1
|
Li N, Webb A, Kennelly J, Sharma R, Whitson BA, Mohler PJ, Hummel JD, Zhao J, Fedorov V. Heart Rate Mystery Unveiled: Sex Differences in Human Sinoatrial Node Genes and Female Tachycardia. Circ Arrhythm Electrophysiol 2025:e013534. [PMID: 40265247 DOI: 10.1161/circep.124.013534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 04/04/2025] [Indexed: 04/24/2025]
Abstract
BACKGROUND Despite over a century of clinical electrocardiographic studies showing that females exhibit a faster resting heart rate (HR), the mechanisms underlying sex differences in HR remain unresolved. Moreover, inappropriate sinus tachycardia primarily affects females, whereas males are at a higher risk for conduction block and atrial fibrillation. We hypothesized that the sexual dimorphism of genes responsible for sinoatrial node (SAN) pacemaking and signaling pathways may contribute to the sex differences in HR and susceptibility to arrhythmias. METHODS Human SAN central pacemaker and right atrial tissue were isolated from nondiseased ex vivo donor hearts. Gene expressions were quantified and validated using the transcriptomic panel and quantitative polymerase chain reaction. Gene set enrichment analysis, Ingenuity Pathway Analysis, and human-specific SAN models were utilized to define regulatory mechanisms and functional impacts of sex-biased gene transcription. RESULTS We identified differentially expressed region- and sex-specific genes, with gene sets enriched in HR regulation (eg, TBX3, HCN1) and metabolism (eg, ADIPOQ, LEP) pathways in female SAN. In contrast, differential genes and gene sets involved in collagen biosynthetic processes, fibrogenesis (eg, EGR1), and immune response (eg, IL6, CXCL8) pathways were enriched in males SAN and right atrial. Ingenuity Pathway Analysis predicted significant roles for TBX3 and estradiol in the sex-specific expression of genes involved in SAN function. Computational simulations showed that the sex-specific SAN differences in If (HCN1) and ICa,L (CACNA1D) can explain the faster HR in females, with females having a lower threshold for inappropriate sinus tachycardia, whereas males are more vulnerable to sinus arrest. CONCLUSIONS The human SAN exhibits region-specific sexual dimorphism in pacemaking gene sets. Higher expression of TBX3 and HCN1 in females may underlie their faster HR and increased susceptibility to inappropriate sinus tachycardia, whereas enriched gene sets related to inflammation and collagen biosynthesis in males may predispose them to conduction impairments and atrial fibrillation risk.
Collapse
Affiliation(s)
- Ning Li
- Department of Physiology and Cell Biology, The Ohio State University College of Medicine Wexner Medical Center, Columbus. (N.L., P.J.M., V.F.)
- Bob and Corrine Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine Wexner Medical Center, Columbus. (N.L., P.J.M., J.D.H., V.F.)
| | - Amy Webb
- Biomedical Informatics, The Ohio State University College of Medicine Wexner Medical Center, Columbus. (A.W.)
| | - James Kennelly
- Auckland Bioengineering Institute, The University of Auckland, New Zealand (J.K., R.S., J.Z.)
| | - Roshan Sharma
- Auckland Bioengineering Institute, The University of Auckland, New Zealand (J.K., R.S., J.Z.)
| | - Bryan A Whitson
- Department of Surgery, Division of Cardiac Surgery, The Ohio State University College of Medicine Wexner Medical Center, Columbus. (B.A.W.)
| | - Peter J Mohler
- Department of Physiology and Cell Biology, The Ohio State University College of Medicine Wexner Medical Center, Columbus. (N.L., P.J.M., V.F.)
- Bob and Corrine Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine Wexner Medical Center, Columbus. (N.L., P.J.M., J.D.H., V.F.)
- Department of Internal Medicine, The Ohio State University College of Medicine Wexner Medical Center, Columbus. (P.J.M., J.D.H.)
| | - John D Hummel
- Bob and Corrine Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine Wexner Medical Center, Columbus. (N.L., P.J.M., J.D.H., V.F.)
- Department of Internal Medicine, The Ohio State University College of Medicine Wexner Medical Center, Columbus. (P.J.M., J.D.H.)
| | - Jichao Zhao
- Auckland Bioengineering Institute, The University of Auckland, New Zealand (J.K., R.S., J.Z.)
| | - Vadim Fedorov
- Department of Physiology and Cell Biology, The Ohio State University College of Medicine Wexner Medical Center, Columbus. (N.L., P.J.M., V.F.)
- Bob and Corrine Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine Wexner Medical Center, Columbus. (N.L., P.J.M., J.D.H., V.F.)
| |
Collapse
|
2
|
Gao Y, Tang X, Liu B, Qiu L. Application of ultrasound for quantitative assessment of body fat mass. Clin Nutr ESPEN 2025; 67:635-644. [PMID: 40204071 DOI: 10.1016/j.clnesp.2025.03.175] [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: 01/16/2025] [Revised: 03/10/2025] [Accepted: 03/26/2025] [Indexed: 04/11/2025]
Abstract
BACKGROUND & AIMS Obesity is a significant health concern associated with various diseases. Accurate measurement of body fat mass (BFM) and local fat thickness (FT) is crucial for health assessment. Ultrasound offers a non-invasive, portable, and cost-effective alternative for measuring FT, but its application for quantitative BFM estimation has not been fully explored. This study aimed to develop and validate a quantitative estimation algorithm for BFM based on local FT measured by ultrasound. METHODS A total of 179 volunteers were randomly divided into modeling and verification groups. BFM was measured using bioelectrical impedance analysis (BIA), and FT was measured at 10 sites throughout the body using ultrasound. In the modeling group, the correlation between FT and BFM at different sites was analyzed, and a BFM estimation algorithm based on FT was developed using multiple linear regression. The accuracy of the estimation equation was validated in the verification group. RESULTS Men had lower BFM than women (P < 0.05). At most sites, the FT of males was less than that of females (P < 0.001). Significant positive correlations were observed between FT at various sites (site 1 to 10) and BFM across all groups (P < 0.01). The estimation algorithm revealed that FT at 4 sites (intra-abdominal, posterior right perinephric, abdominal subcutaneous, and anterior upper arm) contributed to BFM estimation for men, while two additional sites (pre-peritoneal and posterior lower leg) were valuable for women. The R2 for the algorithms was 0.882 for men and 0.907 for women, with the standard error of estimate of 2.04 kg for both. The intraclass correlation coefficient between ultrasound-derived estimated BFM and the BFM measured by BIA in the verification group was 0.848 (P < 0.001). CONCLUSIONS BFM can be quantitatively estimated using a fitting algorithm based on ultrasound-derived local FT.
Collapse
Affiliation(s)
- Yang Gao
- Department of Medical Ultrasound, West China Hospital of Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, Sichuan, China.
| | - Xinyi Tang
- Department of Medical Ultrasound, West China Hospital of Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, Sichuan, China.
| | - Bingjie Liu
- Department of Medical Ultrasound, West China Hospital of Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, Sichuan, China.
| | - Li Qiu
- Department of Medical Ultrasound, West China Hospital of Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, Sichuan, China.
| |
Collapse
|
3
|
Hajsadeghi S, Athar R, Iranpour A, Yahyavi A, Mohebi N, Rahimi AH. The Influence of Metabolic and Bariatric Surgery on Cardiovascular Health in Women: A Comprehensive Study. Obes Surg 2025; 35:1538-1542. [PMID: 39953329 DOI: 10.1007/s11695-024-07661-z] [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: 04/27/2024] [Revised: 12/27/2024] [Accepted: 12/27/2024] [Indexed: 02/17/2025]
Abstract
BACKGROUND Cardiovascular disease (CVD) is the main cause of death in patients with obesity. Bariatric surgery has been shown to promote substantial reduction in carotid intima-media thickness (C-IMT). AIM To comprehensively determine the influence of metabolic and bariatric surgery (MBS) on cardiovascular health in women. METHODS A total of 24 female patients with a body mass index (BMI) > 35 kg/m2 underwent sleeve gastrectomy (SG), Roux-en-Y gastric bypass (RYGB), or one anastomosis gastric bypass (OAGB), a prospective cohort study in women. C-IMT were evaluated at baseline and 24 months post-MBS. RESULTS The results showed a significant reduction in BMI, weight, and C-IMT on the right side 24 months post-MBS (p < 0.001 for BMI and weight and p = 0.004 for C-IMT). There were also significant differences in fasting blood sugar (FBS), triglycerides (TG), total cholesterol, and low-density lipoprotein (LDL) levels. However, high-density lipoprotein (HDL) levels did not show a significant change. CONCLUSION The study underscores the potential of MBS in mitigating cardiovascular risk among women with obesity. However, further research is needed to fully understand the impact of MBS on various parameters. The findings highlight the importance of personalized and gender-specific approaches in the management of obesity and related disease.
Collapse
Affiliation(s)
- Shokoufeh Hajsadeghi
- Research Center for Prevention of Cardiovascular Disease, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran
| | - Rahmatullah Athar
- Department of Surgery, Minimally Invasive Surgery Research Center, Division of Minimally Invasive and Bariatric Surgery, Hazrat-E Fatemeh Hospital Iran University of Medical Sciences, Tehran, Iran
| | - Aida Iranpour
- Research Center for Prevention of Cardiovascular Disease, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran
| | | | - Nafiseh Mohebi
- Neurology Department, Rasool-E Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
4
|
Xu M, Zhou EY, Shi H. Tryptophan and Its Metabolite Serotonin Impact Metabolic and Mental Disorders via the Brain-Gut-Microbiome Axis: A Focus on Sex Differences. Cells 2025; 14:384. [PMID: 40072112 PMCID: PMC11899299 DOI: 10.3390/cells14050384] [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] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 02/27/2025] [Accepted: 03/04/2025] [Indexed: 03/15/2025] Open
Abstract
The crisis of metabolic and mental disorders continues to escalate worldwide. A growing body of research highlights the influence of tryptophan and its metabolites, such as serotonin, beyond their traditional roles in neural signaling. Serotonin acts as a key neurotransmitter within the brain-gut-microbiome axis, a critical bidirectional communication network affecting both metabolism and behavior. Emerging evidence suggests that the gut microbiome regulates brain function and behavior, particularly through microbial influences on tryptophan metabolism and the serotonergic system, both of which are essential for normal functioning. Additionally, sex differences exist in multiple aspects of serotonin-mediated modulation within the brain-gut-microbiome axis, affecting feeding and affective behaviors. This review summarizes the current knowledge from human and animal studies on the influence of tryptophan and its metabolite serotonin on metabolic and behavioral regulation involving the brain and gut microbiome, with a focus on sex differences and the role of sex hormones. We speculate that gut-derived tryptophan and serotonin play essential roles in the pathophysiology that modifies neural circuits, potentially contributing to eating and affective disorders. We propose the gut microbiome as an appealing therapeutic target for metabolic and affective disorders, emphasizing the importance of understanding sex differences in metabolic and behavioral regulation influenced by the brain-gut-microbiome axis. The therapeutic targeting of the gut microbiota and its metabolites may offer a viable strategy for treating serotonin-related disorders, such as eating and affective disorders, with potential differences in treatment efficacy between men and women. This review would promote research on sex differences in metabolic and behavioral regulation impacted by the brain-gut-microbiome axis.
Collapse
Affiliation(s)
- Mengyang Xu
- Program in Cell, Molecular, and Structural Biology, Miami University, Oxford, OH 45056, USA
| | - Ethan Y. Zhou
- Institute for the Environment and Sustainability, Miami University, Oxford, OH 45056, USA
| | - Haifei Shi
- Program in Cell, Molecular, and Structural Biology, Miami University, Oxford, OH 45056, USA
| |
Collapse
|
5
|
Theofilis P, Vlachakis PK, Mantzouranis E, Sakalidis A, Chrysohoou C, Leontsinis I, Lazaros G, Dimitriadis K, Drakopoulou M, Vordoni A, Oikonomou E, Tsioufis K, Tousoulis D. Acute Coronary Syndromes in Women: A Narrative Review of Sex-Specific Characteristics. Angiology 2025; 76:209-224. [PMID: 37995282 DOI: 10.1177/00033197231218331] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2023]
Abstract
Acute coronary syndromes (ACSs) encompass a spectrum of life-threatening cardiovascular conditions, including unstable angina (UA) and myocardial infarction. While significant progress has been made in the understanding and management of ACS over the years, it has become increasingly evident that sex-based differences play a pivotal role in the pathophysiology, presentation, and outcomes of these conditions. Despite this recognition, the majority of clinical research in the field has historically focused on male populations, leading to a significant knowledge gap in understanding the unique aspects of ACS in women. This review article aims to comprehensively explore and synthesize the current body of literature concerning the sex-specific characteristics of ACS, shedding light on the epidemiology, risk factors, clinical presentation, diagnostic challenges, treatment strategies, and prognosis in women. By elucidating the distinct aspects of ACS in women, this review intends to foster greater awareness and improved clinical management, ultimately contributing to enhanced cardiovascular care for female patients.
Collapse
Affiliation(s)
- Panagiotis Theofilis
- 1st Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Panayotis K Vlachakis
- 1st Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Emmanouil Mantzouranis
- 1st Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Athanasios Sakalidis
- 1st Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Christina Chrysohoou
- 1st Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioannis Leontsinis
- 1st Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - George Lazaros
- 1st Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Kyriakos Dimitriadis
- 1st Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria Drakopoulou
- 1st Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Aikaterini Vordoni
- 1st Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Evangelos Oikonomou
- 3rd Department of Cardiology, "Sotiria" Chest Disease Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Konstantinos Tsioufis
- 1st Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitris Tousoulis
- 1st Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
6
|
Wang Q, Yang X, Chen C, Xing Y, Chitakwa N, Jiang J, Wei H, Ding X, Wu D. Sex-specific effects of aged polystyrene microplastics on hepatic AMPK pathway activation and lipid droplet accumulation in MAFLD mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 292:117963. [PMID: 40058092 DOI: 10.1016/j.ecoenv.2025.117963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 02/16/2025] [Accepted: 02/23/2025] [Indexed: 03/17/2025]
Abstract
Microplastics (MPs) are environmental pollutants attracting widespread attention due to their environmental omnipresence and potential health effects. MPs undergo ageing in the environment and our previous research found that aged Polystyrene microplastics (PS-MPs) affected lipid metabolism in healthy female mice, but not males. In this study, we examined the effects of aged PS-MP exposure on lipid metabolism in mice with Metabolic Associated Fatty Liver Disease (MAFLD). 14 female and 14 male mice were furnished with a high-fat diet (HFD) for eight weeks to create MAFLD model mice. They were then orally administered aged PS-MPs for four weeks, and changes in the AMP-activated protein kinase signalling pathway were examined in order to determine PS-MP's effect on hepatic metabolism. The outcomes showed that though serum estradiol, inflammatory gene expression and ROS levels increased significantly in both male and female HFD-aged PS-MP groups, hepatic steatosis was attenuated only in the female group. Furthermore, serum ERα, ERβ, AMPKα, acetyl-CoA carboxylase, sterol regulatory element binding protein-1c, and Fas expressions were significantly increased in the MAFLD mice groups compared to the control group. Combining serum E2 levels, AMPK pathway changes, oxidative stress markers, and inflammatory gene levels, aged PS-MPs may stimulate E2 production and mobilize the liver AMPK signalling pathway of both male and female MAFLD mice. However, lipid metabolism is only affected in female MAFLD mice, suggesting other possible mechanisms besides the AMPK pathway may be at play. These results provide a new perspective on the potential health effects of MP exposure in individuals with metabolic disorders such as MAFLD.
Collapse
Affiliation(s)
- Qing Wang
- Wuxi Medical Center, Nanjing Medical University, Wuxi 214023, China; State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Xiaona Yang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Disease Prevention and Control Center of Linping District, Hangzhou 311100, China
| | - Chuan Chen
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Ying Xing
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Natasha Chitakwa
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Jing Jiang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Hongcheng Wei
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Xinliang Ding
- The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi Medical Center, Nanjing Medical University, Wuxi 214023, China.
| | - Di Wu
- Wuxi Medical Center, Nanjing Medical University, Wuxi 214023, China; State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China.
| |
Collapse
|
7
|
Kim Y, Kim D, Kim J, Yun M, Kang ES. Association between appendicular skeletal muscle mass and myocardial glucose uptake measured by 18F-FDG PET. ESC Heart Fail 2025; 12:467-476. [PMID: 39344859 PMCID: PMC11769618 DOI: 10.1002/ehf2.15086] [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: 04/30/2024] [Revised: 08/18/2024] [Accepted: 09/05/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND Low muscle mass is associated with high insulin resistance and an increased risk of cardiovascular disease. This study aims to determine whether low muscle mass affects the alterations in myocardial substrate metabolism that are associated with the development of cardiovascular disease. METHOD The study included 299 individuals (182 men and 117 women) who underwent examination at the Severance Health Check-up Center between January 2018 and February 2019. Myocardial glucose uptake was assessed using [18F]-fluorodeoxyglucose-positron emission tomography (18F-FDG PET/CT) scanning. Direct segmental bioimpedance analysis was used to measure appendicular skeletal muscle mass (ASM). RESULTS We analysed men and women separately owing to sex-related body composition differences. ASM/Ht2 was significantly positively correlated with myocardial glucose uptake measured by 18F-FDG PET/CT [ln (SUVheart/liver)] only in men (r = 0.154, P = 0.038 in men; r = -0.042, P = 0.652 in women, respectively). In men, myocardial glucose uptake was significantly associated with ASM/Ht2 even after adjusting for multiple confounders in a multivariable linear regression model (standardized β = 0.397, P = 0.004, in men; β = - 0.051, P = 0.698, in women). In women, age (β = -0.424 P = 0.029) was independent determinants of myocardial glucose uptake. CONCLUSIONS In men, ASM was strongly associated with myocardial glucose uptake as measured by 18F-FDG PET/CT. In women, age was significantly correlated with myocardial substrate utilization, but not with ASM.
