1
|
Wu J, Wu P, Wang S, Guan Y, Wang J. Revealing the Landscape Crosstalk Between Reproductive System and Organs Aging. FASEB J 2025; 39:e70572. [PMID: 40289595 DOI: 10.1096/fj.202403410r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 03/30/2025] [Accepted: 04/16/2025] [Indexed: 04/30/2025]
Abstract
The reproductive system is a vital component of the human body. In modern society, due to various socio-economic reasons, an increasing number of couples are choosing to postpone childbearing. Research into the impact of aging on the reproductive system is becoming increasingly important. As people age, there is a decline in the reproductive system across various levels, from the testes in males to spermatogonia cells, and from the ovaries in females to oocytes. The aging of the reproductive system not only affects the system itself but also has implications for other organs and systems in the body. Conversely, the aging of other organs and systems can also damage the reproductive system. This review organizes the changes that occur within the reproductive system as a result of aging and focuses on the interactions between the reproductive system and other systems. Additionally, this review summarizes current therapies aimed at delaying aging, which may provide insights for future interventions targeting the aging of the reproductive system and other systems.
Collapse
Affiliation(s)
- Jiahong Wu
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, P.R. China
| | - Peng Wu
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, P.R. China
| | - Sicheng Wang
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, P.R. China
| | - Yupeng Guan
- School of Medicine, Sun Yat-sen University, Shenzhen, P.R. China
| | - Jiancheng Wang
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, P.R. China
- School of Medicine, Sun Yat-sen University, Shenzhen, P.R. China
| |
Collapse
|
2
|
Ouyang Q, Dong Y, Li R, Hu Y, Xue Q, Yu X, Li J, Zhang P, Wu N, Yang Y, Li F, Wang T, Li Y, Li S, Pan XF. Associations of Hysterectomy, Oophorectomy, and Hormone Replacement Therapy With the Risk of Type 2 Diabetes Mellitus in Postmenopausal Women. Clin Endocrinol (Oxf) 2025. [PMID: 40255188 DOI: 10.1111/cen.15253] [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: 01/20/2025] [Revised: 04/05/2025] [Accepted: 04/07/2025] [Indexed: 04/22/2025]
Abstract
OBJECTIVE Female-specific risk factors warrant attention in the prevention and control of type 2 diabetes mellitus (T2DM). The study aimed to investigate the relationships of hysterectomy, bilateral oophorectomy, and hormone replacement therapy (HRT) with the risk of T2DM in postmenopausal women. DESIGN We included 127,514 postmenopausal women without T2DM at baseline from the UK Biobank. MEASUREMENTS Hysterectomy, bilateral oophorectomy, and HRT were self-reported at baseline, and incident T2DM was identified using ICD-10 code E11 during the follow-up period. RESULTS Compared to no hysterectomy/bilateral oophorectomy, hysterectomy alone (HR, 1.20; 95%CI: 1.09, 1.32) and combined hysterectomy and bilateral oophorectomy (HR, 1.19; 95%CI: 1.08, 1.32) were associated with higher risks of incident T2DM. Independent of other factors, the history of HRT was associated with a higher risk of T2DM (HR, 1.08; 95%CI: 1.03, 1.14), but this positive association was observed only in women without no hysterectomy or bilateral oophorectomy. Within the women without surgical procedures, the association between HRT and T2DM existed only in those younger than 45 years (HR, 1.27; 95%CI: 1.14, 1.41), but not in the older (HR, 1.03; 95%CI: 0.96, 1.09). CONCLUSIONS Hysterectomy, regardless of bilateral oophorectomy status, was associated with a higher risk of T2DM. The HRT use, particularly early use in women without surgical interventions, was associated with a high risk. Our findings indicate that female-specific risk factors such as hysterectomy and bilateral oophorectomy and HRT use should be incorporated into the assessments for potential risk of T2DM in postmenopausal women.
Collapse
Affiliation(s)
- Qingqing Ouyang
- Department of Epidemiology and Biostatistics, School of Public Health, Chengdu Medical College, Chengdu, Sichuan, China
- Section of Epidemiology and Population Health & Department of Gynecology and Obstetrics, Ministry of Education Key Laboratory of Birth Defects and Related Diseases of Women and Children & Children's Medicine Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yidan Dong
- Section of Epidemiology and Population Health & Department of Gynecology and Obstetrics, Ministry of Education Key Laboratory of Birth Defects and Related Diseases of Women and Children & Children's Medicine Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Rui Li
- Section of Epidemiology and Population Health & Department of Gynecology and Obstetrics, Ministry of Education Key Laboratory of Birth Defects and Related Diseases of Women and Children & Children's Medicine Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ying Hu
- Reproductive Endocrinology and Regulation Laboratory & Department of Gynecology and Obstetrics, Ministry of Education Key Laboratory of Birth Defects and Related Diseases of Women and Children & Children's Medicine Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qingping Xue
- Department of Epidemiology and Biostatistics, School of Public Health, Chengdu Medical College, Chengdu, Sichuan, China
| | - Xinyue Yu
- Department of Epidemiology and Biostatistics, School of Public Health, Chengdu Medical College, Chengdu, Sichuan, China
- Section of Epidemiology and Population Health & Department of Gynecology and Obstetrics, Ministry of Education Key Laboratory of Birth Defects and Related Diseases of Women and Children & Children's Medicine Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jingyi Li
