques composed of Aβ and neurofibrillary pathology composed of the microtubule associated protein tau. Another key feature is the dysregulation of autophagy at key steps in the pathway. In AD, disrupted autophagy contributes to disease progression through the failure to clear pathological protein aggregates, insulin resistance, and its role in the synthesis of Aβ. Like many psychiatric and neurodegenerative diseases, the risk of developing AD, and disease course are dependent on the sex of the patient. One potential mechanism through which these differences occur, is the effects of sex hormones on autophagy. In women, the loss of hormones with menopause presents both a risk factor for developing AD, and an obvious example of where sex differences in AD can stem from. However, because AD pathology can begin decades before menopause, this does not provide the full answer. We propose that sex-based differences in autophagy regulation during the lifespan contribute to the increased risk of AD, and greater severity of pathology seen in women.
Collapse
Affiliation(s)
- Erin E Congdon
- Neuroscience and Physiology, School of Medicine, New York University, New York City, NY, United States
| |
Collapse
|