Collapse
Affiliation(s)
- Young‐eun Kim
- Division of Endocrinology and Metabolism, Department of Internal MedicineYonsei University College of MedicineSeoulRepublic of Korea
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University Anam HospitalKorea University College of MedicineSeoulRepublic of Korea
| | - Dongwoo Kim
- Department of Nuclear MedicineYonsei University College of MedicineSeoulSouth Korea
| | - Jiwon Kim
- Division of Endocrinology and Metabolism, Department of Internal MedicineYonsei University College of MedicineSeoulRepublic of Korea
- Department of Internal Medicine, National Health Insurance Service Ilsan HospitalGoyangRepublic of Korea
| | - Mijin Yun
- Department of Nuclear MedicineYonsei University College of MedicineSeoulSouth Korea
| | - Eun Seok Kang
- Division of Endocrinology and Metabolism, Department of Internal MedicineYonsei University College of MedicineSeoulRepublic of Korea
| |
Collapse
|
8
|
Tang R, Xiao G, Liu Y, Jia D, Zeng Z, Jia C, Li D, Li Y, Jiang J, Li S, Bi X. Integrated serum pharmacochemistry, pharmacokinetics, and network analysis to explore active components of BuShao Tiaozhi Capsule on hyperlipidemia. Front Pharmacol 2025; 15:1444967. [PMID: 39830346 PMCID: PMC11738623 DOI: 10.3389/fphar.2024.1444967] [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: 06/06/2024] [Accepted: 11/25/2024] [Indexed: 01/22/2025] Open
Abstract
BuShao Tiaozhi Capsule (BSTZC), a novel drug in China, has been used to treat hyperlipidemia (HLP) in clinical practice for many years. Despite our previous studies suggesting that BSTZC can treat HLP, there is a lack of a rapid and systematic method to explore its active components. Therefore, in this study, we aimed to investigate the active components and mechanisms of BSTZC in treating HLP by integrating serum pharmacology, pharmacokinetics, network analysis, and experimental validation. We first established UPLC fingerprints, calibrated 23 common peaks, and identified 13 common peaks, and the similarity was greater than 0.99 for 10 batches. A total of nine metabolites from BSTZC were identified in serum and considered as PK markers. The pharmacokinetic parameters of the PK markers were compared between the control group and the model group through the pharmacokinetics study to determine the dynamic changes of representative components in rats. Compared with the control group, the Cmax and AUC0→t of OXY, IVT, IVL, and KPF-3-G were significantly higher (P< 0.05); the AUC0→∞ of OXY, PN, and IVT was significantly higher (P< 0.05); and the t1/2 of IVT, SA, and KPF-3-G was significantly different (P< 0.05). In vivo experiments showed that BSTZC and its active components could effectively alleviate lipid metabolism disorders and liver injury, with obvious lipid-lowering effects. Further studies showed that BSTZC alleviated HLP by inhibiting the PI3K/Akt signaling pathway, which was consistent with the results of the network analysis study. Our results revealed the active components and mechanisms of BSTZC in the treatment of HLP, which could provide useful information to guide the clinical application of BSTZC.
Collapse
Affiliation(s)
- Ruiyin Tang
- School of the Fifth Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Guanlin Xiao
- Guangdong Provincial Engineering Technology Research Institute of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Research and Development in Traditional Chinese Medicine, Guangzhou, Guangdong, China
| | - Yanchang Liu
- School of the Fifth Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Dezheng Jia
- School of the Fifth Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Zhihao Zeng
- School of the Fifth Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Canchao Jia
- School of the Fifth Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Dongmei Li
- School of the Fifth Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yangxue Li
- Guangdong Provincial Engineering Technology Research Institute of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Research and Development in Traditional Chinese Medicine, Guangzhou, Guangdong, China
| | - Jieyi Jiang
- Guangdong Provincial Engineering Technology Research Institute of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Research and Development in Traditional Chinese Medicine, Guangzhou, Guangdong, China
| | - Sumei Li
- Guangdong Provincial Engineering Technology Research Institute of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Research and Development in Traditional Chinese Medicine, Guangzhou, Guangdong, China
| | - Xiaoli Bi
- Guangdong Provincial Engineering Technology Research Institute of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Research and Development in Traditional Chinese Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
9
|
Dai J, Wang T, Qiu S, Qi X, Zeng J, Chen C, Wu S, Qiu D, Bai S. Environmental sex reversal in parrotfish does not cause differences in the structure of their gut microbial communities. BMC Microbiol 2024; 24:531. [PMID: 39701987 DOI: 10.1186/s12866-024-03698-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 12/10/2024] [Indexed: 12/21/2024] Open
Abstract
Parrotfish are a common fish in coral reef areas, but little is known about their gut microbial communities. In addition, parrotfish are capable of sex reversal, usually some males are sexually reversed from females, and it is still not known whether this sex reversal leads to significant changes in gut microbial communities. In this study, we investigated the gut microbial communities of three species of parrotfish including Scarus forsteni (4 females and 4 sex-reversed males), Scarus ghobban (5 females and 5 sex-reversed males), and Hipposcarus longiceps (5 females and 5 sex-reversed males) by using high-throughput sequencing technology. The gut microbial communities of these three species were mainly composed of Pseudomonadota (class Gammaproteobacteria) and Bacillota, while at the family level, they mainly included Vibrionaceae, Burkholderiaceae, Enterobacteriaceae, Streptococcacea, and Erwiniaceae. Although at the genus level, there were a large number of unclassified lineages, the remaining gut microorganisms were mainly composed of Vibrio, Photobacterium, Enterococcus and Lactococcus. Furthermore, we did not find significant differences in gut microbial community structure between the female parrotfish and corresponding female reversed males within each species, even in terms of the structure of gut microbial functional information obtained from 16 S rRNA gene sequence predictions. However, the gut microbial communities of these three species of parrotfish differed significantly not only in their community structure but also in their microbial functional information structure, mainly in terms of aspartate and asparagine biosynthesis, histidine degradation, inositol degradation, heptose biosynthesis, chitin derivatives degradation, enterobactin biosynthesis, and thiazole biosynthesis. Our study provides essential gut microbial community data for understanding the physiology and sex reversal phenomenon in parrotfish.
Collapse
Affiliation(s)
- Jingcheng Dai
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Teng Wang
- Scientific Observation and Research Station of Xisha Island Reef Fishery Ecosystem of Hainan Province, Sanya Tropical Fisheries Research Institute, Sanya, 572018, China
| | - Shunda Qiu
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Xiaoxue Qi
- Institute of Deep-sea Science and Engineering, Chinese Academy of Sciences, Sanya, 572000, China
| | - Juntao Zeng
- Institute of Deep-sea Science and Engineering, Chinese Academy of Sciences, Sanya, 572000, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Changcui Chen
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Siqi Wu
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Dongru Qiu
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430023, China
| | - Shijie Bai
- Institute of Deep-sea Science and Engineering, Chinese Academy of Sciences, Sanya, 572000, China.
| |
Collapse
|
10
|
Chai X, Zhang J, Wang Y, Li D, Zhu D, Liang K, Yang C, Wang J, Gong Q, Yang Z, Shao R. Interaction between sex and one-hour post-load glucose on metabolic syndrome and its components among Chinese people at high risk of diabetes. Diabetol Metab Syndr 2024; 16:295. [PMID: 39696709 DOI: 10.1186/s13098-024-01544-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 11/29/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Recently, International Diabetes Federation position statement has adopted one-hour post-load glucose (1hPG) ≥ 8.6 mmol/L for diagnosing intermediate hyperglycemia. We aimed to assess the association of 1hPG ≥ 8.6 mmol/L with metabolic syndrome (MetS) and its components, as well as interaction between sex and 1hPG ≥ 8.6 mmol/L on MetS and its components in Chinese people at high risk of diabetes. METHODS The cross-sectional study was conducted in DaQing city of HeiLongJiang Province, China between August, 2023 and January, 2024. Eligible individuals with fasting glucose of 5.6-6.9 mmol/L and age of 25-55 years in health checkup data in the year of 2023 or with at least one risk factor of diabetes were invited to receive the oral glucose tolerance test and biochemical examinations. Individuals with self-reported presence of diabetes or usage of glucose-lowering medication were excluded. MetS was defined as presence of at least three of the five components according to the Chinese Diabetes Society criteria. Logistic regression was performed to evaluate the association of 1hPG ≥ 8.6 mmol/L with MetS and its components. Additive interaction was estimated using the relative excess risk due to interaction, attributable proportion due to interaction (AP), and synergy index. RESULTS A total of 2419 subjects comprising 1465 men (60.6%) with a mean age of 45.77 ± 6.20 years were included, and the prevalence of MetS was 46.8%, with 59.7% in men and 27.1% in women. 1hPG ≥ 8.6 mmol/L was associated with MetS (aOR = 4.40, 95% CI 3.26-6.01), elevated blood pressure (aOR = 1.46, 95% CI 1.13-1.89), hyperglycemia (aOR = 15.46, 95% CI 11.56-20.98), and reduced HDL-C (aOR = 1.51, 95% CI 1.07-2.15) in the overall population, whereas no significant association between 1hPG ≥ 8.6 mmol/L and elevated blood pressure in men (aOR = 1.36, 95% CI 0.97-1.91) or dyslipidemia in women (elevated TG: aOR = 0.81, 95% CI 0.47-1.39; reduced HDL-C: aOR = 1.08, 95% CI 0.49-2.37). Additive interaction effect between sex and 1hPG ≥ 8.6 mmol/L on MetS was observed, with 31% attributed to the interaction effect between men and 1hPG ≥ 8.6 mmol/L (AP = 0.31, 95% CI 0.06-0.49). CONCLUSIONS There was an additive interaction effect between sex and 1hPG on MetS among Chinese people at high risk of diabetes. 1hPG test and sex-specific strategies should be taken into consideration in cardiometabolic disorder identification and management.
Collapse
Affiliation(s)
- Xin Chai
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Juan Zhang
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China.
| | - Yachen Wang
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Di Li
- Daqing Oilfield General Hospital (Daqing First Hospital), Daqing, 163000, China
| | - Dongli Zhu
- Daqing Oilfield General Hospital (Daqing First Hospital), Daqing, 163000, China
| | - Kaipeng Liang
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Chunyu Yang
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Jinping Wang
- Daqing Oilfield General Hospital (Daqing First Hospital), Daqing, 163000, China
| | - Qiuhong Gong
- Center of Endocrinology, National Center of Cardiology & Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Zhiwei Yang
- Daqing Oilfield General Hospital (Daqing First Hospital), Daqing, 163000, China
| | - Ruitai Shao
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| |
Collapse
|
11
|
Marshall CL, Mostafa D, Hemshehkar M, Lao Y, Balshaw R, Spicer V, Mookherjee N. Biological Sex Is an Effect Modifier of Allergen-Mediated Alteration of the Lung Proteome. J Proteome Res 2024; 23:4203-4215. [PMID: 39214566 DOI: 10.1021/acs.jproteome.4c00025] [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] [Indexed: 09/04/2024]
Abstract
Asthma exhibits a distinct sex bias in the disease prevalence, severity, and response to therapy. However, sex-related differences in alterations of the lung proteome mediated by aeroallergens critical in asthma, such as house dust mites (HDM), remain unknown. In this study, we define sex-related differences in the lung proteome using an HDM-challenged mouse model by 1D LC-MS/MS. Sex-disaggregated data analysis showed that 406 proteins were uniquely altered in females, 273 proteins were uniquely altered in males, and 414 proteins were altered in both females and males in response to HDM. In a linear mixed model analysis, sex modified the HDM exposure effect for 163 proteins, i.e., a significant sex:exposure interaction was identified in 84 proteins in females and 35 proteins in males. Of these, 12 proteins showed a significant sex effect in both female and male lungs. We further selected 3 proteins Tjp1, Lamtor1, and G3BP2 for independent confirmation studies. Our findings detail the sex-specific lung proteome in response to an aeroallergen critical in asthma and demonstrate that sex is a significant effect modifier of HDM response. These results will serve as a valuable resource for delineating sex-specific mechanisms in aeroallergen-driven responses in asthma research.
Collapse
Affiliation(s)
- Courtney Lynn Marshall
- Department of Immunology, Rady Faculty of Health Sciences, University of Manitoba, Mercedes-Benz, Winnipeg R3E0T5, Canada
- Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, Rady Faculty of Health Sciences, University of Manitoba, Mercedes-Benz, Winnipeg R3E3P4, Canada
| | - Dina Mostafa
- Department of Immunology, Rady Faculty of Health Sciences, University of Manitoba, Mercedes-Benz, Winnipeg R3E0T5, Canada
- Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, Rady Faculty of Health Sciences, University of Manitoba, Mercedes-Benz, Winnipeg R3E3P4, Canada
| | - Mahadevappa Hemshehkar
- Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, Rady Faculty of Health Sciences, University of Manitoba, Mercedes-Benz, Winnipeg R3E3P4, Canada
| | - Ying Lao
- Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, Rady Faculty of Health Sciences, University of Manitoba, Mercedes-Benz, Winnipeg R3E3P4, Canada
| | - Robert Balshaw
- George and Fay Yee Centre for Healthcare Innovation, University of Manitoba, Mercedes-Benz, Winnipeg R3E0T6, Canada
| | - Victor Spicer
- Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, Rady Faculty of Health Sciences, University of Manitoba, Mercedes-Benz, Winnipeg R3E3P4, Canada
| | - Neeloffer Mookherjee
- Department of Immunology, Rady Faculty of Health Sciences, University of Manitoba, Mercedes-Benz, Winnipeg R3E0T5, Canada
- Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, Rady Faculty of Health Sciences, University of Manitoba, Mercedes-Benz, Winnipeg R3E3P4, Canada
| |
Collapse
|
12
|
Pisanu C, Congiu D, Meloni A, Paribello P, Severino G, Ardau R, Chillotti C, Als TD, Børglum AD, Del Zompo M, Manchia M, Squassina A. Sex differences in shared genetic determinants between severe mental disorders and metabolic traits. Psychiatry Res 2024; 342:116195. [PMID: 39299147 DOI: 10.1016/j.psychres.2024.116195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 09/02/2024] [Accepted: 09/13/2024] [Indexed: 09/22/2024]
Abstract
High rates of metabolic risk factors contribute to premature mortality in patients with severe mental disorders, but the molecular underpinnings of this association are largely unknown. We performed the first analysis on shared genetic factors between severe mental disorders and metabolic traits considering the effect of sex. We applied an integrated analytical pipeline on the largest sex-stratified genome-wide association datasets available for bipolar disorder (BD), major depressive disorder (MDD), schizophrenia (SZ), and for body mass index (BMI) and waist-to-hip ratio (WHR) (all including participants of European origin). We observed extensive genetic overlap between all severe mental disorders and variants associated with BMI in women or men and identified several genetic loci shared between BD, or SZ and BMI in women (24 and 91, respectively) or men (13 and 208, respectively), with mixed directions of effect. A large part of the identified genetic variants showed sex differences in terms of location, genes modulated in adipose tissue and/or brain regions, and druggable targets. By providing a complete picture of disorder specific and cross-disorder shared genetic determinants, our results highlight potential sex differences in the genetic liability to metabolic comorbidities in patients with severe mental disorders.
Collapse
Affiliation(s)
- Claudia Pisanu
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy.
| | - Donatella Congiu
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
| | - Anna Meloni
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
| | - Pasquale Paribello
- Section of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy; Unit of Clinical Psychiatry, University Hospital Agency of Cagliari, Cagliari, Italy
| | - Giovanni Severino
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
| | - Raffaella Ardau
- Unit of Clinical Pharmacology, University Hospital Agency of Cagliari, Cagliari, Italy
| | - Caterina Chillotti
- Unit of Clinical Pharmacology, University Hospital Agency of Cagliari, Cagliari, Italy
| | - Thomas D Als
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark; Center for Genomics and Personalized Medicine, Aarhus, Denmark
| | - Anders D Børglum
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark; Center for Genomics and Personalized Medicine, Aarhus, Denmark
| | - Maria Del Zompo
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
| | - Mirko Manchia
- Section of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy; Unit of Clinical Psychiatry, University Hospital Agency of Cagliari, Cagliari, Italy; Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
| | - Alessio Squassina
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy.
| |
Collapse
|
13
|
Krolick KN, Cao J, Gulla EM, Bhardwaj M, Marshall SJ, Zhou EY, Kiss AJ, Choueiry F, Zhu J, Shi H. Subregion-specific transcriptomic profiling of rat brain reveals sex-distinct gene expression impacted by adolescent stress. Neuroscience 2024; 553:19-39. [PMID: 38977070 PMCID: PMC11444371 DOI: 10.1016/j.neuroscience.2024.07.002] [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: 01/18/2024] [Revised: 05/14/2024] [Accepted: 07/02/2024] [Indexed: 07/10/2024]
Abstract
Stress during adolescence clearly impacts brain development and function. Sex differences in adolescent stress-induced or exacerbated emotional and metabolic vulnerabilities could be due to sex-distinct gene expression in hypothalamic, limbic, and prefrontal brain regions. However, adolescent stress-induced whole-genome expression changes in key subregions of these brain regions were unclear. In this study, female and male adolescent Sprague Dawley rats received one-hour restraint stress daily from postnatal day (PD) 32 to PD44. Corticosterone levels, body weights, food intake, body composition, and circulating adiposity and sex hormones were measured. On PD44, brain and blood samples were collected. Using RNA-sequencing, sex-specific differences in stress-induced differentially expressed (DE) genes were identified in subregions of the hypothalamus, limbic system, and prefrontal cortex. Canonical pathways reflected well-known sex-distinct maladies and diseases, substantiating the therapeutic potential of the DE genes found in the current study. Thus, we proposed specific sex distinct, adolescent stress-induced transcriptional changes found in the current study as examples of the molecular bases for sex differences witnessed in stress induced or exacerbated emotional and metabolic disorders. Future behavioral studies and single-cell studies are warranted to test the implications of the DE genes identified in this study in sex-distinct stress-induced susceptibilities.