- Department of Epidemiology and Biostatistics, School of Public Health, Chengdu Medical College, Chengdu, Sichuan, China
- Section of Epidemiology and Population Health & Department of Gynecology and Obstetrics, Ministry of Education Key Laboratory of Birth Defects and Related Diseases of Women and Children & Children's Medicine Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Peiqi Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Chengdu Medical College, Chengdu, Sichuan, China
- Section of Epidemiology and Population Health & Department of Gynecology and Obstetrics, Ministry of Education Key Laboratory of Birth Defects and Related Diseases of Women and Children & Children's Medicine Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Nianwei Wu
- Section of Epidemiology and Population Health & Department of Gynecology and Obstetrics, Ministry of Education Key Laboratory of Birth Defects and Related Diseases of Women and Children & Children's Medicine Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yunhaonan Yang
- Section of Epidemiology and Population Health & Department of Gynecology and Obstetrics, Ministry of Education Key Laboratory of Birth Defects and Related Diseases of Women and Children & Children's Medicine Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Fan Li
- Section of Epidemiology and Population Health & Department of Gynecology and Obstetrics, Ministry of Education Key Laboratory of Birth Defects and Related Diseases of Women and Children & Children's Medicine Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Tianlei Wang
- Section of Epidemiology and Population Health & Department of Gynecology and Obstetrics, Ministry of Education Key Laboratory of Birth Defects and Related Diseases of Women and Children & Children's Medicine Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yingru Li
- Section of Epidemiology and Population Health & Department of Gynecology and Obstetrics, Ministry of Education Key Laboratory of Birth Defects and Related Diseases of Women and Children & Children's Medicine Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Shuo Li
- Section of Epidemiology and Population Health & Department of Gynecology and Obstetrics, Ministry of Education Key Laboratory of Birth Defects and Related Diseases of Women and Children & Children's Medicine Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiong-Fei Pan
- Section of Epidemiology and Population Health & Department of Gynecology and Obstetrics, Ministry of Education Key Laboratory of Birth Defects and Related Diseases of Women and Children & Children's Medicine Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Shuangliu Institute of Women's and Children's Health, Shuangliu Maternal and Child Health Hospital, Chengdu, Sichuan, China
| |
Collapse
|
3
|
Kuznetsova EA, Fedorov NS, Zakyrjanova GF, Malomouzh AI, Petrov AM. 25-Hydroxycholesterol as a negative regulator of diaphragm muscle contractions via estrogen receptor and Ca 2+ -dependent pathway. Histochem Cell Biol 2025; 163:42. [PMID: 40178695 DOI: 10.1007/s00418-025-02370-9] [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] [Accepted: 03/06/2025] [Indexed: 04/05/2025]
Abstract
Cholesterol is involved in the regulation of various signaling processes, and oxysterols are essential lipid messengers. The cholesterol derivative 25-hydroxycholesterol (25-HC) is overproduced by muscle macrophages in sarcopenia, myasthenia, and neurodegenerative diseases. Herein, we examined the effects of 25-HC on Ca2+ signaling and contractions of the mouse diaphragm, the main respiratory muscle. We found that 25-HC increased resting levels of cytosolic Ca2+ in muscle fibers. This effect was dependent on estrogen receptor α (ERα) and was mediated by Ca2+-efflux from intracellular stores via dantrolene-insensitive and TMB-8-sensitive channels, presumably inositol trisphosphate receptors (IP3Rs). In addition, 25-HC suppressed diaphragm contractile responses to direct stimulation of the muscle fibers. The negative effect of 25-HC on contraction force was inhibited by blockers of ERα and Ca2+ mobilization. Thus, 25-HC may suppress diaphragm muscle contractility due to activation of an ERα/IP3R/Ca2+in axis in muscle fibers. At the same time, 25-HC did not significantly modify the contractions elicited by phrenic nerve stimulation and respiratory activity in vivo. We discuss that the previously found enhancement of neuromuscular transmission mediated by 25-HC in the diaphragm can compensate for the reduction in the muscle contractions in the case of phrenic nerve activation.
Collapse
Affiliation(s)
- Eva A Kuznetsova
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", 2/31 Lobachevsky Street, Box 30, Kazan, 420111, Russia
| | - Nikita S Fedorov
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", 2/31 Lobachevsky Street, Box 30, Kazan, 420111, Russia
| | - Guzel F Zakyrjanova
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", 2/31 Lobachevsky Street, Box 30, Kazan, 420111, Russia
- Department of Human and Animal Physiology, Lomonosov Moscow State University, Leninskiye Gory, 1, 12, Moscow, 119234, Russia
| | - Artem I Malomouzh
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", 2/31 Lobachevsky Street, Box 30, Kazan, 420111, Russia
- Kazan National Research Technical University, 10, K. Marx St, Kazan, 420111, Russia
| | - Alexey M Petrov
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", 2/31 Lobachevsky Street, Box 30, Kazan, 420111, Russia.
- Department of Normal Physiology, Institute of Neuroscience, Kazan State Medical University, 49 Butlerova Street, Kazan, 420012, Russia.
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420008, Kazan, Russia.
| |
Collapse
|
4
|
Lundqvist T, Stenlid R, Halldin M. Hormonal Crossroads in Inborn Errors of the Metabolism Impact of Puberty and Dietary Interventions on Metabolic Health. Metabolites 2025; 15:235. [PMID: 40278364 PMCID: PMC12029320 DOI: 10.3390/metabo15040235] [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: 02/28/2025] [Revised: 03/21/2025] [Accepted: 03/26/2025] [Indexed: 04/26/2025] Open
Abstract
Background/Objectives: Inborn errors of metabolism (IEMs) represent a diverse group of genetic disorders characterized by enzymatic defects that disrupt metabolic pathways, leading to toxic metabolite accumulation, deficits, or impaired macromolecule synthesis. While strict dietary interventions are critical for managing many of these conditions, hormonal and metabolic changes during puberty introduce new challenges. Advancements in early diagnosis and treatment have significantly extended the lifespan of individuals with IEMs. However, this increased longevity is associated with heightened risks of new medical problems, including obesity, insulin resistance, and type 2 diabetes mellitus (T2DM), as these complications share mechanistic features with those seen in obesity and T2DM. Methods: This mini-review examines current knowledge of the intricate interplay between pubertal hormones and metabolic pathways in IEM patients. Results: We address critical questions, such as if puberty intensifies the risk of metabolic derangements in these individuals and if there is a metabolic intersection where these disorders converge, leading to shared complications. We highlight the impact of puberty-induced hormonal fluctuations, such as growth hormone (GH) surges and sex steroid activity, on disorders like phenylketonuria, urea cycle defects, and fatty acid oxidation disorders. Moreover, we explore the role of dietary interventions in mitigating or exacerbating these effects, emphasizing the importance of balancing nutritional needs during growth spurts. Conclusions: A multidisciplinary approach integrating endocrinology, nutrition, and emerging therapies is advocated to optimize metabolic health during puberty. Addressing these challenges is critical for improving long-term outcomes for individuals with IEMs, particularly during this pivotal developmental phase.
Collapse
Affiliation(s)
- Thomas Lundqvist
- Department of Women’s and Children’s Health, Karolinska Institutet, 171 76 Stockholm, Sweden
- Unit for Pediatric Endocrinology and Metabolic Disorders, Karolinska University Hospital, 171 76 Stockholm, Sweden
| | - Rasmus Stenlid
- Department of Medical Cell Biology, Uppsala University, 751 23 Uppsala, Sweden
| | - Maria Halldin
- Department of Women’s and Children’s Health, Karolinska Institutet, 171 76 Stockholm, Sweden
- Unit for Pediatric Endocrinology and Metabolic Disorders, Karolinska University Hospital, 171 76 Stockholm, Sweden
| |
Collapse
|
5
|
Wang J, Tian S, Du J, Du S, Chen W, Liu Y. The hypothalamic estrogen receptor α pathway is involved in high-intensity interval training-induced visceral fat loss in premenopausal rats. Lipids Health Dis 2025; 24:118. [PMID: 40148843 PMCID: PMC11948781 DOI: 10.1186/s12944-025-02533-6] [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] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 03/14/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Visceral adipose tissue (VAT) is strongly associated with metabolic diseases. Both high-intensity interval training (HIT) and moderate-intensity training (MIT) reduce VAT effectively; however, HIT might mediate greater VAT loss in females. The estrogen receptor α (ERα) pathway may play a key role. The aim of the present study was to confirm the role of adipose/hypothalamic ERα in HIT/MIT-mediated VAT loss, as well as the associated hypothalamic electrophysiology and body catabolism changes in pre- and post-menopausal animal models. METHODS Ovariectomy (OVX) or sham surgeries were conducted to establish pre/postmenopausal female rat models. After distance-matched long-term HIT and MIT interventions, ERα expression in hypothalamic/VAT, as well as food intake, spontaneous physical activity (SPA), VAT mass and morphology, local field potential (LFPs) in paraventricular nuclei (PVN) and excessive post-exercise oxygen consumption (EPOC), were observed. A target chemical block during the post-exercise recovery period was executed to further verify the role of the hypothalamic ERα pathway. RESULTS HIT enhanced the expression of ERα in the hypothalamus rather than VAT in the pre-, but not the postmenopausal group, which was accompanied by elevated LFP power density in α and β bands, enhanced EPOC and larger VAT loss than MIT. Chemical blockade of ERα suppressed EPOC and VAT catabolism mediated by HIT. CONCLUSION During the post-exercise recovery period, the hypothalamic ERα pathway involved in HIT induced EPOC elevation and VAT reduction in premenopausal female rats.
Collapse
Affiliation(s)
- Juanjuan Wang
- School of Physical Education, Hebei Normal University, Shijiazhuang, China
- School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing, China
| | - Shuai Tian
- School of Physical Education, Hebei Normal University, Shijiazhuang, China
| | - Jinchan Du
- School of Physical Education, Hebei Normal University, Shijiazhuang, China
| | - Sihao Du
- School of Physical Education, Hebei Normal University, Shijiazhuang, China
| | - Wei Chen
- School of Physical Education, Hebei Normal University, Shijiazhuang, China
- Provincial Key Lab of Measurement and Evaluation in Human Movement and Bio- Information, Hebei Normal University, Shijiazhuang, China
| | - Yang Liu
- School of Physical Education, Hebei Normal University, Shijiazhuang, China.
- Provincial Key Lab of Measurement and Evaluation in Human Movement and Bio- Information, Hebei Normal University, Shijiazhuang, China.
- School of Physical Education, Hebei Normal University, No. 20, South Second Ring Road East, Shijiazhuang, Hebei, China.