Collapse
Affiliation(s)
| | - Jingyi Cao
- Department of Biology, Miami University, Oxford, OH 45056, USA.
| | - Evelyn M Gulla
- Department of Biology, Miami University, Oxford, OH 45056, USA.
| | - Meeta Bhardwaj
- Department of Biology, Miami University, Oxford, OH 45056, USA.
| | | | - Ethan Y Zhou
- Department of Biology, Miami University, Oxford, OH 45056, USA.
| | - Andor J Kiss
- Center for Bioinformatics & Functional Genomics, Miami University, Oxford, OH 45056, USA.
| | - Fouad Choueiry
- Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA.
| | - Jiangjiang Zhu
- Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA; James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA.
| | - Haifei Shi
- Department of Biology, Miami University, Oxford, OH 45056, USA.
| |
Collapse
|
14
|
Mahapatra G, Gao Z, Bateman JR, Lockhart SN, Bergstrom J, Piloso JE, Craft S, Molina AJA. Peripheral Blood Cells From Older Adults Exhibit Sex-Associated Differences in Mitochondrial Function. J Gerontol A Biol Sci Med Sci 2024; 79:glae098. [PMID: 38602189 PMCID: PMC11059251 DOI: 10.1093/gerona/glae098] [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: 11/22/2023] [Indexed: 04/12/2024] Open
Abstract
Blood-based mitochondrial bioenergetic profiling is a feasible, economical, and minimally invasive approach that can be used to examine mitochondrial function and energy metabolism in human subjects. In this study, we use 2 complementary respirometric techniques to evaluate mitochondrial bioenergetics in both intact and permeabilized peripheral blood mononuclear cells (PBMCs) and platelets to examine sex dimorphism in mitochondrial function among older adults. Employing equal numbers of PBMCs and platelets to assess mitochondrial bioenergetics, we observe significantly higher respiration rates in female compared to male participants. Mitochondrial bioenergetic differences remain significant after controlling for independent parameters including demographic parameters (age, years of education), and cognitive parameters (mPACC5, COGDX). Our study illustrates that circulating blood cells, immune cells in particular, have distinctly different mitochondrial bioenergetic profiles between females and males. These differences should be taken into account as blood-based bioenergetic profiling is now commonly used to understand the role of mitochondrial bioenergetics in human health and aging.
Collapse
Affiliation(s)
- Gargi Mahapatra
- Division of Geriatrics, Gerontology, and Palliative Care, Department of Medicine, University of California San Diego School of Medicine, La Jolla, California, USA
| | - Zhengrong Gao
- Section on Gerontology and Geriatrics, Department of Internal Medicine, Sticht Center for Healthy Aging and Alzheimer’s Prevention, Wake Forest Baptist Medical Center, Winston-Salem, North Carolina, USA
| | - James R Bateman
- Department of Neurology, Wake Forest Baptist Medical Center, Winston-Salem, North Carolina, USA
- Section on Gerontology and Geriatrics, Department of Internal Medicine, Sticht Center for Healthy Aging and Alzheimer’s Prevention, Wake Forest Baptist Medical Center, Winston-Salem, North Carolina, USA
| | - Samuel Neal Lockhart
- Section on Gerontology and Geriatrics, Department of Internal Medicine, Sticht Center for Healthy Aging and Alzheimer’s Prevention, Wake Forest Baptist Medical Center, Winston-Salem, North Carolina, USA
| | - Jaclyn Bergstrom
- Division of Geriatrics, Gerontology, and Palliative Care, Department of Medicine, University of California San Diego School of Medicine, La Jolla, California, USA
| | - Jemima Elizabeth Piloso
- Section on Gerontology and Geriatrics, Department of Internal Medicine, Sticht Center for Healthy Aging and Alzheimer’s Prevention, Wake Forest Baptist Medical Center, Winston-Salem, North Carolina, USA
| | - Suzanne Craft
- Section on Gerontology and Geriatrics, Department of Internal Medicine, Sticht Center for Healthy Aging and Alzheimer’s Prevention, Wake Forest Baptist Medical Center, Winston-Salem, North Carolina, USA
| | - Anthony J A Molina
- Division of Geriatrics, Gerontology, and Palliative Care, Department of Medicine, University of California San Diego School of Medicine, La Jolla, California, USA
| |
Collapse
|
15
|
Liu DS, Wang XS, Zhong XH, Cao H, Zhang F. Sexual dimorphism in the gut microbiota and sexual dimorphism in chronic diseases: Association or causation? J Steroid Biochem Mol Biol 2024; 237:106451. [PMID: 38154505 DOI: 10.1016/j.jsbmb.2023.106451] [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: 10/20/2022] [Revised: 03/31/2023] [Accepted: 12/24/2023] [Indexed: 12/30/2023]
Abstract
Understanding the sexual dimorphism in diseases is essential to investigate the pathogenesis of some chronic diseases (e.g., autoimmune diseases, etc). The gut microbiota has been found to show a notable impact on the pathology of several chronic diseases in recent years. Intriguingly, the composition of the gut microbiota varies between sexes. Here, we reviewed 'facts and fiction' regarding sexual dimorphism in chronic diseases and sexual dimorphism in the gut microbiota respectively. The association and causative relationship between them aiming to elucidate the pathological mechanisms of sexual dimorphism in chronic diseases were further explored. The development of gender-special food products based on the sexual dimorphism in the gut microbiota were recommended, which would be beneficial to facilitating the personalized treatment.
Collapse
Affiliation(s)
- Dong-Song Liu
- Affiliated Hospital of Jiangnan University, Wuxi, China; Nantong University, Nantong, China
| | - Xue-Song Wang
- Affiliated Hospital of Jiangnan University, Wuxi, China; Nantong University, Nantong, China; Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Xiao-Hui Zhong
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Hong Cao
- Affiliated Hospital of Jiangnan University, Wuxi, China; Nantong University, Nantong, China; Wuxi School of Medicine, Jiangnan University, Wuxi, China.
| | - Feng Zhang
- Affiliated Hospital of Jiangnan University, Wuxi, China; Wuxi School of Medicine, Jiangnan University, Wuxi, China.
| |
Collapse
|
16
|
Teh CH, Rampal S, Kee CC, Azahadi O, Tahir A. Body mass index and waist circumference trajectories across the life course and birth cohorts, 1996-2015 Malaysia: sex and ethnicity matter. Int J Obes (Lond) 2023; 47:1302-1308. [PMID: 37833560 PMCID: PMC10663154 DOI: 10.1038/s41366-023-01391-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 09/19/2023] [Accepted: 09/22/2023] [Indexed: 10/15/2023]
Abstract
OBJECTIVE The global obesity epidemic remains a significant threat to public health and the economy. Age-period-cohort (APC) analysis is one method to model the trajectory of obesity. However, there is scarce published evidence of such analyses among the South East Asian population. This study aims to explore the sex and ethnic variations of BMI and waist circumference trajectories over time among non-institutionalized Malaysian adults aged 18 to 80 years. METHODS Data from four population-based National Health and Morbidity Surveys conducted in 1996, 2006, 2010, and 2015 were pooled. Hierarchical Age-Period-Cohort (HAPC) analysis explored the trajectories of BMI and waist circumference across the life course and birth cohorts by sex and ethnicity. These models assumed no period effect. RESULTS Generally, BMI and waist circumference trajectories increased across age and birth cohorts. These trajectories varied by sex and ethnicity. Females have more profound increasing BMI and waist circumference trajectories than their male counterparts as they age and as cohort recency increases. Chinese have less profound BMI and waist circumference increases across the life course and birth cohorts than other ethnic groups. CONCLUSIONS The profound increasing cohort trajectories of obesity, regardless of sex and ethnicity, are alarming. Future studies should focus on identifying factors associated with the less profound cohort effect among the Chinese to reduce the magnitude of trajectories in obesity, particularly among future generations.
Collapse
Affiliation(s)
- Chien Huey Teh
- Centre for Epidemiology and Evidence-based Practice, Department of Social and Preventive Medicine, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
- Institute for Medical Research, National Institutes of Health, Ministry of Health Malaysia, 40170, Setia Alam, Malaysia
| | - Sanjay Rampal
- Centre for Epidemiology and Evidence-based Practice, Department of Social and Preventive Medicine, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia.
| | - Chee Cheong Kee
- Sector for Biostatistics and Data Repository, National Institutes of Health, Ministry of Health Malaysia, 40170, Setia Alam, Malaysia
| | - Omar Azahadi
- Sector for Biostatistics and Data Repository, National Institutes of Health, Ministry of Health Malaysia, 40170, Setia Alam, Malaysia
| | - Aris Tahir
- Institute for Medical Research, National Institutes of Health, Ministry of Health Malaysia, 40170, Setia Alam, Malaysia
| |
Collapse
|
17
|
Zhou B, Gong N, He Q, Huang X, Zhu J, Zhang L, Huang Y, Tan X, Xia Y, Zheng Y, Shi Q, Qin C. Clustering of lifestyle behaviours and analysis of their associations with MAFLD: a cross-sectional study of 196,515 individuals in China. BMC Public Health 2023; 23:2303. [PMID: 37990228 PMCID: PMC10664514 DOI: 10.1186/s12889-023-17177-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 11/07/2023] [Indexed: 11/23/2023] Open
Abstract
BACKGROUND The aggregation of lifestyle behaviours and their association with metabolic-associated fatty liver disease (MAFLD) remain unclear. We identified lifestyle patterns and investigated their association with the risk of developing MAFLD in a sample of Chinese adults who underwent annual physical examinations. METHODS Annual physical examination data of Chinese adults from January 2016 to December 2020 were used in this study. We created a scoring system for lifestyle items combining a statistical method (multivariate analysis of variance) and clinical expertise (Delphi method). Subsequently, principal component analysis and two-step cluster analysis were implemented to derive the lifestyle patterns of men and women. Binary logistic regression analysis was used to explore the prevalence risk of MAFLD among lifestyle patterns stratified by sex. RESULTS A total of 196,515 subjects were included in the analysis. Based on the defined lifestyle scoring system, nine and four lifestyle patterns were identified for men and women, respectively, which included "healthy or unhealthy" patterns and mixed patterns containing a combination of healthy and risky lifestyle behaviours. This study showed that subjects with an unhealthy or mixed pattern had a significantly higher risk of developing MAFLD than subjects with a relatively healthy pattern, especially among men. CONCLUSIONS Clusters of unfavourable behaviours are more prominent in men than in women. Lifestyle patterns, as important factors influencing the development of MAFLD, show significant sex differences in the risk of MAFLD. There is a strong need for future research to develop targeted MAFLD interventions based on the identified behavioural clusters by sex stratification.
Collapse
Affiliation(s)
- Bingqian Zhou
- Department of Health Management Center, The Third Xiangya Hospital, Central South University, No.138, Tongzipo Road, Changsha, 410013, China
- Xiangya Nursing School, Central South University, Changsha, 410013, China
| | - Ni Gong
- Department of Health Management Center, The Third Xiangya Hospital, Central South University, No.138, Tongzipo Road, Changsha, 410013, China
- Xiangya Nursing School, Central South University, Changsha, 410013, China
| | - Qingnan He
- Department of Health Management Center, The Third Xiangya Hospital, Central South University, No.138, Tongzipo Road, Changsha, 410013, China
| | - Xinjuan Huang
- Xiangya Nursing School, Central South University, Changsha, 410013, China
| | - Jingchi Zhu
- Jishou University School of Medicine, Jishou, 416000, China
| | - Lijun Zhang
- State Key Laboratory of Ultrasound in Medicine and Engineering, School of Public Health and Management, Chongqing Medical University, No.1, Medical College Road , Chongqing, 400016, China
| | - Yanyan Huang
- State Key Laboratory of Ultrasound in Medicine and Engineering, School of Public Health and Management, Chongqing Medical University, No.1, Medical College Road , Chongqing, 400016, China
| | - Xinyun Tan
- Xiangya Nursing School, Central South University, Changsha, 410013, China
| | - Yuanqin Xia
- Jishou University School of Medicine, Jishou, 416000, China
| | - Yu Zheng
- Xiangya Nursing School, Central South University, Changsha, 410013, China
| | - Qiuling Shi
- State Key Laboratory of Ultrasound in Medicine and Engineering, School of Public Health and Management, Chongqing Medical University, No.1, Medical College Road , Chongqing, 400016, China.
| | - Chunxiang Qin
- Department of Health Management Center, The Third Xiangya Hospital, Central South University, No.138, Tongzipo Road, Changsha, 410013, China.
- Xiangya Nursing School, Central South University, Changsha, 410013, China.
| |
Collapse
|
18
|
Bloyd M, Sinaii N, Faucz FR, Iben J, Coon SL, Caprio S, Santoro N, Stratakis CA, London E. High-frequency variants in PKA signaling-related genes within a large pediatric cohort with obesity or metabolic abnormalities. Front Endocrinol (Lausanne) 2023; 14:1272939. [PMID: 38027204 PMCID: PMC10679389 DOI: 10.3389/fendo.2023.1272939] [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: 08/04/2023] [Accepted: 10/05/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction Pediatric obesity has steadily increased in recent decades. Large-scale genome-wide association studies (GWAS) conducted primarily in Eurocentric adult populations have identified approximately 100 loci that predispose to obesity and type II diabetes. GWAS in children and individuals of non-European descent, both disproportionately affected by obesity, are fewer. Rare syndromic and monogenic obesities account for only a small portion of childhood obesity, so understanding the role of other genetic variants and their combinations in heritable obesities is key to developing targeted and personalized therapies. Tight and responsive regulation of the cAMP-dependent protein kinase (PKA) signaling pathway is crucial to maintaining healthy energy metabolism, and mutations in PKA-linked genes represent the most common cause of monogenic obesity. Methods For this study, we performed targeted exome sequencing of 53 PKA signaling-related genes to identify variants in genomic DNA from a large, ethnically diverse cohort of obese or metabolically challenged youth. Results We confirmed 49 high-frequency variants, including a novel variant in the PDE11A gene (c.152C>T). Several other variants were associated with metabolic characteristics within ethnic groups. Discussion We conclude that a PKA pathway-specific variant search led to the identification of several new genetic associations with obesity in an ethnically diverse population.
Collapse
Affiliation(s)
- Michelle Bloyd
- Section on Endocrinology and Genetics, Program on Developmental Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Bethesda, MD, United States
| | - Ninet Sinaii
- Biostatistics and Clinical Epidemiology Service, National Institutes of Health (NIH) Clinical Center, Bethesda, MD, United States
| | - Fabio Rueda Faucz
- Section on Endocrinology and Genetics, Program on Developmental Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Bethesda, MD, United States
| | - James Iben
- Molecular Genomics Core, National Institute of Child Health and Human Development (NICHD), Bethesda, MD, United States
| | - Steven L. Coon
- Molecular Genomics Core, National Institute of Child Health and Human Development (NICHD), Bethesda, MD, United States
| | - Sonia Caprio
- Section on Pediatric Endocrinology and Diabetes, Yale University, New Haven, CT, United States
| | - Nicola Santoro
- Section on Pediatric Endocrinology and Diabetes, Yale University, New Haven, CT, United States
- Department of Medicine and Health Sciences, “V. Tiberio” University of Molise, Campobasso, Italy
| | - Constantine A. Stratakis
- Section on Endocrinology and Genetics, Program on Developmental Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Bethesda, MD, United States
- Human Genetics and Precision Medicine, Institute for Molecular Biology and Biotechnology (IMBB), Foundation for Research & Technology Hellas (FORTH), Heraklion, ELPEN Research Institute, Athens, Greece
| | - Edra London
- Section on Endocrinology and Genetics, Program on Developmental Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Bethesda, MD, United States
| |
Collapse
|
19
|
Wagner VA, Holl KL, Clark KC, Reho JJ, Lehmler HJ, Wang K, Grobe JL, Dwinell MR, Raff H, Kwitek AE. The Power of the Heterogeneous Stock Rat Founder Strains in Modeling Metabolic Disease. Endocrinology 2023; 164:bqad157. [PMID: 37882530 PMCID: PMC10637104 DOI: 10.1210/endocr/bqad157] [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: 07/07/2023] [Revised: 10/04/2023] [Accepted: 10/24/2023] [Indexed: 10/27/2023]
Abstract
Metabolic diseases are a host of complex conditions, including obesity, diabetes mellitus, and metabolic syndrome. Endocrine control systems (eg, adrenals, thyroid, gonads) are causally linked to metabolic health outcomes. N/NIH Heterogeneous Stock (HS) rats are a genetically heterogeneous outbred population developed for genetic studies of complex traits. Genetic mapping studies in adult HS rats identified loci associated with cardiometabolic risks, such as glucose intolerance, insulin resistance, and increased body mass index. This study determined underappreciated metabolic health traits and the associated endocrine glands within available substrains of the HS rat founders. We hypothesize that the genetic diversity of the HS rat founder strains causes a range of endocrine health conditions contributing to the diversity of cardiometabolic disease risks. ACI/EurMcwi, BN/NHsdMcwi, BUF/MnaMcwi, F344/StmMcwi, M520/NRrrcMcwi, and WKY/NCrl rats of both sexes were studied from birth until 13 weeks of age. Birth weight was recorded, body weight was measured weekly, metabolic characteristics were assessed, and blood and tissues were collected. Our data show wide variation in endocrine traits and metabolic health states in ACI, BN, BUF, F344, M520, and WKY rat strains. This is the first report to compare birth weight, resting metabolic rate, endocrine gland weight, hypothalamic-pituitary-thyroid axis hormones, and brown adipose tissue weight in these rat strains. Importantly, this work unveils new potential for the HS rat population to model early life adversity and adrenal and thyroid pathophysiology. The HS population likely inherited risk alleles for these strain-specific traits, making the HS rat a powerful model to investigate interventions on endocrine and metabolic health.
Collapse
Affiliation(s)
- Valerie A Wagner
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Katie L Holl
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Karen C Clark
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - John J Reho
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Comprehensive Rodent Metabolic Phenotyping Core, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Hans-Joachim Lehmler
- Department of Occupational and Environmental Health, University of Iowa, Iowa City, IA 52242, USA
| | - Kai Wang
- Department of Biostatistics, University of Iowa, Iowa City, IA 52242, USA
| | - Justin L Grobe
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Comprehensive Rodent Metabolic Phenotyping Core, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Melinda R Dwinell
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Hershel Raff
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Endocrine Research Laboratory, Aurora St. Luke's Medical Center, Advocate Aurora Research Institute, Milwaukee, WI 53233, USA
| | - Anne E Kwitek
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Rat Genome Database, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
20
|
Kvandova M, Puzserova A, Balis P. Sexual Dimorphism in Cardiometabolic Diseases: The Role of AMPK. Int J Mol Sci 2023; 24:11986. [PMID: 37569362 PMCID: PMC10418890 DOI: 10.3390/ijms241511986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/18/2023] [Accepted: 07/24/2023] [Indexed: 08/13/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of mortality and disability among both males and females. The risk of cardiovascular diseases is heightened by the presence of a risk factor cluster of metabolic syndrome, covering obesity and obesity-related cardiometabolic risk factors such as hypertension, glucose, and lipid metabolism dysregulation primarily. Sex hormones contribute to metabolic regulation and make women and men susceptible to obesity development in a different manner, which necessitates sex-specific management. Identifying crucial factors that protect the cardiovascular system is essential to enhance primary and secondary prevention of cardiovascular diseases and should be explicitly studied from the perspective of sex differences. It seems that AMP-dependent protein kinase (AMPK) may be such a factor since it has the protective role of AMPK in the cardiovascular system, has anti-diabetic properties, and is regulated by sex hormones. Those findings highlight the potential cardiometabolic benefits of AMPK, making it an essential factor to consider. Here, we review information about the cross-talk between AMPK and sex hormones as a critical point in cardiometabolic disease development and progression and a target for therapeutic intervention in human disease.
Collapse
Affiliation(s)
- Miroslava Kvandova
- Institute of Normal and Pathological Physiology, Centre of Experimental Medicine, Slovak Academy of Sciences, 841 04 Bratislava, Slovakia; (A.P.); (P.B.)
| | | | | |
Collapse
|
21
|
Laupsa-Borge J, Grytten E, Bohov P, Bjørndal B, Strand E, Skorve J, Nordrehaug JE, Berge RK, Rostrup E, Mellgren G, Dankel SN, Nygård OK. Sex-specific responses in glucose-insulin homeostasis and lipoprotein-lipid components after high-dose supplementation with marine n-3 PUFAs in abdominal obesity: a randomized double-blind crossover study. Front Nutr 2023; 10:1020678. [PMID: 37404855 PMCID: PMC10315503 DOI: 10.3389/fnut.2023.1020678] [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: 08/16/2022] [Accepted: 06/01/2023] [Indexed: 07/06/2023] Open
Abstract
Background Clinical studies on effects of marine-derived omega-3 (n-3) polyunsaturated fatty acids (PUFAs), mainly eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), and the plant-derived omega-6 (n-6) PUFA linoleic acid (LA) on lipoprotein-lipid components and glucose-insulin homeostasis have shown conflicting results, which may partly be explained by differential responses in females and males. However, we have lacked data on sexual dimorphism in the response of cardiometabolic risk markers following increased consumption of n-3 or n-6 PUFAs. Objective To explore sex-specific responses after n-3 (EPA + DHA) or n-6 (LA) PUFA supplementation on circulating lipoprotein subfractions, standard lipids, apolipoproteins, fatty acids in red blood cell membranes, and markers of glycemic control/insulin sensitivity among people with abdominal obesity. Methods This was a randomized double-blind crossover study with two 7-week intervention periods separated by a 9-week washout phase. Females (n = 16) were supplemented with 3 g/d of EPA + DHA (fish oil) or 15 g/d of LA (safflower oil), while males (n = 23) received a dose of 4 g/d of EPA + DHA or 20 g/d of LA. In fasting blood samples, we measured lipoprotein particle subclasses, standard lipids, apolipoproteins, fatty acid profiles, and markers of glycemic control/insulin sensitivity. Results The between-sex difference in relative change scores was significant after n-3 for total high-density lipoproteins (females/males: -11%*/-3.3%, p = 0.036; *: significant within-sex change), high-density lipoprotein particle size (+2.1%*/-0.1%, p = 0.045), and arachidonic acid (-8.3%*/-12%*, p = 0.012), and after n-6 for total (+37%*/+2.1%, p = 0.041) and small very-low-density lipoproteins (+97%*/+14%, p = 0.021), and lipoprotein (a) (-16%*/+0.1%, p = 0.028). Circulating markers of glucose-insulin homeostasis differed significantly after n-3 for glucose (females/males: -2.1%/+3.9%*, p = 0.029), insulin (-31%*/+16%, p < 0.001), insulin C-peptide (-12%*/+13%*, p = 0.001), homeostasis model assessment of insulin resistance index 2 (-12%*/+14%*, p = 0.001) and insulin sensitivity index 2 (+14%*/-12%*, p = 0.001), and quantitative insulin sensitivity check index (+4.9%*/-3.4%*, p < 0.001). Conclusion We found sex-specific responses after high-dose n-3 (but not n-6) supplementation in circulating markers of glycemic control/insulin sensitivity, which improved in females but worsened in males. This may partly be related to the sex differences we observed in several components of the lipoprotein-lipid profile following the n-3 intervention. Clinical trial registration https://clinicaltrials.gov/, identifier [NCT02647333].
Collapse
Affiliation(s)
- Johnny Laupsa-Borge
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Elise Grytten
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Pavol Bohov
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Bodil Bjørndal
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Elin Strand
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Jon Skorve
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Jan Erik Nordrehaug
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| | - Rolf K. Berge
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Espen Rostrup
- Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| | - Gunnar Mellgren
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
- Mohn Research Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Simon N. Dankel
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
- Mohn Research Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Ottar K. Nygård
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
22
|
Lombrea A, Romanescu M, Jianu N, Andor M, Suciu M, Man DE, Danciu C, Dehelean CA, Buda V. Sex-Related Differences in the Pharmacological Response in SARS-CoV-2 Infection, Dyslipidemia, and Diabetes Mellitus: A Narrative Review. Pharmaceuticals (Basel) 2023; 16:853. [PMID: 37375800 DOI: 10.3390/ph16060853] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 05/26/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Pharmacological responses vary by sex in several illnesses. This narrative review summarizes sex variations in pharmaceutical response in SARS-CoV-2 infection, dyslipidemia, and diabetes mellitus. Infection with SARS-CoV-2 is more severe and deadly in men than women. This may be attributed to immunological responses, genetics, and hormones. Some research shows that men may respond better to genomic vaccinations and females to antiviral medications such as remdesivir (Moderna and Pfizer-BioNTech). In dyslipidemia, women tend to have greater HDL-C and lower LDL-C than men. Some studies show that females may need lower statin dosages than men to obtain equal LDL-C reductions. Ezetimibe co-administered with a statin significantly improved lipid profile indicators in men compared to women. Statins reduce dementia risk. Atorvastatin decreased dementia risk in males (adjusted HR 0.92, 95% CI 0.88-0.97), whereas lovastatin lowered dementia risk in women (HR 0.74, 95% CI 0.58-0.95). In diabetes mellitus, evidence suggests that females may have a higher risk of developing certain complications such as diabetic retinopathy and neuropathy, despite having lower rates of cardiovascular disease than males. This could be the result of differences in hormonal influences and genetic factors. Some research shows females may respond better to oral hypoglycemic medications such as metformin. In conclusion, sex-related differences in pharmacological response have been observed in SARS-CoV-2 infection, dyslipidemia, and diabetes mellitus. Further research is needed to better understand these differences and to develop personalized treatment strategies for males and females with these conditions.
Collapse
Affiliation(s)
- Adelina Lombrea
- Doctoral School, "Victor Babeş" University of Medicine and Pharmacy, 2 Eftimie Murgu Street, 300041 Timisoara, Romania
- Research Center for Pharmaco-Toxicological Evaluation, "Victor Babeş" University of Medicine and Pharmacy, 2 Eftimie Murgu Street, 300041 Timisoara, Romania
| | - Mirabela Romanescu
- Doctoral School, "Victor Babeş" University of Medicine and Pharmacy, 2 Eftimie Murgu Street, 300041 Timisoara, Romania
| | - Narcisa Jianu
- Doctoral School, "Victor Babeş" University of Medicine and Pharmacy, 2 Eftimie Murgu Street, 300041 Timisoara, Romania
| | - Minodora Andor
- Faculty of Medicine, "Victor Babeş" University of Medicine and Pharmacy, 2 Eftimie Murgu Street, 300041 Timisoara, Romania
- Multidisciplinary Heart Research Center, "Victor Babeş" University of Medicine and Pharmacy, 2 Eftimie Murgu Street, 340001 Timisoara, Romania
| | - Maria Suciu
- Research Center for Pharmaco-Toxicological Evaluation, "Victor Babeş" University of Medicine and Pharmacy, 2 Eftimie Murgu Street, 300041 Timisoara, Romania
- Faculty of Pharmacy, "Victor Babeş" University of Medicine and Pharmacy, 2 Eftimie Murgu Street, 300041 Timisoara, Romania
| | - Dana Emilia Man
- Faculty of Medicine, "Victor Babeş" University of Medicine and Pharmacy, 2 Eftimie Murgu Street, 300041 Timisoara, Romania
| | - Corina Danciu
- Research Center for Pharmaco-Toxicological Evaluation, "Victor Babeş" University of Medicine and Pharmacy, 2 Eftimie Murgu Street, 300041 Timisoara, Romania
- Faculty of Pharmacy, "Victor Babeş" University of Medicine and Pharmacy, 2 Eftimie Murgu Street, 300041 Timisoara, Romania
| | - Cristina Adriana Dehelean
- Research Center for Pharmaco-Toxicological Evaluation, "Victor Babeş" University of Medicine and Pharmacy, 2 Eftimie Murgu Street, 300041 Timisoara, Romania
- Faculty of Pharmacy, "Victor Babeş" University of Medicine and Pharmacy, 2 Eftimie Murgu Street, 300041 Timisoara, Romania
| | - Valentina Buda
- Research Center for Pharmaco-Toxicological Evaluation, "Victor Babeş" University of Medicine and Pharmacy, 2 Eftimie Murgu Street, 300041 Timisoara, Romania
- Faculty of Pharmacy, "Victor Babeş" University of Medicine and Pharmacy, 2 Eftimie Murgu Street, 300041 Timisoara, Romania
- Ineu City Hospital, 2 Republicii Street, 315300 Ineu, Romania
| |
Collapse
|
23
|
Suwanno J, Phonphet C, Mayurapak C, Ninla-Aesong P, Thiamwong L. Sex-based differences in risk of cardiovascular disease development and cardiovascular risk factors among individuals with hypertension: A cross-sectional study from primary care facilities. JOURNAL OF VASCULAR NURSING 2023; 41:62-71. [PMID: 37356872 DOI: 10.1016/j.jvn.2023.04.002] [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: 02/09/2022] [Revised: 04/10/2023] [Accepted: 04/10/2023] [Indexed: 06/27/2023]
Abstract
BACKGROUND Over the last two decades, the understanding of cardiovascular disease (CVD) has expanded in Asian countries. Despite this progress, there have been limited investigations into sex-based differences in the development of CVD and cardiovascular risk factors (CVRFs). AIM We investigated whether males and females with hypertension had different risks of developing CVD and CVRFs. METHODS We used a stratified multi-stage sampling design involving 15 primary care centers in Thailand. We recruited 1,448 individuals aged 35-74 years old. The Framingham cardiovascular risk algorithm was used to determine the risk of CVD development. RESULTS Female patients were overall more likely to have lower CVD risk scores. However, they demonstrated higher scores in the moderate-risk (p < 0.001) and high-risk (p < 0.001) groups as compared with males. One in four females was at a high risk of developing CVD. Females had higher rates of all CVRFs against males across sub-risk groups, with the highest odds ratio observed in the high-risk group, which persisted after adjusting for covariations. Overall, female patients had higher rates of diabetes, hyperlipidemia, obesity, and abdominal obesity as compared with males. Females in the overall group had a lower prevalence of uncontrolled hypertension than males, in contrast to the high-risk group. Female patients also had a lower prevalence of cigarette smoking and alcohol consumption than males. CONCLUSION There is evidence of sex-based differences in the risk of CVD development in hypertensive individuals. The interaction of CVRFs with a high risk of developing CVD was noted in females.
Collapse
Affiliation(s)
- Jom Suwanno
- School of Nursing, Walailak University, Nakhon Si Thammarat, 80160, Thailand; The Excellent Center of Community Health Promotion of Walailak University, Thailand.
| | - Chennet Phonphet
- School of Nursing, Walailak University, Nakhon Si Thammarat, 80160, Thailand; The Excellent Center of Community Health Promotion of Walailak University, Thailand
| | - Chidchanog Mayurapak
- School of Nursing, Walailak University, Nakhon Si Thammarat, 80160, Thailand; The Excellent Center of Community Health Promotion of Walailak University, Thailand
| | | | - Ladda Thiamwong
- College of Nursing, University of Central Florida, Orlando, FL 32826 USA
| |
Collapse
|
24
|
Vandenberg LN, Zoeller RT, Prins GS, Trasande L. Evaluating adverse effects of environmental agents in food: a brief critique of the US FDA's criteria. Environ Health 2023; 22:38. [PMID: 37085808 PMCID: PMC10120250 DOI: 10.1186/s12940-023-00971-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 02/12/2023] [Indexed: 05/03/2023]
Abstract
BACKGROUND In the US, the Food and Drug Administration (US FDA) is charged with protecting the safety of food from both pathogens and chemicals used in food production and food packaging. To protect the public in a transparent manner, the FDA needs to have an operational definition of what it considers to be an "adverse effect" so that it can take action against harmful agents. The FDA has recently published two statements where, for the first time, it defines the characteristics of an adverse effect that it uses to interpret toxicity studies. OBJECTIVE In this brief review, we examine two recent actions by the FDA, a proposed rule regarding a color additive used in vegetarian burgers and a decision not to recall fish with high levels of scombrotoxin. We evaluated the FDA's description of the criteria used to determine which outcomes should be considered adverse. OVERVIEW We describe three reasons why the FDA's criteria for "adverse effects" is not public health protective. These include an unscientific requirement for a monotonic dose response, which conflates hazard assessment and dose response assessment while also ignoring evidence for non-linear and non-monotonic effects for many environmental agents; a requirement that the effect be observed in both sexes, which fails to acknowledge the many sex- and gender-specific effects on physiology, disease incidence and severity, and anatomy; and a requirement that the effects are irreversible, which does not acknowledge the role of exposure timing or appreciate transgenerational effects that have been demonstrated for environmental chemicals. CONCLUSIONS The FDA's criteria for identifying adverse effects are inadequate because they are not science-based. Addressing this is important, because the acknowledgement of adverse effects is central to regulatory decisions and the protection of public health.
Collapse
Affiliation(s)
- Laura N Vandenberg
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts - Amherst, 240G Goessmann, 686 N. Pleasant Street, Amherst, MA, 01003, USA.
| | - R Thomas Zoeller
- Department of Biology, University of Massachusetts Amherst, Amherst, MA, USA
- Visiting Professor, Örebro University, Örebro, Sweden
| | - Gail S Prins
- Department of Urology, School of Medicine, Division of Epidemiology & Biostatistics, School of Public Health, University of Illinois at Chicago, Chicago, USA
| | - Leonardo Trasande
- Departments of Pediatrics, Environmental Medicine, and Population Health, New York University School of Medicine, New York, NY, USA
| |
Collapse
|
25
|
Sandoval DA, Patti ME. Glucose metabolism after bariatric surgery: implications for T2DM remission and hypoglycaemia. Nat Rev Endocrinol 2023; 19:164-176. [PMID: 36289368 PMCID: PMC10805109 DOI: 10.1038/s41574-022-00757-5] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/28/2022] [Indexed: 11/09/2022]
Abstract
Although promising therapeutics are in the pipeline, bariatric surgery (also known as metabolic surgery) remains our most effective strategy for the treatment of obesity and type 2 diabetes mellitus (T2DM). Of the many available options, Roux-en-Y gastric bypass (RYGB) and vertical sleeve gastrectomy (VSG) are currently the most widely used procedures. RYGB and VSG have very different anatomical restructuring but both surgeries are effective, to varying degrees, at inducing weight loss and T2DM remission. Both weight loss-dependent and weight loss-independent alterations in multiple tissues (such as the intestine, liver, pancreas, adipose tissue and skeletal muscle) yield net improvements in insulin resistance, insulin secretion and insulin-independent glucose metabolism. In a subset of patients, post-bariatric hypoglycaemia can develop months to years after surgery, potentially reflecting the extreme effects of potent glucose reduction after surgery. This Review addresses the effects of bariatric surgery on glucose regulation and the potential mechanisms responsible for both the resolution of T2DM and the induction of hypoglycaemia.