| |
Collapse
|
6
|
Chang RC, Huang Y, To K, Whitlock RS, Nguyen KU, Joemon MC, Lopez M, Deeprompt KG, Shioda T, Blumberg B. Transgenerational Effects of the Obesogen Tributyltin on Metabolic Health in Mice: Interactions With a Western Diet. Endocrinology 2025; 166:bqaf063. [PMID: 40179257 PMCID: PMC11986328 DOI: 10.1210/endocr/bqaf063] [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: 11/04/2024] [Revised: 02/26/2025] [Accepted: 03/30/2025] [Indexed: 04/05/2025]
Abstract
Obesity is a global health crisis, with increasing evidence linking environmental factors such as exposure to endocrine-disrupting chemicals (EDCs) to its development. This study examines the transgenerational effects of exposure to the model obesogen, tributyltin (TBT), on obesity and metabolic health, specifically focusing on how these effects interact with a diet modeling the 50th percentile of US dietary consumption [the Total Western Diet (TWD)]. Pregnant F0 dams were exposed to TBT, and their offspring were subjected at adulthood to different diets, including a high-fat diet and TWD, across multiple subsequent generations (F1-F3). We found that TBT exposure predisposed male offspring to increased fat accumulation, insulin resistance, and metabolic dysfunction, effects that were exacerbated by the TWD. Notably, male offspring displayed elevated leptin levels, hepatic fibrosis, and inflammatory responses under TWD exposure, suggesting an additive or synergistic relationship between obesogen exposure and dietary fat intake. These transgenerational effects were largely absent in female offspring, underscoring sex-specific vulnerabilities to environmental and dietary factors. Our results demonstrated that the combination of prenatal TBT exposure and TWD amplifies metabolic disturbances across generations, highlighting the need to consider both environmental chemicals and dietary patterns in addressing the obesity pandemic. This study underscores the critical role of early-life EDC exposures and dietary factors in shaping long-term metabolic health and the potential for transgenerational programming of susceptibility to obesity and metabolic disorders.
Collapse
Affiliation(s)
- Richard C Chang
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697-2300, USA
| | - Yikai Huang
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697-2300, USA
| | - Kaitlin To
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697-2300, USA
| | - Ryan Scott Whitlock
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697-2300, USA
| | - Katelyn Uyen Nguyen
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697-2300, USA
| | - Michelle Clara Joemon
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697-2300, USA
| | - Miranda Lopez
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697-2300, USA
| | - Kritin Guy Deeprompt
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697-2300, USA
| | - Toshi Shioda
- Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Bruce Blumberg
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697-2300, USA
- Department of Biomedical Engineering, University of California, Irvine, CA 92697-2300, USA
| |
Collapse
|
7
|
Zhou H, Feng S, Cai J, Shao X, Zhu S, Zhou H, Cao Y, Wang R, Lin X, Wang J. Oestrogen suppresses the adipogenesis of fibro/adipogenic progenitors through reactivating the METTL3-ESR1-mediated loop in post-menopausal females. Clin Transl Med 2025; 15:e70206. [PMID: 39875775 PMCID: PMC11774659 DOI: 10.1002/ctm2.70206] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 01/12/2025] [Accepted: 01/19/2025] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Post-menopausal women experience more severe muscular fatty infiltration, though the mechanisms remain unclear. The decline in estrogen levels is considered as a critical physiological alteration during post-menopause. Fibro/adipogenic progenitors (FAPs) are identified as major contributors to muscular fatty infiltration. This study aimed to investigate the detailed mechanism underlying the excessive muscular fatty infiltration in postmenopausal females. METHODS Supraspinatus muscle samples were collected from female patients with or without menopause, and from mice with or without ovariectomy (OVX), to evaluate muscular fatty infiltration and isolated FAPs. The expressions of (estrogen receptor 1) ESR1, methyltransferase-like 3 (METTL3), and adipogenesis ability in FAPs from post-menopausal women and OVX mice were investigated. RNA sequencing (RNA-Seq) was performed to explore the gene expression profiles and potential mechanisms in FAPs from Pdgfrα-CreERT2; Esr1 knockout (Esr1 KO) mice and Esr1 flox/flox (Esr1 f/f) mice. The interplay of the METTL3-ESR1 mediated loop and its role in regulating adipogenesis in FAPs were investigated using dual luciferase reporter assays, chromatin immunoprecipitation (ChIP), and protein and RNA stability assays. The effects of estrogen supplementation on muscular fatty infiltration and locomotor function in OVX mice were evaluated by immunofluorescent staining and functional analysis. RESULTS Decreased expression of ESR1/METTL3 and increased adipogenesis ability in FAPs was found in post-menopausal female. METTL3-mediated m6A methylation promoted ESR1 mRNA stability at the post-transcriptional level in FAPs. METTL3-mediated m6A modification promoted ESR1 expression by stabilizing ESR1 mRNA, while ESR1 acted as a transcription factor that enhanced METTL3 transcription in turn. ESR1 also suppressed the transcription of the adipogenic transcription factor peroxisome proliferator-activated receptor gamma (PPARγ), thereby inhibiting adipogenesis in FAPs. Reactivation of the METTL3-ESR1 mediated loop by estrogen alleviated excessive adipogenesis in FAPs from post-menopausal women, and it also reduced muscular fatty infiltration, and improved locomotor function in OVX mice. CONCLUSION Excessive muscular fatty infiltration in post-menopausal women arose from the disruption of the METTL3-ESR1 mediated loop of FAPs due to estrogen deficiency. Reactivation of the METTL3-ESR1 mediated loop by estrogen may serve as a novel intervention to inhibit excessive adipogenesis of post-menopausal female FAPs, thereby ameliorating muscular fatty infiltration and improving locomotor function in post-menopausal females. KEY POINTS Oestrogen insufficiency disrupted the METTL3ESR1 loop in post-menopausal FAPs, causing excessive muscular fatty infiltration. METTL3-mediated m6A modification stabilized ESR1 mRNA and enhanced ESR1 expression, while increased ESR1 further promoted METTL3 transcription. ESR1 inhibited the transcription of adipogenic factor PPARγ, ameliorating adipogenesis in FAPs. Reactivating the METTL3ESR1 loop via oestrogen in FAPs reduced muscular fatty infiltration and improved locomotor function.