Collapse
Affiliation(s)
- Darleen A Sandoval
- Department of Paediatrics, Section of Nutrition, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | | |
Collapse
|
26
|
Khodursky S, Jiang CS, Zheng EB, Vaughan R, Schrider DR, Zhao L. Sex differences in interindividual gene expression variability across human tissues. PNAS NEXUS 2022; 1:pgac243. [PMID: 36712323 PMCID: PMC9802459 DOI: 10.1093/pnasnexus/pgac243] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 10/21/2022] [Indexed: 11/07/2022]
Abstract
Understanding phenotypic sex differences has long been a goal of biology from both a medical and evolutionary perspective. Although much attention has been paid to mean differences in phenotype between the sexes, little is known about sex differences in phenotypic variability. To gain insight into sex differences in interindividual variability at the molecular level, we analyzed RNA-seq data from 43 tissues from the Genotype-Tissue Expression project (GTEx). Within each tissue, we identified genes that show sex differences in gene expression variability. We found that these sex-differentially variable (SDV) genes are associated with various important biological functions, including sex hormone response, immune response, and other signaling pathways. By analyzing single-cell RNA sequencing data collected from breast epithelial cells, we found that genes with sex differences in gene expression variability in breast tissue tend to be expressed in a cell-type-specific manner. We looked for an association between SDV expression and Graves' disease, a well-known heavily female-biased disease, and found a significant enrichment of Graves' associated genes among genes with higher variability in females in thyroid tissue. This suggests a possible role for SDV expression in sex-biased disease. We then examined the evolutionary constraints acting on genes with sex differences in variability and found that they exhibit evidence of increased selective constraint. Through analysis of sex-biased eQTL data, we found evidence that SDV expression may have a genetic basis. Finally, we propose a simple evolutionary model for the emergence of SDV expression from sex-specific constraints.
Collapse
Affiliation(s)
- Samuel Khodursky
- Laboratory of Evolutionary Genetics and Genomics, The Rockefeller University, New York, NY 10065, USA
| | - Caroline S Jiang
- Department of Biostatistics, The Rockefeller University, New York, NY 10065, USA
| | - Eric B Zheng
- Laboratory of Evolutionary Genetics and Genomics, The Rockefeller University, New York, NY 10065, USA
| | - Roger Vaughan
- Department of Biostatistics, The Rockefeller University, New York, NY 10065, USA
| | - Daniel R Schrider
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Li Zhao
- Laboratory of Evolutionary Genetics and Genomics, The Rockefeller University, New York, NY 10065, USA
| |
Collapse
|
27
|
Emiliano AB, Lopatinsky NR, Kraljević M, Higuchi S, He Y, Haeusler RA, Schwartz GJ. Sex-specific differences in metabolic outcomes after sleeve gastrectomy and intermittent fasting in obese middle-aged mice. Am J Physiol Endocrinol Metab 2022; 323:E107-E121. [PMID: 35658544 PMCID: PMC9273270 DOI: 10.1152/ajpendo.00017.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 05/19/2022] [Accepted: 05/19/2022] [Indexed: 01/21/2023]
Abstract
Despite the high prevalence of obesity among middle-aged subjects, it is unclear if sex differences in middle age affect the metabolic outcomes of obesity therapies. Accordingly, in this study, middle-aged obese female and male mice were randomized to one of three groups: sleeve gastrectomy (SG), sham surgery ad libitum (SH-AL), or sham surgery with weight matching to SG through intermittent fasting with calorie restriction (SH-IF). Comprehensive measures of energy and glucose homeostasis, including energy intake, body weight, energy expenditure, glucose and insulin tolerance, and interscapular brown adipose tissue (iBAT) sympathetic innervation density were obtained. At the end of 8 wk, SG and SH-IF females had better metabolic outcomes than their male counterparts. SG females had improved weight loss maintenance, preservation of fat-free mass (FFM), higher total energy expenditure (TEE), normal locomotor activity, and reduced plasma insulin and white adipose tissue (WAT) inflammatory markers. SH-IF females also had lower plasma insulin and WAT inflammatory markers, and higher TEE than SH-IF males, despite their lower FFM. In addition, SH-IF females had higher iBAT sympathetic nerve density than SG and SH-AL females, whereas there were no differences among males. Notably, SH-IF mice of both sexes had the most improved glucose tolerance, highlighting the benefits of fasting, irrespective of weight loss. Results from this study demonstrate that in middle-aged obese mice, female sex is associated with better metabolic outcomes after SG or IF with calorie restriction. Clinical studies are needed to determine if sex differences should guide the choice of obesity therapies.NEW & NOTEWORTHY SG or IF with calorie restriction produces better metabolic outcomes in females than in males. IF with calorie restriction prevents metabolic adaptation, even in the face of fat-free mass loss. IF with calorie restriction in females only, is associated with increased iBAT sympathetic innervation, which possibly mitigates reductions in energy expenditure secondary to fat-free mass loss. Lastly, IF leads to better glucose homeostasis than SG, irrespective of sex.
Collapse
Affiliation(s)
| | | | - Marko Kraljević
- Columbia University Medical Center, New York, New York
- Clarunis University Center for Gastrointestinal and Liver Diseases, St. Clara Hospital and University Hospital, Basel, Switzerland
| | - Sei Higuchi
- Columbia University Medical Center, New York, New York
| | - Ying He
- Columbia University Medical Center, New York, New York
| | | | | |
Collapse
|
28
|
Dwaib HS, AlZaim I, Ajouz G, Eid AH, El-Yazbi A. Sex Differences in Cardiovascular Impact of Early Metabolic Impairment: Interplay between Dysbiosis and Adipose Inflammation. Mol Pharmacol 2022; 102:481-500. [PMID: 34732528 DOI: 10.1124/molpharm.121.000338] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 10/23/2021] [Indexed: 11/22/2022] Open
Abstract
The evolving view of gut microbiota has shifted toward describing the colonic flora as a dynamic organ in continuous interaction with systemic physiologic processes. Alterations of the normal gut bacterial profile, known as dysbiosis, has been linked to a wide array of pathologies. Of particular interest is the cardiovascular-metabolic disease continuum originating from positive energy intake and high-fat diets. Accumulating evidence suggests a role for sex hormones in modulating the gut microbiome community. Such a role provides an additional layer of modulation of the early inflammatory changes culminating in negative metabolic and cardiovascular outcomes. In this review, we will shed the light on the role of sex hormones in cardiovascular dysfunction mediated by high-fat diet-induced dysbiosis, together with the possible involvement of insulin resistance and adipose tissue inflammation. Insights into novel therapeutic interventions will be discussed as well. SIGNIFICANCE STATEMENT: Increasing evidence implicates a role for dysbiosis in the cardiovascular complications of metabolic dysfunction. This minireview summarizes the available data on the sex-based differences in gut microbiota alterations associated with dietary patterns leading to metabolic impairment. A role for a differential impact of adipose tissue inflammation across sexes in mediating the cardiovascular detrimental phenotype following diet-induced dysbiosis is proposed. Better understanding of this pathway will help introduce early approaches to mitigate cardiovascular deterioration in metabolic disease.
Collapse
Affiliation(s)
- Haneen S Dwaib
- Department of Pharmacology and Toxicology, Faculty of Medicine (H.S.D., I.A., G.A., A.E.-Y.), Department of Nutrition and Food Sciences, Faculty of Agricultural and Food Sciences (H.S.D.), American University of Beirut, Beirut, Lebanon; Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon (I.A.); Department of Basic Medical Sciences, College of Medicine (A.H.E.), Biomedical and Pharmaceutical Research Unit, QU Health (A.H.E.), Qatar University, Doha, Qatar; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt (A.E.-Y.); and Faculty of Pharmacy, Alalamein International University, Alalamein, Egypt (A.E.-Y.)
| | - Ibrahim AlZaim
- Department of Pharmacology and Toxicology, Faculty of Medicine (H.S.D., I.A., G.A., A.E.-Y.), Department of Nutrition and Food Sciences, Faculty of Agricultural and Food Sciences (H.S.D.), American University of Beirut, Beirut, Lebanon; Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon (I.A.); Department of Basic Medical Sciences, College of Medicine (A.H.E.), Biomedical and Pharmaceutical Research Unit, QU Health (A.H.E.), Qatar University, Doha, Qatar; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt (A.E.-Y.); and Faculty of Pharmacy, Alalamein International University, Alalamein, Egypt (A.E.-Y.)
| | - Ghina Ajouz
- Department of Pharmacology and Toxicology, Faculty of Medicine (H.S.D., I.A., G.A., A.E.-Y.), Department of Nutrition and Food Sciences, Faculty of Agricultural and Food Sciences (H.S.D.), American University of Beirut, Beirut, Lebanon; Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon (I.A.); Department of Basic Medical Sciences, College of Medicine (A.H.E.), Biomedical and Pharmaceutical Research Unit, QU Health (A.H.E.), Qatar University, Doha, Qatar; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt (A.E.-Y.); and Faculty of Pharmacy, Alalamein International University, Alalamein, Egypt (A.E.-Y.)
| | - Ali H Eid
- Department of Pharmacology and Toxicology, Faculty of Medicine (H.S.D., I.A., G.A., A.E.-Y.), Department of Nutrition and Food Sciences, Faculty of Agricultural and Food Sciences (H.S.D.), American University of Beirut, Beirut, Lebanon; Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon (I.A.); Department of Basic Medical Sciences, College of Medicine (A.H.E.), Biomedical and Pharmaceutical Research Unit, QU Health (A.H.E.), Qatar University, Doha, Qatar; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt (A.E.-Y.); and Faculty of Pharmacy, Alalamein International University, Alalamein, Egypt (A.E.-Y.)
| | - Ahmed El-Yazbi
- Department of Pharmacology and Toxicology, Faculty of Medicine (H.S.D., I.A., G.A., A.E.-Y.), Department of Nutrition and Food Sciences, Faculty of Agricultural and Food Sciences (H.S.D.), American University of Beirut, Beirut, Lebanon; Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon (I.A.); Department of Basic Medical Sciences, College of Medicine (A.H.E.), Biomedical and Pharmaceutical Research Unit, QU Health (A.H.E.), Qatar University, Doha, Qatar; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt (A.E.-Y.); and Faculty of Pharmacy, Alalamein International University, Alalamein, Egypt (A.E.-Y.)
| |
Collapse
|
29
|
DeVeaux SA, Ogle ME, Vyshnya S, Chiappa NF, Leitmann B, Rudy R, Day A, Mortensen LJ, Kurtzberg J, Roy K, Botchwey EA. Characterizing human mesenchymal stromal cells' immune-modulatory potency using targeted lipidomic profiling of sphingolipids. Cytotherapy 2022; 24:608-618. [PMID: 35190267 PMCID: PMC10725732 DOI: 10.1016/j.jcyt.2021.12.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 11/29/2021] [Accepted: 12/06/2021] [Indexed: 12/17/2022]
Abstract
Cell therapies are expected to increase over the next decade owing to increasing demand for clinical applications. Mesenchymal stromal cells (MSCs) have been explored to treat a number of diseases, with some successes in early clinical trials. Despite early successes, poor MSC characterization results in lessened therapeutic capacity once in vivo. Here, we characterized MSCs derived from bone marrow (BM), adipose tissue and umbilical cord tissue for sphingolipids (SLs), a class of bioactive lipids, using liquid chromatography/tandem mass spectrometry. We found that ceramide levels differed based on the donor's sex in BM-MSCs. We detected fatty acyl chain variants in MSCs from all three sources. Linear discriminant analysis revealed that MSCs separated based on tissue source. Principal component analysis showed that interferon-γ-primed and unstimulated MSCs separated according to their SL signature. Lastly, we detected higher ceramide levels in low indoleamine 2,3-dioxygenase MSCs, indicating that sphingomyelinase or ceramidase enzymatic activity may be involved in their immune potency.
Collapse
Affiliation(s)
- S’Dravious A. DeVeaux
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory, Atlanta, GA
- Petit Institute of Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA
| | - Molly E. Ogle
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory, Atlanta, GA
- Petit Institute of Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA
| | - Sofiya Vyshnya
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory, Atlanta, GA
- Petit Institute of Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA
| | - Nathan F. Chiappa
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory, Atlanta, GA
- Petit Institute of Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA
| | - Bobby Leitmann
- Regenerative Bioscience Center, Rhodes Center for ADS, University of Georgia, Athens, GA
- School of Chemical, Materials and Biomedical Engineering, University of Georgia, Athens, GA
| | - Ryan Rudy
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory, Atlanta, GA
- Petit Institute of Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA
| | - Abigail Day
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory, Atlanta, GA
- Petit Institute of Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA
| | - Luke J. Mortensen
- Regenerative Bioscience Center, Rhodes Center for ADS, University of Georgia, Athens, GA
- School of Chemical, Materials and Biomedical Engineering, University of Georgia, Athens, GA
| | - Joanne Kurtzberg
- Marcus Center for Cellular Cures, Duke University School of Medicine, Durham, NC
| | - Krishnendu Roy
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory, Atlanta, GA
- Marcus Center for Therapeutic Cell Characterization and Manufacturing, Georgia Institute of Technology, Atlanta, GA
- NSF Engineering Research Center (ERC) for Cell Manufacturing Technologies (CMaT), Georgia Institute of Technology, Atlanta, GA
| | - Edward A. Botchwey
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory, Atlanta, GA
- Petit Institute of Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA
| |
Collapse
|
30
|
Prasetyo M, Andreas S, Sunardi D, Prihartono J, Imanuel Setiawan S, Christian A. Ultrasonographic measurement of abdominal and gluteal-femoral fat thickness as a predictor for android/gynoid ratio. Eur J Radiol 2022; 154:110387. [PMID: 35660916 DOI: 10.1016/j.ejrad.2022.110387] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 04/29/2022] [Accepted: 05/26/2022] [Indexed: 11/03/2022]
Abstract
OBJECTIVE To evaluate the use of ultrasonography (US) as an alternative to dual-energy x-ray absorptiometry (DXA) to predict the percentage ratio of android/gynoid (A/G) fat mass. METHODS This was a cross-sectional study. Twenty-eight participants included in the study underwent whole-body DXA examinations and the A/G ratio was calculated. Soft-tissue US was performed in several standardised anthropometric areas of the body. Correlation analysis between abdominal and gluteal-femoral fat thickness based on US and A/G ratio was conducted using the Pearson or Spearman test depending on the data normality. Multiple regression analysis using the backward stepwise method was performed to establish an equation for estimating the A/G ratio. RESULTS There was a strong and significant correlation between fat thickness in the six anthropometric areas and the A/G ratio in female participants. The analysis revealed three anthropometric areas: upper abdomen (S4), lower abdomen (S5), and mid-xiphoid-umbilical region (S7), that can accurately predict the A/G ratio by 82.3%. (P < 0.05). However, no such correlation was found in male participants. CONCLUSIONS US measurement of fat thickness can predict A/G ratio in the female population. However, this method is not recommended for men.
Collapse
Affiliation(s)
- Marcel Prasetyo
- Department of Radiology, Faculty of Medicine Universitas Indonesia - Dr. Cipto Mangunkusumo National Central General Hospital, Jakarta, Indonesia.
| | - Steven Andreas
- Department of Radiology, Faculty of Medicine Universitas Indonesia - Dr. Cipto Mangunkusumo National Central General Hospital, Jakarta, Indonesia
| | - Diana Sunardi
- Department of Clinical Nutrition, Faculty of Medicine Universitas Indonesia - Dr. Cipto Mangunkusumo National Central General Hospital, Jakarta, Indonesia
| | - Joedo Prihartono
- Department of Community Medicine, Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia
| | - Stefanus Imanuel Setiawan
- Department of Radiology, Faculty of Medicine Universitas Indonesia - Dr. Cipto Mangunkusumo National Central General Hospital, Jakarta, Indonesia
| | - Andreas Christian
- Department of Radiology, Faculty of Medicine Universitas Indonesia - Dr. Cipto Mangunkusumo National Central General Hospital, Jakarta, Indonesia
| |
Collapse
|
31
|
Global Proteome Profiling of the Temporal Cortex of Female Rats Exposed to Chronic Stress and the Western Diet. Nutrients 2022; 14:nu14091934. [PMID: 35565902 PMCID: PMC9103025 DOI: 10.3390/nu14091934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 04/25/2022] [Accepted: 05/02/2022] [Indexed: 11/16/2022] Open
Abstract
The increasing consumption of highly processed foods with high amounts of saturated fatty acids and simple carbohydrates is a major contributor to the burden of overweight and obesity. Additionally, an unhealthy diet in combination with chronic stress exposure is known to be associated with the increased prevalence of central nervous system diseases. In the present study, the global brain proteome approach was applied to explore protein alterations after exposure to the Western diet and/or stress. Female adult rats were fed with the Western diet with human snacks and/or subjected to chronic stress induced by social instability for 12 weeks. The consumption of the Western diet resulted in an obese phenotype and induced changes in the serum metabolic parameters. Consuming the Western diet resulted in changes in only 5.4% of the proteins, whereas 48% of all detected proteins were affected by chronic stress, of which 86.3% were down-regulated due to this exposure to chronic stress. However, feeding with a particular diet modified stress-induced changes in the brain proteome. The down-regulation of proteins involved in axonogenesis and mediating the synaptic clustering of AMPA glutamate receptors (Nptx1), as well as proteins related to metabolic processes (Atp5i, Mrps36, Ndufb4), were identified, while increased expression was detected for proteins involved in the development and differentiation of the CNS (Basp1, Cend1), response to stress, learning and memory (Prrt2), and modulation of synaptic transmission (Ncam1, Prrt2). In summary, global proteome analysis provides information about the impact of the combination of the Western diet and stress exposure on cerebrocortical protein alterations and yields insight into the underlying mechanisms and pathways involved in functional and morphological brain alterations as well as behavioral disturbances described in the literature.