Collapse
Affiliation(s)
- Hao Zhou
- Xinhua Hospital Affiliated to Shanghai Jiaotong University School of MedicineShanghaiChina
| | - Shujing Feng
- School of Exercise and HealthShanghai University of SportShanghaiChina
| | - Jinkui Cai
- Wuhan Third HospitalTongren Hospital of Wuhan UniversityWuhanChina
| | - Xiexiang Shao
- Xinhua Hospital Affiliated to Shanghai Jiaotong University School of MedicineShanghaiChina
| | - Siyuan Zhu
- Department of Hand SurgeryHuashan HospitalFudan UniversityShanghaiChina
| | - Han Zhou
- Xinhua Hospital Affiliated to Shanghai Jiaotong University School of MedicineShanghaiChina
| | - Yongmin Cao
- School of Exercise and HealthShanghai University of SportShanghaiChina
| | - Ru Wang
- School of Exercise and HealthShanghai University of SportShanghaiChina
| | | | - Jianhua Wang
- Xinhua Hospital Affiliated to Shanghai Jiaotong University School of MedicineShanghaiChina
| |
Collapse
|
8
|
Pietrowicz M, Root-Bernstein R. Capsaicin (But Not Other Vanillins) Enhances Estrogen Binding to Its Receptor: Implications for Power Sports and Cancers. Life (Basel) 2025; 15:208. [PMID: 40003617 PMCID: PMC11856108 DOI: 10.3390/life15020208] [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: 12/19/2024] [Revised: 01/20/2025] [Accepted: 01/27/2025] [Indexed: 02/27/2025] Open
Abstract
Capsaicin (CAP), the pain-inducing compound in chili peppers, exerts its effects mainly through the transient receptor potential vanilloid channel 1 (TRPV1), which mediates pain perception and some metabolic functions. CAP has also been demonstrated to improve performance in power sports (but not endurance sports) and does so mainly for females. CAP may also have anti-cancer effects. Many mechanisms have been explored to explain these phenomena, particularly the effects of TRPV1 activation for calcium influx, glucose transporter (GLUT) upregulation and inhibition of insulin (INS) production, but two important ones seem to have been missed. We demonstrate here that CAP binds to both INS and to the estrogen receptor (ESR1), enhancing estradiol binding. Other TRPV1 agonists, such as vanillin, vanillic acid and acetaminophen, have either no effect or inhibit estrogen binding. Notably, TRPV1, ESR1 and INS share significant regions of homology that may aid in identifying the CAP-binding site on the ESR1. Because activation of the estrogen receptor upregulates GLUT expression and thereby glucose transport, we propose that the observed enhancement of performance in power sports, particularly among women, may result, in part, from CAP enhancement of ESR1 function and prevent INS degradation. Chronic exposure to CAP, however, may result in downregulation and internalization of ESR1, as well as TRPV1 stimulation of glucagon-like peptide 1 (GLP-1) expression, both of which downregulate GLUT expression, thereby starving cancer cells of glucose. The binding of capsaicin to the ESR1 may also enhance ESR1 antagonists such as tamoxifen, benefiting some cancer patients.
Collapse
Affiliation(s)
- Maja Pietrowicz
- Independent Researcher, 37430 Tall Oak Dr., Clinton Township, MI 48036, USA;
| | | |
Collapse
|
9
|
Zhou JX, Zheng ZY, Peng ZX, Yang YT, Ni HG. Predictive model in silicon and pathogenicity mechanism of metabolic syndrome: Impacts of heavy metal exposure. JOURNAL OF ENVIRONMENTAL MANAGEMENT 2025; 373:124001. [PMID: 39746257 DOI: 10.1016/j.jenvman.2024.124001] [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: 08/29/2024] [Revised: 12/03/2024] [Accepted: 12/29/2024] [Indexed: 01/04/2025]
Abstract
Although the association between heavy metals in human and the development of metabolic syndrome (MetS) have been extensively studied, the pathogenic mechanism of MetS affected by metals is not clear to date. In this study, a predictive model was developed with machine learning base on the large-scale dataset. These proposed models were evaluated via comparatively analysis of their accuracy and robustness. With the optimal model, two metals significantly correlated with MetS were screened and were employed to infer the pathogenicity mechanism of MetS via molecular docking. Significant associations between heavy metals and MetS were found. Molecular docking provided insights into the interactions between metal ions and key protein receptors involved in metabolic regulation, suggesting a mechanism by which heavy metals interfere with metabolic functions. Specifically, Ba and Cd affect the development of MetS thru their interactions with insulin and estrogen receptors. This study attempted to explore heavy metals' potential roles in MetS at the molecular level. These findings emphasize the importance of addressing environmental exposures in the prevention and treatment of MetS.
Collapse
Affiliation(s)
- Jing-Xuan Zhou
- School of Urban Planning and Design, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Zi-Yi Zheng
- School of Urban Planning and Design, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Zhao-Xing Peng
- School of Urban Planning and Design, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Yu-Ting Yang
- School of Urban Planning and Design, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Hong-Gang Ni
- School of Urban Planning and Design, Peking University Shenzhen Graduate School, Shenzhen, 518055, China.