Collapse
|
32
|
Renin–Angiotensin System in Liver Metabolism: Gender Differences and Role of Incretins. Metabolites 2022; 12:metabo12050411. [PMID: 35629915 PMCID: PMC9143858 DOI: 10.3390/metabo12050411] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/12/2022] [Accepted: 04/27/2022] [Indexed: 02/04/2023] Open
Abstract
The impaired hepatic lipids and carbohydrates metabolism result in various metabolic disorders, including obesity, diabetes, insulin resistance, hyperlipidemia and metabolic syndrome. The renin–angiotensin system (RAS) has been identified in the liver and it is now recognized as an important modulator of body metabolic processes. This review is intended to provide an update of the impact of the renin–angiotensin system on lipid and carbohydrate metabolism, regarding gender difference and prenatal undernutrition, specifically focused on the role of the liver. The discovery of angiotensin-converting enzyme 2 (ACE2) has renewed interest in the potential therapeutic role of RAS modulation. RAS is over activated in non-alcoholic fatty liver disease (NAFLD) and hepatocellular carcinoma. Glucagon-like peptide-1 (GLP-1) has been shown to modulate RAS. The GLP-I analogue liraglutide antagonizes hepatocellular steatosis and exhibits liver protection. Liraglutide has a negative effect on the ACE/AngII/AT1R axis and a positive impact on the ACE2/Ang(1-7)/Mas axis. Activation of the ACE2/Ang(1-7)/Mas counter-regulatory axis is able to prevent liver injuries. Angiotensin(1-7) and ACE2 shows more favorable effects on lipid homeostasis in males but there is a need to do more investigation in female models. Prenatal undernutrition exerts long-term effects in the liver of offspring and is associated with a number of metabolic and endocrine alterations. These findings provide a novel therapeutic regimen to prevent and treat many chronic diseases by accelerating the effect of the ACE2/Ang1-7/Mas axis and inhibiting the ACE/AngII/AT1R axis.
Collapse
|
33
|
Estrogenic Action in Stress-Induced Neuroendocrine Regulation of Energy Homeostasis. Cells 2022; 11:cells11050879. [PMID: 35269500 PMCID: PMC8909319 DOI: 10.3390/cells11050879] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/16/2022] [Accepted: 02/28/2022] [Indexed: 01/27/2023] Open
Abstract
Estrogens are among important contributing factors to many sex differences in neuroendocrine regulation of energy homeostasis induced by stress. Research in this field is warranted since chronic stress-related psychiatric and metabolic disturbances continue to be top health concerns, and sex differences are witnessed in these aspects. For example, chronic stress disrupts energy homeostasis, leading to negative consequences in the regulation of emotion and metabolism. Females are known to be more vulnerable to the psychological consequences of stress, such as depression and anxiety, whereas males are more vulnerable to the metabolic consequences of stress. Sex differences that exist in the susceptibility to various stress-induced disorders have led researchers to hypothesize that gonadal hormones are regulatory factors that should be considered in stress studies. Further, estrogens are heavily recognized for their protective effects on metabolic dysregulation, such as anti-obesogenic and glucose-sensing effects. Perturbations to energy homeostasis using laboratory rodents, such as physiological stress or over-/under- feeding dietary regimen prevalent in today’s society, offer hints to the underlying mechanisms of estrogenic actions. Metabolic effects of estrogens primarily work through estrogen receptor α (ERα), which is differentially expressed between the sexes in hypothalamic nuclei regulating energy metabolism and in extrahypothalamic limbic regions that are not typically associated with energy homeostasis. In this review, we discuss estrogenic actions implicated in stress-induced sex-distinct metabolic disorders.
Collapse
|
34
|
Chow AJ, Iverson R, Lamoureux M, Tingley K, Jordan I, Pallone N, Smith M, Al-Baldawi Z, Chakraborty P, Brehaut J, Chan A, Cohen E, Dyack S, Gillis LJ, Goobie S, Graham ID, Greenberg CR, Grimshaw JM, Hayeems RZ, Jain-Ghai S, Jolly A, Khangura S, MacKenzie JJ, Major N, Mitchell JJ, Nicholls SG, Pender A, Potter M, Prasad C, Prosser LA, Schulze A, Siriwardena K, Sparkes R, Speechley K, Stockler S, Taljaard M, Teitelbaum M, Trakadis Y, van Karnebeek C, Walia JS, Wilson BJ, Wilson K, Potter BK. Families' healthcare experiences for children with inherited metabolic diseases: protocol for a mixed methods cohort study. BMJ Open 2022; 12:e055664. [PMID: 35193919 PMCID: PMC8867352 DOI: 10.1136/bmjopen-2021-055664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
INTRODUCTION Children with inherited metabolic diseases (IMDs) often have complex and intensive healthcare needs and their families face challenges in receiving high-quality, family centred health services. Improvement in care requires complex interventions involving multiple components and stakeholders, customised to specific care contexts. This study aims to comprehensively understand the healthcare experiences of children with IMDs and their families across Canada. METHODS AND ANALYSIS A two-stage explanatory sequential mixed methods design will be used. Stage 1: quantitative data on healthcare networks and encounter experiences will be collected from 100 parent/guardians through a care map, 2 baseline questionnaires and 17 weekly diaries over 5-7 months. Care networks will be analysed using social network analysis. Relationships between demographic or clinical variables and ratings of healthcare experiences across a range of family centred care dimensions will be analysed using generalised linear regression. Other quantitative data related to family experiences and healthcare experiences will be summarised descriptively. Ongoing analysis of quantitative data and purposive, maximum variation sampling will inform sample selection for stage 2: a subset of stage 1 participants will participate in one-on-one videoconference interviews to elaborate on the quantitative data regarding care networks and healthcare experiences. Interview data will be analysed thematically. Qualitative and quantitative data will be merged during analysis to arrive at an enhanced understanding of care experiences. Quantitative and qualitative data will be combined and presented narratively using a weaving approach (jointly on a theme-by-theme basis) and visually in a side-by-side joint display. ETHICS AND DISSEMINATION The study protocol and procedures were approved by the Children's Hospital of Eastern Ontario's Research Ethics Board, the University of Ottawa Research Ethics Board and the research ethics boards of each participating study centre. Findings will be published in peer-reviewed journals and presented at scientific conferences.
Collapse
Affiliation(s)
- Andrea J Chow
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Ontario, Canada
| | - Ryan Iverson
- Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| | | | - Kylie Tingley
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Ontario, Canada
| | | | - Nicole Pallone
- Patient Partner, Canadian PKU & Allied Disorders Inc, Toronto, Ontario, Canada
| | - Maureen Smith
- Patient Partner, Canadian Organization for Rare Disorders, Toronto, Ontario, Canada
| | - Zobaida Al-Baldawi
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Ontario, Canada
| | - Pranesh Chakraborty
- Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
- Department of Pediatrics, University of Ottawa, Ottawa, Ontario, Canada
| | - Jamie Brehaut
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Ontario, Canada
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Alicia Chan
- Department of Medical Genetics, University of Alberta, Edmonton, Alberta, Canada
| | - Eyal Cohen
- Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada
- Child Health Evaluative Sciences, The Hospital for Sick Children, Toronto, Ontario, Canada
- Division of Pediatric Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Sarah Dyack
- Division of Medical Genetics, Department of Pediatrics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Lisa Jane Gillis
- Department of Medicine, Johns Hopkins All Children's Hospital, St Petersburg, Florida, USA
| | - Sharan Goobie
- Division of Medical Genetics, Department of Pediatrics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Ian D Graham
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Ontario, Canada
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Cheryl R Greenberg
- Rady Faculty of Health Sciences, Max Rady College of Medicine, Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, Manitoba, Canada
- Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| | - Jeremy M Grimshaw
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Robin Z Hayeems
- Child Health Evaluative Sciences, The Hospital for Sick Children, Toronto, Ontario, Canada
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, ON, Canada
| | - Shailly Jain-Ghai
- Department of Medical Genetics, University of Alberta, Edmonton, Alberta, Canada
| | - Ann Jolly
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Ontario, Canada
- Contagion Consulting Group, Ottawa, Ontario, Canada
| | - Sara Khangura
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Ontario, Canada
| | - Jennifer J MacKenzie
- Hamilton Health Sciences, McMaster Children's Hospital, Hamilton, Ontario, Canada
| | - Nathalie Major
- Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
- Department of Pediatrics, University of Ottawa, Ottawa, Ontario, Canada
| | - John J Mitchell
- Division of Pediatric Endocrinology, Department of Pediatrics, Montreal Children's Hospital, Montreal, Quebec, Canada
| | - Stuart G Nicholls
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Ottawa Methods Centre, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Amy Pender
- Hamilton Health Sciences, McMaster Children's Hospital, Hamilton, Ontario, Canada
| | - Murray Potter
- Hamilton Health Sciences, McMaster Children's Hospital, Hamilton, Ontario, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Chitra Prasad
- Department of Pediatrics, Western University, London, Ontario, Canada
| | - Lisa A Prosser
- Department of Pediatrics, Susan B. Meister Child Health Evaluation and Research Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Andreas Schulze
- Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
- Clinical and Metabolic Genetics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Komudi Siriwardena
- Department of Medical Genetics, University of Alberta, Edmonton, Alberta, Canada
| | - Rebecca Sparkes
- Departments of Medical Genetics and Pediatrics, Alberta Children's Hospital, Calgary, Alberta, Canada
| | - Kathy Speechley
- Department of Pediatrics, Western University, London, Ontario, Canada
| | - Sylvia Stockler
- Department of Pediatrics, BC Children's Hospital, Vancouver, British Columbia, Canada
| | - Monica Taljaard
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Ontario, Canada
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Mari Teitelbaum
- Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| | - Yannis Trakadis
- Department of Specialized Medicine, Division of Medical Genetics, McGill University Health Centre, Montreal, Quebec, Canada
| | - Clara van Karnebeek
- Department of Pediatrics, Radboud University Medical Center, Nijmegen, Gelderland, The Netherlands
- Department of Pediatrics, British Columbia Children's Hospital, Vancouver, British Columbia, Canada
| | - Jagdeep S Walia
- Medical Genetics, Department of Pediatrics, Kingston Health Sciences Centre, Kingston, Ontario, Canada
- Department of Pediatrics, Queen's University, Kingston, Ontario, Canada
| | - Brenda J Wilson
- Faculty of Medicine Division of Community Health and Humanities, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| | - Kumanan Wilson
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Ontario, Canada
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Bruyere Research Institute, Ottawa, Ontario, Canada
| | - Beth K Potter
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
35
|
Mills EL, Harmon C, Jedrychowski MP, Xiao H, Gruszczyk AV, Bradshaw GA, Tran N, Garrity R, Laznik-Bogoslavski D, Szpyt J, Prendeville H, Lynch L, Murphy MP, Gygi SP, Spiegelman BM, Chouchani ET. Cysteine 253 of UCP1 regulates energy expenditure and sex-dependent adipose tissue inflammation. Cell Metab 2022; 34:140-157.e8. [PMID: 34861155 PMCID: PMC8732317 DOI: 10.1016/j.cmet.2021.11.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 09/15/2021] [Accepted: 11/08/2021] [Indexed: 01/07/2023]
Abstract
Uncoupling protein 1 (UCP1) is a major regulator of brown and beige adipocyte energy expenditure and metabolic homeostasis. However, the widely employed UCP1 loss-of-function model has recently been shown to have a severe deficiency in the entire electron transport chain of thermogenic fat. As such, the role of UCP1 in metabolic regulation in vivo remains unclear. We recently identified cysteine-253 as a regulatory site on UCP1 that elevates protein activity upon covalent modification. Here, we examine the physiological importance of this site through the generation of a UCP1 cysteine-253-null (UCP1 C253A) mouse, a precise genetic model for selective disruption of UCP1 in vivo. UCP1 C253A mice exhibit significantly compromised thermogenic responses in both males and females but display no measurable effect on fat accumulation in an obesogenic environment. Unexpectedly, we find that a lack of C253 results in adipose tissue redox stress, which drives substantial immune cell infiltration and systemic inflammatory pathology in adipose tissues and liver of male, but not female, mice. Elevation of systemic estrogen reverses this male-specific pathology, providing a basis for protection from inflammation due to loss of UCP1 C253 in females. Together, our results establish the UCP1 C253 activation site as a regulator of acute thermogenesis and sex-dependent tissue inflammation.
Collapse
Affiliation(s)
- Evanna L Mills
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Cathal Harmon
- Department of Immunology, Harvard Medical School, Boston, MA, USA; Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital and Harvard Medical School, Boston, USA
| | - Mark P Jedrychowski
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Haopeng Xiao
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Anja V Gruszczyk
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge CB2 0XY, UK
| | - Gary A Bradshaw
- Harvard Program in Therapeutic Science, Harvard Medical School, Boston, MA, USA
| | - Nhien Tran
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Ryan Garrity
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - John Szpyt
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Hannah Prendeville
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Lydia Lynch
- Department of Immunology, Harvard Medical School, Boston, MA, USA; Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital and Harvard Medical School, Boston, USA; School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Michael P Murphy
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge CB2 0XY, UK; Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Bruce M Spiegelman
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Edward T Chouchani
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
36
|
Lei Z, Wu H, Yang Y, Hu Q, Lei Y, Liu W, Nie Y, Yang L, Zhang X, Yang C, Lin T, Tong F, Zhu J, Guo J. Dihydroartemisinin improves hypercholesterolemia in ovariectomized mice via enhancing vectorial transport of cholesterol and bile acids from blood to bile. Bioorg Med Chem 2022; 53:116520. [PMID: 34847494 DOI: 10.1016/j.bmc.2021.116520] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 11/07/2021] [Accepted: 11/16/2021] [Indexed: 12/12/2022]
Abstract
The increase of concentrations of total cholesterol (TC) and low-density lipoprotein cholesterol (LDL-C) in the serum of postmenopausal women is the important risk factor of the high morbidity of cardiovascular diseases of old women worldwide. To test the anti-hypercholesterolemia function of dihydroartemisinin (DHA) in postmenopausal women, ovariectomized (OVX) mice were generated, and DHA were administrated to OVX mice for 4 weeks. The blood and liver tissues were collected for biochemical and histological tests respectively. The mRNA and protein expression levels of genes related to metabolism and transport of cholesterol, bile acid and fatty acid in the liver or ileum were checked through qPCR and western blot. DHA could significantly reduce the high concentrations of TC and LDL-C in the serum and the lipid accumulation in the liver of ovariectomized mice. The expression of ABCG5/8 was reduced in liver of OVX mice, and DHA could up-regulate the expression of them. Genes of transport proteins for bile salt transport from blood to bile, including Slc10a1, Slco1b2 and Abcb11, were also significantly up-regulated by DHA. DHA also down-regulated the expression of Slc10a2 in the ileum of OVX mice to reduce the absorption of bile salts. Genes required for fatty acid synthesis and uptake, such as Fasn and CD36, were reduced in the liver of OVX mice, and DHA administration could significantly up-regulate the expression of them. These results demonstrated that DHA could improve hypercholesterolemia in OVX mice through enhancing the vectorial transport of cholesterol and bile acid from blood to bile.
Collapse
Affiliation(s)
- Zili Lei
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China; Institute of Chinese Medicine, Guangdong Pharmaceutical University; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, PR China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, PR China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, PR China.
| | - Huijuan Wu
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China; Institute of Chinese Medicine, Guangdong Pharmaceutical University; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, PR China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, PR China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, PR China; School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, Guangzhou 510006, PR China
| | - Yanhong Yang
- The First Affiliated Hospital (School of Clinical Medicine), Guangdong Pharmaceutical University, Nong-Lin-Xia Road 19(#), Yue-Xiu District, Guangzhou 510080, PR China
| | - Qing Hu
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China; Institute of Chinese Medicine, Guangdong Pharmaceutical University; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, PR China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, PR China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, PR China
| | - Yuting Lei
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China; Institute of Chinese Medicine, Guangdong Pharmaceutical University; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, PR China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, PR China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, PR China
| | - Wanwan Liu
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China; Institute of Chinese Medicine, Guangdong Pharmaceutical University; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, PR China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, PR China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, PR China
| | - Ya Nie
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China; Institute of Chinese Medicine, Guangdong Pharmaceutical University; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, PR China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, PR China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, PR China; School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, Guangzhou 510006, PR China
| | - Lanxiang Yang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China; Institute of Chinese Medicine, Guangdong Pharmaceutical University; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, PR China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, PR China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, PR China; School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, Guangzhou 510006, PR China
| | - Xueying Zhang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China; Institute of Chinese Medicine, Guangdong Pharmaceutical University; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, PR China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, PR China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, PR China; School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, Guangzhou 510006, PR China
| | - Changyuan Yang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China; Institute of Chinese Medicine, Guangdong Pharmaceutical University; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, PR China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, PR China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, PR China; School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, Guangzhou 510006, PR China
| | - Ting Lin
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China; Institute of Chinese Medicine, Guangdong Pharmaceutical University; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, PR China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, PR China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, PR China
| | - Fengxue Tong
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China; Institute of Chinese Medicine, Guangdong Pharmaceutical University; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, PR China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, PR China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, PR China
| | - Jiamin Zhu
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China; Institute of Chinese Medicine, Guangdong Pharmaceutical University; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, PR China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, PR China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, PR China
| | - Jiao Guo
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China; Institute of Chinese Medicine, Guangdong Pharmaceutical University; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, PR China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, PR China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, PR China.
| |
Collapse
|
37
|
Nowacka-Chmielewska MM, Liśkiewicz D, Grabowska K, Liśkiewicz A, Marczak Ł, Wojakowska A, Pondel N, Grabowski M, Barski JJ, Małecki A. Effects of Simultaneous Exposure to a Western Diet and Wheel-Running Training on Brain Energy Metabolism in Female Rats. Nutrients 2021; 13:nu13124242. [PMID: 34959794 PMCID: PMC8707360 DOI: 10.3390/nu13124242] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/18/2021] [Accepted: 11/24/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND In the pathogenesis of central nervous system disorders (e.g., neurodegenerative), an important role is attributed to an unhealthy lifestyle affecting brain energy metabolism. Physical activity in the prevention and treatment of lifestyle-related diseases is getting increasing attention. METHODS We performed a series of assessments in adult female Long Evans rats subjected to 6 weeks of Western diet feeding and wheel-running training. A control group of lean rats was fed with a standard diet. In all experimental groups, we measured physiological parameters (animal weights, body composition, serum metabolic parameters). We assessed the impact of simultaneous exposure to a Western diet and wheel-running on the cerebrocortical protein expression (global proteomic profiling), and in the second part of the experiment, we measured the cortical levels of protein related to brain metabolism (Western blot). RESULTS Western diet led to an obese phenotype and induced changes in the serum metabolic parameters. Wheel-running did not reduce animal weights or fat mass but significantly decreased serum glucose level. The global proteome analysis revealed that the altered proteins were functionally annotated as they were involved mostly in metabolic pathways. Western blot analysis showed the downregulation of the mitochondrial protein-Acyl-CoA dehydrogenase family member 9, hexokinase 1 (HK1)-enzyme involved in principal glucose metabolism pathways and monocarboxylate transporter 2 (MCT2). Wheel-running reversed this decline in the cortical levels of HK1 and MCT2. CONCLUSION The cerebrocortical proteome is affected by a combination of physical activity and Western diet in female rats. An analysis of the cortical proteins involved in brain energy metabolism provides a valuable basis for the deeper investigation of changes in the brain structure and function induced by simultaneous exposure to a Western diet and physical activity.
Collapse
Affiliation(s)
- Marta Maria Nowacka-Chmielewska
- Laboratory of Molecular Biology, Institute of Physiotherapy and Health Sciences, Academy of Physical Education, 40-065 Katowice, Poland; (D.L.); (N.P.); (A.M.)
- Correspondence: ; Tel.: +48-509-505-836
| | - Daniela Liśkiewicz
- Laboratory of Molecular Biology, Institute of Physiotherapy and Health Sciences, Academy of Physical Education, 40-065 Katowice, Poland; (D.L.); (N.P.); (A.M.)
| | - Konstancja Grabowska
- Department for Experimental Medicine, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland; (K.G.); (M.G.); (J.J.B.)
| | - Arkadiusz Liśkiewicz
- Department of Physiology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland;
| | - Łukasz Marczak
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, 61-704 Poznań, Poland; (Ł.M.); (A.W.)
| | - Anna Wojakowska
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, 61-704 Poznań, Poland; (Ł.M.); (A.W.)
| | - Natalia Pondel
- Laboratory of Molecular Biology, Institute of Physiotherapy and Health Sciences, Academy of Physical Education, 40-065 Katowice, Poland; (D.L.); (N.P.); (A.M.)
| | - Mateusz Grabowski
- Department for Experimental Medicine, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland; (K.G.); (M.G.); (J.J.B.)
| | - Jarosław Jerzy Barski
- Department for Experimental Medicine, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland; (K.G.); (M.G.); (J.J.B.)
- Department of Physiology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland;
| | - Andrzej Małecki
- Laboratory of Molecular Biology, Institute of Physiotherapy and Health Sciences, Academy of Physical Education, 40-065 Katowice, Poland; (D.L.); (N.P.); (A.M.)
| |
Collapse
|
38
|
Sung HH, Gi MY, Cha JA, Cho HE, Moon AE, Yoon H. Gender difference in the relationship between lipid accumulation product index and pulse pressure in nondiabetic Korean adults: The Korean National Health and Nutrition Examination Survey 2013-2014. Clin Exp Hypertens 2021; 44:146-153. [PMID: 34821192 DOI: 10.1080/10641963.2021.2007943] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The present study was conducted to assess the association between the lipid accumulation product index (LAP) and pulse pressure (PP) by gender in nondiabetic Korean adults. This study used the data of 8,240 nondiabetic adults (3,577 men and 4,663 women) aged ≥ 20 years from the Korean National Health and Nutrition Examination Survey 2013-2014. Key findings from the study were as follows: first, the mean values of age for the overall population, men, and women were 49.59 ± 15.73 years, 49.26 ± 16.04 years, and 49.85 ± 15.47 years, respectively. Second, in women (n = 4,663), after adjustment for related variables and with quartile 1 of LAP as a reference, the odds ratios (ORs) of high PP (PP > 60 mmHg) were significantly higher in quartile 3 [1.735 (95% confidence interval [CI], 1.064-2.831)] and quartile 4 of LAP [2.271 (95% CI, 1.325-3.893)]. Third, high PP in men (n = 3,577) was not associated with the quartiles of LAP. Forth, after adjustment for related variables, the PP level was positively associated with the quartiles of LAP in women (p < .001) but not in men (p = .400). PP was positively associated with LAP in nondiabetic Korean women but not in men.
Collapse
Affiliation(s)
- Hyun Ho Sung
- Department of Clinical Laboratory Science, Dongnam Health University, Suwonsi, South Korea
| | - Mi Young Gi
- Department of Nursing, Christian College of Nursing, Gwangju, South Korea
| | - Ju Ae Cha
- Department of Nursing, Chunnam Technouniversity, Gokseong-gun, South Korea
| | - Hye Eun Cho
- Department of Dental Hygiene, Kwangju Womens's University, Gwangsan-gu, South Korea
| | - Ae Eun Moon
- Department of Dental Hygiene, Honam University, Gwangju, South Korea
| | - Hyun Yoon
- Department of Clinical Laboratory Science, Wonkwang Health Science University, Iksan-si, South Korea
| |
Collapse
|
39
|
Terburgh K, Lindeque JZ, van der Westhuizen FH, Louw R. Cross-comparison of systemic and tissue-specific metabolomes in a mouse model of Leigh syndrome. Metabolomics 2021; 17:101. [PMID: 34792662 DOI: 10.1007/s11306-021-01854-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 11/03/2021] [Indexed: 01/03/2023]
Abstract
INTRODUCTION The value of metabolomics in multi-systemic mitochondrial disease research has been increasingly recognized, with the ability to investigate a variety of biofluids and tissues considered a particular advantage. Although minimally invasive biofluids are the generally favored sample type, it remains unknown whether systemic metabolomes provide a clear reflection of tissue-specific metabolic alterations. OBJECTIVES Here we cross-compare urine and tissue-specific metabolomes in the Ndufs4 knockout mouse model of Leigh syndrome-a complex neurometabolic MD defined by progressive focal lesions in specific brain regions-to identify and evaluate the extent of common and unique metabolic alterations on a systemic and brain regional level. METHODS Untargeted and semi-targeted multi-platform metabolomics were performed on urine, four brain regions, and two muscle types of Ndufs4 KO (n≥19) vs wildtype (n≥20) mice. RESULTS Widespread alterations were evident in alanine, aspartate, glutamate, and arginine metabolism in Ndufs4 KO mice; while brain-region specific metabolic signatures include the accumulation of branched-chain amino acids, proline, and glycolytic intermediates. Furthermore, we describe a systemic dysregulation in one-carbon metabolism and the tricarboxylic acid cycle, which was not clearly reflected in the Ndufs4 KO brain. CONCLUSION Our results confirm the value of urinary metabolomics when evaluating MD-associated metabolites, while cautioning against mechanistic studies relying solely on systemic biofluids.
Collapse
Affiliation(s)
- Karin Terburgh
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University (Potchefstroom Campus), Private Bag X6001, Potchefstroom, South Africa
| | - Jeremie Z Lindeque
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University (Potchefstroom Campus), Private Bag X6001, Potchefstroom, South Africa
| | - Francois H van der Westhuizen
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University (Potchefstroom Campus), Private Bag X6001, Potchefstroom, South Africa
| | - Roan Louw
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University (Potchefstroom Campus), Private Bag X6001, Potchefstroom, South Africa.
| |
Collapse
|
40
|
Cohen CC, Perng W, Sundaram SS, Scherzinger A, Shankar K, Dabelea D. Hepatic Fat in Early Childhood Is Independently Associated With Estimated Insulin Resistance: The Healthy Start Study. J Clin Endocrinol Metab 2021; 106:3140-3150. [PMID: 34289064 PMCID: PMC8530740 DOI: 10.1210/clinem/dgab541] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND Fatty liver disease is a common metabolic abnormality in adolescents with obesity but remains understudied in early childhood. OBJECTIVES To describe hepatic fat deposition in prepubertal children and examine cross-sectional associations with metabolic markers and body composition. METHODS Data were from 286 children ages 4 to 8 years old in the Healthy Start Study, a longitudinal prebirth cohort in Colorado (USA). Assessments included magnetic resonance imaging to quantify hepatic and abdominal fats, fasting blood draws to measure metabolic markers, and air displacement plethysmography to measure body composition (fat mass and fat-free mass). RESULTS The median (interquartile range) for hepatic fat was 1.65% (1.24%, 2.11%). Log-transformed hepatic fat was higher in Hispanic [mean (95% CI): 0.63 (0.52, 0.74)] vs non-Hispanic white children [0.46 (0.38, 0.53), P = 0.01] and children with overweight/obesity [0.64 (0.49, 0.79)] vs normal-weight [0.47 (0.40, 0.53), P = 0.02]. Higher log-hepatic fat was associated with higher insulin [β (95% CI): 1.47 (0.61, 2.33) uIU/mL, P = 0.001] and estimated insulin resistance (homeostatic model assessment) [0.40 (0.20, 0.60), P < 0.001] in the full sample and glucose [5.53 (2.84, 8.21) mg/dL, P < 0.001] and triglycerides [10.92 (2.92,18.91) mg/dL, P = 0.008] in boys, in linear regression models adjusted for sociodemographics, maternal/perinatal confounders, and percentage body fat. Log-hepatic fat was also associated with abdominal subcutaneous adipose tissue [SAT; 7.37 (1.12,13.60) mm2, P = 0.02] in unadjusted models, but this was attenuated and insignificant after adjusting for confounders. CONCLUSIONS While hepatic fat was low in children 4 to 8 years old, it was independently associated with estimated insulin resistance and exhibited sex-specific associations with glucose and triglycerides, suggesting hepatic fat may be an early indicator of metabolic dysfunction in youth.
Collapse
Affiliation(s)
- Catherine C Cohen
- Department of Pediatrics, University of Colorado School of Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, Colorado School of Public Health, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Wei Perng
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, Colorado School of Public Health, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
- Department of Nutritional Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Shikha S Sundaram
- Department of Pediatrics, University of Colorado School of Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Ann Scherzinger
- Department of Radiology, University of Colorado, School of Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Kartik Shankar
- Department of Pediatrics, University of Colorado School of Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, Colorado School of Public Health, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Dana Dabelea
- Department of Pediatrics, University of Colorado School of Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, Colorado School of Public Health, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
41
|
Untargeted metabolomics reveals sex-specific differences in lipid metabolism of adult rats exposed to dexamethasone in utero. Sci Rep 2021; 11:20342. [PMID: 34645877 PMCID: PMC8514544 DOI: 10.1038/s41598-021-99598-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 09/22/2021] [Indexed: 12/13/2022] Open
Abstract
Prenatal stress through glucocorticoid (GC) exposure leads to an increased risk of developing diseases such as cardiovascular disease, metabolic syndrome and hypertension in adulthood. We have previously shown that administration of the synthetic glucocorticoid, dexamethasone (Dex), to pregnant Wistar-Kyoto dams produces offspring with elevated blood pressures and disrupted circadian rhythm signaling. Given the link between stress, circadian rhythms and metabolism, we performed an untargeted metabolomic screen on the livers of offspring to assess potential changes induced by prenatal Dex exposure. This metabolomic analysis highlighted 18 significantly dysregulated metabolites in females and 12 in males. Pathway analysis using MetaboAnalyst 4.0 highlighted key pathway-level metabolic differences: glycerophospholipid metabolism, purine metabolism and glutathione metabolism. Gene expression analysis revealed significant upregulation of several lipid metabolism genes in females while males showed no dysregulation. Triglyceride concentrations were also found to be significantly elevated in female offspring exposed to Dex in utero, which may contribute to lipid metabolism activation. This study is the first to conduct an untargeted metabolic profile of liver from GC exposed offspring. Corroborating metabolic, gene expression and lipid profiling results demonstrates significant sex-specific lipid metabolic differences underlying the programming of hepatic metabolism.
Collapse
|
42
|
Yaribeygi H, Maleki M, Sathyapalan T, Jamialahmadi T, Sahebkar A. Pathophysiology of Physical Inactivity-Dependent Insulin Resistance: A Theoretical Mechanistic Review Emphasizing Clinical Evidence. J Diabetes Res 2021; 2021:7796727. [PMID: 34660812 PMCID: PMC8516544 DOI: 10.1155/2021/7796727] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 09/24/2021] [Accepted: 09/27/2021] [Indexed: 12/17/2022] Open
Abstract
The modern lifestyle has a negative impact on health. It is usually accompanied by increased stress levels and lower physical activity, which interferes with body homeostasis. Diabetes mellitus is a relatively common metabolic disorder with increasing prevalence globally, associated with various risk factors, including lower physical activity and a sedentary lifestyle. It has been shown that sedentary behavior increases the risk of insulin resistance, but the intermediate molecular mechanisms are not fully understood. In this mechanistic review, we explore the possible interactions between physical inactivity and insulin resistance to help better understand the pathophysiology of physical inactivity-dependent insulin resistance and finding novel interventions against these deleterious pathways.
Collapse
Affiliation(s)
- Habib Yaribeygi
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran
| | - Mina Maleki
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Thozhukat Sathyapalan
- Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School, University of Hull, UK
| | - Tannaz Jamialahmadi
- Department of Food Science and Technology, Quchan Branch, Islamic Azad University, Quchan, Iran
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
43
|
Shin D, Lee KW. Dietary carbohydrates interact with AMY1 polymorphisms to influence the incidence of type 2 diabetes in Korean adults. Sci Rep 2021; 11:16788. [PMID: 34408213 PMCID: PMC8373941 DOI: 10.1038/s41598-021-96257-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 07/30/2021] [Indexed: 01/23/2023] Open
Abstract
The relationship between AMY1 single nucleotide polymorphisms (SNPs), dietary carbohydrates, and the risk of type 2 diabetes is unclear. We aimed to evaluate this association using an ongoing large-scale prospective study, namely the Korean Genome and Epidemiology Study. We selected six genetic variants of the AMY1 gene: rs10881197, rs4244372, rs6696797, rs1566154, rs1930212, and rs1999478. Baseline dietary data were obtained using a semi-quantitative food frequency questionnaire. Type 2 diabetes was defined according to the criteria of the World Health Organization and American Diabetes Association. During an average follow-up period of 12 years (651,780 person-years), 1082 out of 4552 (23.8%) patients had type 2 diabetes. Three AMY1 SNPs were significantly associated with diabetes incidence among patients with carbohydrate intake > 65% of total energy: rs6696797, rs4244372, and rs10881197. In multivariable Cox models, Korean women with the rs6696797 AG or AA genotype had 28% higher incidence of type 2 diabetes (hazard ratio 1.28, 95% confidence interval 1.06-1.55) than Korean women with the rs6696797 GG genotype. We did not observe significant associations between AMY1 SNPs, dietary carbohydrates, and diabetes incidence in Korean men. We conclude that AMY1 genetic variants and dietary carbohydrate intake influence the incidence of type 2 diabetes in Korean women only. Korean women who are minor carriers of the AMY1 rs6696797, rs4244372, and rs10881197 genotypes may benefit from a low-carbohydrate diet to prevent the future risk of type 2 diabetes.
Collapse
Affiliation(s)
- Dayeon Shin
- Department of Food and Nutrition, Inha University, Incheon, 22212, Republic of Korea
| | - Kyung Won Lee
- Department of Home Economics Education, Korea National University of Education, Cheongju, 28173, Republic of Korea.
| |
Collapse
|
44
|
Ahmadizar F, Wang K, Aribas E, Fani L, Heshmatollah A, Ikram MK, Kavousi M. Impaired fasting glucose, type 2 diabetes mellitus, and lifetime risk of cardiovascular disease among women and men: the Rotterdam Study. BMJ Open Diabetes Res Care 2021; 9:9/1/e002406. [PMID: 34433547 PMCID: PMC8388277 DOI: 10.1136/bmjdrc-2021-002406] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 08/15/2021] [Indexed: 12/20/2022] Open
Abstract
INTRODUCTION Data on sex-specific lifetime risk of cardiovascular disease (CVD) across the glycemic spectrum, in particular in impaired fasting glucose (IFG) state, are scarce. Whether overweight/obesity modifies the CVD burden also remains unclear. RESEARCH DESIGN AND METHODS Using a prospective population-based Rotterdam Study, normoglycemia, IFG, and type 2 diabetes mellitus (T2D) were defined. First incident cases of coronary heart disease, heart failure, and stroke during a follow-up time until January 1, 2015 were identified and formed the composite CVD end point. The remaining lifetime risks of CVD were estimated in each glucose category at 55, 65, 75, and 85 years of age, using a modified version of survival analysis adjusted for the competing risk of death. RESULTS Among 5698 women and 3803 men free of CVD at baseline, the mean age was 64.5 years (SD 9.6) and 60.0% of participants were women. At age 55 years, the remaining lifetime risk of any CVD event among women was 55.1% (95% CI 48.3 to 61.9) for IFG, compared with 52.7% (95% CI 49.5 to 55.9) for normoglycemia and 61.5% (95% CI 54.7 to 68.3) for T2D. For men, the remaining lifetime risk of any CVD event was 62.1% (95% CI 55.2 to 69.1) for IFG, compared with 59.1% (95% CI 55.5 to 62.7) for normoglycemia and 60.3% (95% CI 53.1 to 67.5) for T2D. At age 55 years, the lifetime risk for incident CVD was higher, although not statistically significant, among women and men with IFG who were overweight or had obesity compared with normal-weight women and men. CONCLUSION IFG carried a large lifetime risk for incident CVD among both women and men compared with normoglycemia. In particular among men, the risk was comparable to that of T2D. Overweight/Obesity modifies the risk and conferred a larger burden of lifetime CVD risk among women and men with IFG.