| |
Collapse
|
10
|
Masemola M, Mendham AE, Micklesfield LK, Pheiffer C, Hawley J, Kengne AP, Chikowore T, Kufe CN, Crowther NJ, Norris S, Storbeck KH, Olsson T, Karpe F, Goedecke JH. Regional Adiposity and Insulin Sensitivity-Interactions With Menopause and HIV in Middle-Aged Black African Women. J Clin Endocrinol Metab 2024; 110:16-29. [PMID: 38950129 DOI: 10.1210/clinem/dgae447] [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: 03/14/2024] [Revised: 06/27/2024] [Accepted: 06/27/2024] [Indexed: 07/03/2024]
Abstract
OBJECTIVE To explore depot-specific functional aspects of adipose tissue, examining the putative role for menopause and HIV status on insulin sensitivity (SI) and beta-cell function in Black South African women. METHODS Women (n = 92) from the Middle-Aged Soweto Cohort, including premenopausal HIV-negative women (n = 21); premenopausal women living with HIV (LWH; n = 11); postmenopausal HIV-negative women (n = 42); and postmenopausal women LWH (n = 18) underwent the following tests: body composition (dual-energy x-ray absorptiometry); fasting bloods for sex hormones, inflammation, and adipokines; frequently sampled intravenous glucose tolerance test for SI and beta-cell function (disposition index, DI); abdominal (aSAT) and gluteal subcutaneous adipose tissue (gSAT) biopsies for cell size, and mRNA expression of adipokines, inflammation, and estrogen receptors (ER). RESULTS Depot-specific associations between gene expression and insulin parameters did not differ by HIV or menopause status. Pooled analysis showed significant models for SI (P = .002) and DI (P = .003). Higher SI was associated with lower leptin and CD11c expression in aSAT and higher adiponectin in gSAT. Higher DI was associated with higher aSAT and gSAT expression of adiponectin, lipoprotein lipase, ERα, and PPARγ, and lower leptin in aSAT. Women LWH had higher expression of adiponectin and lower expression of leptin in both aSAT (P = .002 and P = .005) and gSAT (P = .004 and P = .002), respectively, and a larger proportion of smaller cells in aSAT (P < .001). CONCLUSION Insulin sensitivity and beta-cell function were distinctively associated with aSAT and gSAT. While menopause did not influence these relationships, HIV had a significant effect on adipose tissue, characterized by variations in cell size distribution and transcript levels within the depots.
Collapse
Affiliation(s)
- Maphoko Masemola
- South African Medical Research Council/WITS Developmental Pathways for Health Research Unit (DPHRU), Department of Paediatrics, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2193, South Africa
| | - Amy E Mendham
- South African Medical Research Council/WITS Developmental Pathways for Health Research Unit (DPHRU), Department of Paediatrics, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2193, South Africa
- Riverland Academy of Clinical Excellence, Riverland Mallee Coorong Local Health Network, South Australia Health, Berri, 5343, South Australia, Australia
- Health through Physical Activity, Lifestyle and Sport Research Centre (HPALS), FIMS International Collaborating Centre of Sports Medicine, Division of Physiological Sciences, Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, 7700, South Africa
| | - Lisa K Micklesfield
- South African Medical Research Council/WITS Developmental Pathways for Health Research Unit (DPHRU), Department of Paediatrics, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2193, South Africa
| | - Carmen Pheiffer
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, Cape Town, 7505, South Africa
- Centre for Cardiometabolic Research in Africa (CARMA), Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, Cape Town, 7505, South Africa
- Department of Obstetrics and Gynaecology, Faculty of Health Sciences, University of Pretoria, Pretoria, 0002, South Africa
| | - James Hawley
- Department of Biochemistry, Manchester University, NHS foundation Trust, Manchester, M13 9WL, UK
| | - Andre Pascal Kengne
- Non-Communicable Diseases Research Unit, South African Medical Research Council, Tygerberg, Cape Town, 7505, South Africa
| | - Tinashe Chikowore
- South African Medical Research Council/WITS Developmental Pathways for Health Research Unit (DPHRU), Department of Paediatrics, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2193, South Africa
| | - Clement Nyuyki Kufe
- South African Medical Research Council/WITS Developmental Pathways for Health Research Unit (DPHRU), Department of Paediatrics, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2193, South Africa
- Department of Anaesthesiology, School of Clinical Medicine, University of the Witwatersrand, Faculty of Health Sciences, Johannesburg, 2050, South Africa
| | - Nigel J Crowther
- Department of Chemical Pathology, National Health Laboratory Service and University of the Witwatersrand, Faculty of Health Sciences, Johannesburg, 2192, South Africa
| | - Shane Norris
- South African Medical Research Council/WITS Developmental Pathways for Health Research Unit (DPHRU), Department of Paediatrics, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2193, South Africa
| | - Karl-Heinz Storbeck
- Department of Biochemistry, Stellenbosch University, Stellenbosch, 7600, South Africa
| | - Tommy Olsson
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, 901 87, Sweden
| | - Fredrik Karpe
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 7LE, UK
- National Institute for Health Research, Oxford Biomedical Research Centre, Oxford University Hospitals Trust, Oxford, OX3 7LE, UK
| | - Julia H Goedecke
- South African Medical Research Council/WITS Developmental Pathways for Health Research Unit (DPHRU), Department of Paediatrics, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2193, South Africa
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, Cape Town, 7505, South Africa
| |
Collapse
|
11
|
Hinton A, Neikirk K, Le H, Harris C, Oliver A, Martin P, Gaye A. Estrogen receptors in mitochondrial metabolism: age-related changes and implications for pregnancy complications. AGING ADVANCES 2024; 1:154-171. [PMID: 39839811 PMCID: PMC11748122 DOI: 10.4103/agingadv.agingadv-d-24-00012] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 11/24/2024] [Indexed: 01/23/2025]
Abstract
Estrogen hormones are primarily associated with their role as female sex hormones responsible for primary and secondary sexual development. Estrogen receptors are known to undergo age-dependent decreases due to age-related changes in hormone production. In the mitochondria, estrogen functions by reducing the production of reactive oxygen species in the electron transport chain, inhibiting apoptosis, and regulating mitochondrial DNA content. Moreover, estrogen receptors may be the key components in maintaining mitochondrial membrane potential and structure. Although estrogen plays a crucial role in the development of pregnancy, our understanding of how estrogen receptors change with aging during pregnancy remains limited. During pregnancy, estrogen levels are significantly elevated, with a corresponding upregulation of estrogen receptors, which play various roles in pregnancy. However, the exact role of estrogen receptors in pregnancy complications remains to be further investigated. The paper reviews the role of estrogen receptors in the regulation of mitochondrial metabolism and in pregnancy complications, with a special focus on the effect of age-related changes on estrogen levels and estrogen receptors function. We also address how estrogen maintains mitochondrial function, including reducing the production of reactive oxygen species in the electron transport chain, inhibiting apoptosis, regulating mitochondrial DNA content, and maintaining mitochondrial membrane potential and structure. However, the effects of estrogen on mitochondria-endoplasmic reticulum contacts have not been well studied. Based on these emergent roles in mitochondria, the differential roles of estrogen receptors in pregnancy complications are of great relevance. The paper emphasizes the association between maternal health and estrogen receptors and indicates the need for future research to elucidate the interdependence of estrogen receptor-regulated maternal health with mitochondrial function and their relationship with the gut microbiome. Overall, we summarize the important role of estrogen receptors during pregnancy and highlight the need for further research to better understand the role of estrogen receptors in aging and pregnancy complications. This not only helps to reveal the mechanism underlying the role of estrogen in maternal health but also has potential clinical implications for the development of new therapies targeting age-related diseases and pregnancy complications.