Collapse
Affiliation(s)
- Fariba Ahmadizar
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Kan Wang
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Elif Aribas
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Lana Fani
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Alis Heshmatollah
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - M Kamran Ikram
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Department of Neurology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Maryam Kavousi
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
45
|
Tamura Y, Ohhata T, Niida H, Sakai S, Uchida C, Masumoto K, Katou F, Wutz A, Kitagawa M. Homologous recombination is reduced in female embryonic stem cells by two active X chromosomes. EMBO Rep 2021; 22:e52190. [PMID: 34309165 DOI: 10.15252/embr.202052190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 06/21/2021] [Accepted: 06/22/2021] [Indexed: 12/16/2022] Open
Abstract
The reactivation of X-linked genes is observed in some primary breast tumors. Two active X chromosomes are also observed in female embryonic stem cells (ESCs), but whether double doses of X-linked genes affect DNA repair efficiency remains unclear. Here, we establish isogenic female/male ESCs and show that the female ESCs are more sensitive to camptothecin and have lower gene targeting efficiency than male ESCs, suggesting that homologous recombination (HR) efficiency is reduced in female ESCs. We also generate Xist-inducible female ESCs and show that the lower HR efficiency is restored when X chromosome inactivation is induced. Finally, we assess the X-linked genes with a role in DNA repair and find that Brcc3 is one of the genes involved in a network promoting proper HR. Our findings link the double doses of X-linked genes with lower DNA repair activity, and this may have relevance for common diseases in female patients, such as breast cancer.
Collapse
Affiliation(s)
- Yuka Tamura
- Department of Molecular Biology, Hamamatsu University School of Medicine, Hamamatsu, Japan.,Department of Oral and Maxillofacial Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Tatsuya Ohhata
- Department of Molecular Biology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hiroyuki Niida
- Department of Molecular Biology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Satoshi Sakai
- Department of Molecular Biology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Chiharu Uchida
- Advanced Research Facilities & Services, Preeminent Medical Photonics Education & Research Center, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kazuma Masumoto
- Department of Oral and Maxillofacial Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Fuminori Katou
- Department of Oral and Maxillofacial Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Anton Wutz
- Institute of Molecular Health Sciences, ETH Zürich, Zurich, Switzerland
| | - Masatoshi Kitagawa
- Department of Molecular Biology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| |
Collapse
|
46
|
Karras SN, Koufakis T, Dimakopoulos G, Adamidou L, Karalazou P, Thisiadou K, Bais A, Tzotzas T, Manthou E, Makedou K, Kotsa K. Vitamin D equilibrium affects sex-specific changes in lipid concentrations during Christian Orthodox fasting. J Steroid Biochem Mol Biol 2021; 211:105903. [PMID: 33933575 DOI: 10.1016/j.jsbmb.2021.105903] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/07/2021] [Accepted: 04/26/2021] [Indexed: 01/06/2023]
Abstract
We aimed to evaluate sex differences in changes of lipid profiles in a cohort of metabolically healthy adults following Orthodox fasting (OF), as well as to assess a potential role of vitamin D status in mediating these variations. 45 individuals (24 premenopausal females, 53.3 %) with mean age 48.3 ± 9.1 years and mean Body Mass Index 28.7 ± 5.8 kg/m2 were prospectively followed for 12 weeks. Anthropometry, dietary and biochemical data regarding serum lipids, and vitamin D status were collected at baseline, 7 weeks after the implementation of OF, and 5 weeks after fasters returned to their standard dietary habits (12 weeks from baseline). According to 25-hydroxy-vitamin D [25(OH)D] measurements, participants were divided into two groups: those with concentrations above and below the median of values. Females with 25(OH)D concentrations below the median manifested a non-significant reduction by approximately 15 % in total and low-density lipoprotein cholesterol during the fasting period, followed by a significant increase 5 weeks after OF cessation (170.7 vs. 197.5 and 99.6 vs. 121.0 mg/dl respectively, p < 0.001). In contrast, males with 25(OH)D levels below the median demonstrated an inverse, non-significant trend of increase in lipid concentrations during the whole study period. Our findings suggest strikingly different inter-gender lipid responses to a dietary model of low-fat, mediated by vitamin D status. Further studies are necessary to reveal the underlying mechanisms and assess the importance of these differences with respect to cardiovascular health and the benefit of vitamin D supplementation strategies.
Collapse
Affiliation(s)
- Spyridon N Karras
- Division of Endocrinology and Metabolism, First Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, Greece
| | - Theocharis Koufakis
- Division of Endocrinology and Metabolism, First Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, Greece
| | - Georgios Dimakopoulos
- Medical Statistics, Epirus Science and Technology Park Campus of the University of Ioannina, Ioannina, Greece
| | - Lilian Adamidou
- Department of Dietetics and Nutrition, AHEPA University Hospital, Thessaloniki, Greece
| | - Paraskevi Karalazou
- Laboratory of Biochemistry, AHEPA General Hospital, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Katerina Thisiadou
- Laboratory of Biochemistry, AHEPA General Hospital, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Alkiviadis Bais
- Aristotle University of Thessaloniki, Laboratory of Atmospheric Physics, Thessaloniki, Greece
| | | | - Eleni Manthou
- Division of Endocrinology and Metabolism, First Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, Greece
| | - Kali Makedou
- Laboratory of Biochemistry, AHEPA General Hospital, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Kalliopi Kotsa
- Division of Endocrinology and Metabolism, First Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, Greece.
| |
Collapse
|
47
|
Artegoitia VM, Krishnan S, Bonnel EL, Stephensen CB, Keim NL, Newman JW. Healthy eating index patterns in adults by sex and age predict cardiometabolic risk factors in a cross-sectional study. BMC Nutr 2021; 7:30. [PMID: 34154665 PMCID: PMC8218401 DOI: 10.1186/s40795-021-00432-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 04/14/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Associations between diet and cardiometabolic disease (CMD) risk may vary in men and women owing to sex differences in eating habits and physiology. The current secondary analysis sought to determine the ability of sex differences in dietary patterns to discriminate groups with or without CMD risk factors (CMDrf) in the adult population and if this was influenced by age. METHODS Diet patterns and quality were evaluated using 24 h recall-based Healthy Eating Index (HEI-2015) in free-living apparently healthy men (n = 184) and women (n = 209) 18-65 y of age with BMIs of 18-44 kg/m2. Participants were stratified into low- and high-CMDrf groups based on the presence/absence of at least one CMDrf: BMI > 25 kg/m2; fasting triglycerides > 150 mg/dL; HDL cholesterol < 50 mg/dL-women or < 40 mg/dL-men; HOMA > 2; HbA1c > 5.7. Sex by age dietary patterns were stratified by multivariate analyses, with metabolic variable associations established by stepwise discriminant analysis. RESULTS Diet quality increased with age in both sexes (P < 0.01), while women showed higher fruit, vegetable and saturated fat intake as a percentage of total energy (P < 0.05). The total-HEI score (i.e. diet quality) was lower in the high-CMDrf group (P = 0.01), however, diet quality parameters predicted CMDrf presence more accurately when separated by sex. Lower 'total vegetable' intake in the high-CMDrf group in both sexes, while high-CMDrf men also had lower 'total vegetables', 'greens and beans' intake, and high-CMDrf women had lower 'total fruits', 'whole-fruits', 'total vegetables', 'seafood and plant-proteins', 'fatty acids', and 'saturated fats' intakes (P < 0.05). Moreover, 'dairy' intake was higher in high-CMDrf women but not in men (sex by 'dairy' interaction P = 0.01). Sex by age diet pattern models predicted CMDrf with a 93 and 89% sensitivity and 84 and 92% specificity in women and men, respectively. CONCLUSIONS Sex and age differences in dietary patterns classified participants with and without accepted CMDrfs, supporting an association between specific diet components and CMD risk that differs by sex. Including sex specific dietary patterns into health assessments may provide targeted nutritional guidance to reduce the burden of cardiovascular disease. TRIAL REGISTRATION ClinicalTrials.gov : NCT02367287 . ClinicalTrials.gov : NCT02298725 .
Collapse
Affiliation(s)
- Virginia M Artegoitia
- Obesity and Metabolism Research Unit, United States Department of Agriculture-Agricultural Research Services-Western Human Nutrition Research Center, 430 West Health Sciences Drive, Davis, CA, 95616, USA
| | - Sridevi Krishnan
- Department of Nutrition, University of California Davis, Davis, CA, USA
| | - Ellen L Bonnel
- Department of Nutrition, University of California Davis, Davis, CA, USA
- Human Studies Unit, United States Department of Agriculture-Agricultural Research Services-Western Human Nutrition Research Center, Davis, CA, USA
| | - Charles B Stephensen
- Department of Nutrition, University of California Davis, Davis, CA, USA
- Immunity and Disease Prevention Research Unit, United States Department of Agriculture-Agricultural Research Services-Western Human Nutrition Research Center, Davis, CA, USA
| | - Nancy L Keim
- Obesity and Metabolism Research Unit, United States Department of Agriculture-Agricultural Research Services-Western Human Nutrition Research Center, 430 West Health Sciences Drive, Davis, CA, 95616, USA
- Department of Nutrition, University of California Davis, Davis, CA, USA
| | - John W Newman
- Obesity and Metabolism Research Unit, United States Department of Agriculture-Agricultural Research Services-Western Human Nutrition Research Center, 430 West Health Sciences Drive, Davis, CA, 95616, USA.
- Department of Nutrition, University of California Davis, Davis, CA, USA.
- West Coast Metabolomics Center, Genome Center, University of California Davis, Davis, CA, USA.
| |
Collapse
|
48
|
Mulholland M, Jakobsson G, Lei Y, Sundius L, Ljungcrantz I, Rattik S, Tietge UJF, Engelbertsen D. IL-2Rβγ signalling in lymphocytes promotes systemic inflammation and reduces plasma cholesterol in atherosclerotic mice. Atherosclerosis 2021; 326:1-10. [PMID: 33945906 DOI: 10.1016/j.atherosclerosis.2021.04.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 04/08/2021] [Accepted: 04/21/2021] [Indexed: 11/20/2022]
Abstract
BACKGROUND AND AIMS The relationship between inflammation and lipid metabolism is complex and bidirectional. Lymphocyte-driven inflammation has been shown to modulate both atherosclerotic plaque development and cholesterol levels, but the mechanisms are incompletely understood. METHODS The cardiometabolic effects of IL-2Rβγ signalling in atherosclerotic Apoe-/- mice were investigated by treatment with an agonistic IL-2Rβγ-targeting IL-2/anti-IL-2 complex or a monoclonal anti-CD122 (IL-2Rβ) blocking antibody. RESULTS Administration of IL-2Rβγ agonistic IL-2/anti-IL-2 complexes to Apoe-/- mice augmented opposing arms of the adaptive immune system. Expansion of effector/memory T cells and increased levels of circulating pro-inflammatory cytokines were observed along with elevated levels of regulatory T cells and IL-10. Notably, IL-2/anti-IL-2 treatment did not affect plaque size but decreased levels of plasma cholesterol. The cholesterol lowering effect of IL-2Rβγ agonism was not affected by anti-CD8 or anti-NK1.1 depleting antibody treatment but was contingent on the presence of adaptive immunity. Expression of multiple liver X receptor (LXR)-related genes, including Pltp and Srebp1c in the liver, was decreased by IL-2/anti-IL-2 treatment. Although IL-2Rβγ agonism lowered cholesterol levels, blocking IL-2Rβγ signalling using an anti-CD122 monoclonal antibody did not impact cholesterol levels or plaque burden in Apoe-/- mice. CONCLUSIONS Elevated IL-2Rβγ signalling results in activation of both inflammatory and regulatory lymphocytes with a net zero effect on atherosclerosis and decreased plasma cholesterol levels. Changes in cholesterol levels were associated with reductions in hepatic LXR-related gene expression. Further studies are needed to investigate the clinical significance of IL-2 mediated modulation of hepatic LXR signalling in inflammatory disorders.
Collapse
Affiliation(s)
- Megan Mulholland
- Department of Clinical Sciences, Lund University, Skåne University Hospital, 20502, Malmö, Sweden.
| | - Gabriel Jakobsson
- Department of Clinical Sciences, Lund University, Skåne University Hospital, 20502, Malmö, Sweden
| | - Yu Lei
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, S-14183, Stockholm, Sweden
| | - Lena Sundius
- Department of Clinical Sciences, Lund University, Skåne University Hospital, 20502, Malmö, Sweden
| | - Irena Ljungcrantz
- Department of Clinical Sciences, Lund University, Skåne University Hospital, 20502, Malmö, Sweden
| | - Sara Rattik
- Department of Clinical Sciences, Lund University, Skåne University Hospital, 20502, Malmö, Sweden
| | - Uwe J F Tietge
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, S-14183, Stockholm, Sweden; Clinical Chemistry, Karolinska University Laboratory, Karolinska University Hospital, SE-141 86, Stockholm, Sweden
| | - Daniel Engelbertsen
- Department of Clinical Sciences, Lund University, Skåne University Hospital, 20502, Malmö, Sweden.
| |
Collapse
|
49
|
Haider A, Bengs S, Luu J, Osto E, Siller-Matula JM, Muka T, Gebhard C. Sex and gender in cardiovascular medicine: presentation and outcomes of acute coronary syndrome. Eur Heart J 2021; 41:1328-1336. [PMID: 31876924 DOI: 10.1093/eurheartj/ehz898] [Citation(s) in RCA: 179] [Impact Index Per Article: 44.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 09/01/2019] [Accepted: 12/02/2019] [Indexed: 12/14/2022] Open
Abstract
Although health disparities in women presenting with acute coronary syndrome (ACS) have received growing attention in recent years, clinical outcomes from ACS are still worse for women than for men. Women continue to experience higher patient and system delays and receive less aggressive invasive treatment and pharmacotherapies. Gender- and sex-specific variables that contribute to ACS vulnerability remain largely unknown. Notwithstanding the sex differences in baseline coronary anatomy and function, women and men are treated the same based on guidelines that were established from experimental and clinical trial data over-representing the male population. Importantly, younger women have a particularly unfavourable prognosis and a plethora of unanswered questions remains in this younger population. The present review summarizes contemporary evidence for gender and sex differences in vascular biology, clinical presentation, and outcomes of ACS. We further discuss potential mechanisms and non-traditional risk conditions modulating the course of disease in women and men, such as unrecognized psychosocial factors, sex-specific vascular and neural stress responses, and the potential impact of epigenetic modifications.
Collapse
Affiliation(s)
- Ahmed Haider
- Department of Nuclear Medicine, University Hospital Zurich, Raemistrasse 100, 8091 Zurich, Switzerland.,Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
| | - Susan Bengs
- Department of Nuclear Medicine, University Hospital Zurich, Raemistrasse 100, 8091 Zurich, Switzerland.,Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
| | - Judy Luu
- Division of Cardiology, Department of Internal Medicine, University of Manitoba, 820 Sherbrook Street, Winnipeg MB R3A, Manitoba, Canada
| | - Elena Osto
- Institute of Clinical Chemistry, University of Zurich and University Hospital Zurich, Raemistrasse 100, 8091 Zurich, Switzerland.,Cardiology, University Heart Center, University Hospital Zurich, Raemistrasse 100, 8091 Zurich, Switzerland
| | - Jolanta M Siller-Matula
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria.,Centre for Preclinical Research and Technology, Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Zwirki i Wigury 61, 02-091 Warsaw, Poland
| | - Taulant Muka
- Institute of Social and Preventive Medicine, University of Bern, Mittelstrasse 43, 3012 Bern, Switzerland
| | - Catherine Gebhard
- Department of Nuclear Medicine, University Hospital Zurich, Raemistrasse 100, 8091 Zurich, Switzerland.,Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland.,Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| |
Collapse
|
50
|
Yang X, Liu S, Zhang Z. Sex difference in blood pressure, a combinatorial consequence of the differential in RAAS components, sex hormones and time course. Curr Hypertens Rev 2021; 18:11-16. [PMID: 33992057 DOI: 10.2174/1573402117666210511011444] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/26/2021] [Accepted: 03/02/2021] [Indexed: 11/22/2022]
Abstract
The longitudinal increment of blood pressure (BP) with age is attributed to lifestyle, internal and external environments. It is not limited to systemic brain-derived neurotrophic factor (BDNF), signaling to allow the individuals to better adapt to the developmental and environmental change. This regulation is necessary for all lives, regardless of sex. Basic levels of renin-angiotensin- aldosterone system (RAAS) components in males and females define the fundamental sex difference in BP, which may be set by prenatal programming and profoundly influence BP after birth. The innate sex difference in BP is magnified during puberty growth and further modified by menopause. At the age of 70 or older, blood pressure was similar in men and women. The understanding of the prenatal setup and development of sexual dimorphism in BP may provide preventative therapeutic strategies, including timing and drugs, for individuals with abnormal BP.
Collapse
Affiliation(s)
- Xiaomei Yang
- College of Basic Medicine, Henan University of Chinese Medicine,156,Jinshui East Road ,Zhengdong New District, Zhengzhou, Henan 450046. China
| | - Shien Liu
- College of Basic Medicine, Henan University of Chinese Medicine,156,Jinshui East Road ,Zhengdong New District, Zhengzhou, Henan 450046. China
| | - Zhongming Zhang
- College of Chinese Medicine, and Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Nanyang Institute of Technology; 80 Changjiang Rd. Nanyang, Henan, 473004. China
| |
Collapse
|