Collapse
Affiliation(s)
- Antentor Hinton
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Kit Neikirk
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Han Le
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Chanel Harris
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, USA
- Department of Biomedical Sciences, Meharry Medical College, Nashville, TN, USA
| | - Ashton Oliver
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, USA
- Department of Biomedical Sciences, Meharry Medical College, Nashville, TN, USA
| | - Pamela Martin
- Department of Biomedical Sciences, Meharry Medical College, Nashville, TN, USA
| | - Amadou Gaye
- Department of Integrative Genomics and Epidemiology, Meharry Medical College, Nashville, TN, USA
| |
Collapse
|
12
|
Gao Q, Huan C, Jia Z, Cao Q, Yuan P, Li X, Wang C, Mao Z, Huo W. SOCS3 Methylation Partially Mediated the Association of Exposure to Triclosan but Not Triclocarban with Type 2 Diabetes Mellitus: A Case-Control Study. Int J Mol Sci 2024; 25:12113. [PMID: 39596180 PMCID: PMC11594987 DOI: 10.3390/ijms252212113] [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] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 11/05/2024] [Accepted: 11/06/2024] [Indexed: 11/28/2024] Open
Abstract
This study aimed to evaluate the association of TCs (triclosan (TCS) and triclocarban) exposure with T2DM and glucose metabolism-related indicators and the mediating effect of SOCS3 methylation on their associations. A total of 956 participants (330 T2DM and 626 controls) were included in this case-control study. Logistic regression and generalized linear models were used to assess the effect of TCs on T2DM and glucose metabolism-related indicators. The dose-response relationship between TCs and T2DM was analyzed by restricted cubic spline. Finally, after evaluating the association between TCs and SOCS3 methylation levels, the mediating effect of SOCS3 methylation on the TC-associated T2DM was estimated. Each 1-unit increase in TCS levels was associated with a 13.2% increase in the risk of T2DM (OR = 1.132, 95% CI: 1.062, 1.207). A linear dose-response relationship was found between TCS and T2DM. TCS was negatively associated with Chr17:76356190 methylation. Moreover, mediation analysis revealed that Chr17:76356190 methylation mediated 14.54% of the relationship between TCS exposure and T2DM. Exposure to TCS was associated with a higher prevalence of T2DM. SOCS3 methylation partially mediated the association of TCS with T2DM. Our findings may provide new insights into the treatment of T2DM, and the study of the biological mechanisms of T2DM.
Collapse
Affiliation(s)
- Qian Gao
- Department of Occupational and Environmental Health, College of Public Health, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, China; (Q.G.)
| | - Changsheng Huan
- Department of Occupational and Environmental Health, College of Public Health, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, China; (Q.G.)
| | - Zexin Jia
- Department of Occupational and Environmental Health, College of Public Health, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, China; (Q.G.)
| | - Qingqing Cao
- Department of Occupational and Environmental Health, College of Public Health, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, China; (Q.G.)
| | - Pengcheng Yuan
- Department of Occupational and Environmental Health, College of Public Health, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, China; (Q.G.)
| | - Xin Li
- Department of Occupational and Environmental Health, College of Public Health, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, China; (Q.G.)
| | - Chongjian Wang
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Zhenxing Mao
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Wenqian Huo
- Department of Occupational and Environmental Health, College of Public Health, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, China; (Q.G.)
| |
Collapse
|
13
|
Antelo-Cea DA, Martínez-Rojas L, Cabrerizo-Ibáñez I, Roudi Rashtabady A, Hernández-Alvarez MI. Regulation of Mitochondrial and Peroxisomal Metabolism in Female Obesity and Type 2 Diabetes. Int J Mol Sci 2024; 25:11237. [PMID: 39457018 PMCID: PMC11508381 DOI: 10.3390/ijms252011237] [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: 10/03/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
Obesity and type 2 diabetes (T2D) are widespread metabolic disorders that significantly impact global health today, affecting approximately 17% of adults worldwide with obesity and 9.3% with T2D. Both conditions are closely linked to disruptions in lipid metabolism, where peroxisomes play a pivotal role. Mitochondria and peroxisomes are vital organelles responsible for lipid and energy regulation, including the β-oxidation and oxidation of very long-chain fatty acids (VLCFAs), cholesterol biosynthesis, and bile acid metabolism. These processes are significantly influenced by estrogens, highlighting the interplay between these organelles' function and hormonal regulation in the development and progression of metabolic diseases, such as obesity, metabolic dysfunction-associated fatty liver disease (MAFLD), and T2D. Estrogens modulate lipid metabolism through interactions with nuclear receptors, like peroxisome proliferator-activated receptors (PPARs), which are crucial for maintaining metabolic balance. Estrogen deficiency, such as in postmenopausal women, impairs PPAR regulation, leading to lipid accumulation and increased risk of metabolic disorders. The disruption of peroxisomal-mitochondrial function and estrogen regulation exacerbates lipid imbalances, contributing to insulin resistance and ROS accumulation. This review emphasizes the critical role of these organelles and estrogens in lipid metabolism and their implications for metabolic health, suggesting that therapeutic strategies, including hormone replacement therapy, may offer potential benefits in treating and preventing metabolic diseases.
Collapse
Affiliation(s)
- Damián A. Antelo-Cea
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain; (D.A.A.-C.); (L.M.-R.); (I.C.-I.); (A.R.R.)
- IBUB Universitat de Barcelona—Institut de Biomedicina de la Universitat de Barcelona, 08028 Barcelona, Spain
| | - Laura Martínez-Rojas
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain; (D.A.A.-C.); (L.M.-R.); (I.C.-I.); (A.R.R.)
| | - Izan Cabrerizo-Ibáñez
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain; (D.A.A.-C.); (L.M.-R.); (I.C.-I.); (A.R.R.)
| | - Ayda Roudi Rashtabady
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain; (D.A.A.-C.); (L.M.-R.); (I.C.-I.); (A.R.R.)
- IBUB Universitat de Barcelona—Institut de Biomedicina de la Universitat de Barcelona, 08028 Barcelona, Spain
| | - María Isabel Hernández-Alvarez
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain; (D.A.A.-C.); (L.M.-R.); (I.C.-I.); (A.R.R.)
- IBUB Universitat de Barcelona—Institut de Biomedicina de la Universitat de Barcelona, 08028 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
14
|
Liu S, Mu Z, Chen X, Xu Y. The impact of sex hormones on metabolic syndrome: univariable and multivariable Mendelian randomization studies. Diabetol Metab Syndr 2024; 16:215. [PMID: 39223618 PMCID: PMC11370018 DOI: 10.1186/s13098-024-01443-4] [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: 05/25/2024] [Accepted: 08/07/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Observational studies have found associations between sex hormones and metabolic syndrome(Mets), but the causal relationships remains unclear. This study utilizes univariable and multivariable Mendelian randomization (MR) to elucidate the associations between sex hormones (including sex hormone-binding globulin(SHBG), estradiol(E2), testosterone(T)) and Mets and its subtypes (including waist circumference(WC), fasting blood glucose(FBG), high blood pressure(HBP), high-density lipoprotein(HDL-C), triglycerides(TG)). METHODS We utilized summary data from large-scale genome-wide association studies. Univariable Mendelian randomization (UMVMR) analysis was primarily conducted using the inverse variance weighted method (IVW), with secondary analyses employing the weighted median, MR-Egger regression, simple mode method, and weighted mode method. Subsequently, multivariable Mendelian randomization (MVMR) was employed to assess the causal relationships between SHBG, T, E2, and MetS and its components: WC, FPG, HBP, HDL-C, and TG. Sensitivity analyses were conducted to assess result reliability. RESULTS Genetically predicted SHBG was significantly negatively associated with MetS (UMVMR: β=-0.72; 95% CI = 0.41 to 0.57; P = 1.28e-17; MVMR: β=-0.60; 95% CI=-0.83 to -0.38; P < 0.001). Positive causal relationships were observed between SHBG and WC(MVMR: β = 0.10; 95% CI = 0.03 to 0.17; P = 0.01) and HDL-C (MVMR: β = 0.41; 95% CI = 0.21 to 0.60; P < 0.001), while negative causal relationships were found between SHBG and HBP (MVMR: β=-0.02; 95% CI=-0.04 to -0.00; P = 0.02), TG (MVMR: β=-0.48; 95% CI=-0.70 to -0.26; P < 0.001). Genetically predicted E2 exhibited a negative association with TG (MVMR: β=-1.49; 95% CI=-2.48 to -0.50; P = 0.003). Genetically predicted T was negatively associated with TG (MVMR: β=-0.36; 95% CI=-0.71 to -0.00; P = 0.049) and WC (MVMR: β=-0.13; 95% CI=-0.24 to -0.02; P = 0.02), with inconsistent sensitivity analyses. Additionally, No other causal associations were found. CONCLUSION Our study indicates that SHBG is a protective factor for MetS, potentially delaying its onset and progression through improvements in HBP and TG. Furthermore, T and E2 may improve TG levels, with T also reducing WC levels. Importantly, our study provides new insights for the prevention and treatment of MetS.
Collapse
Affiliation(s)
- Siyuan Liu
- The Third Clinical Medical College of Zhejiang, University of Traditional Chinese Medicine, Hangzhou, Zhejiang, China
| | - Zhuosong Mu
- Jiangnan Hospital Affiliated to Zhejiang, Chinese Medical University (Hangzhou Xiaoshan Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, China
| | - Xinyi Chen
- The Third Clinical Medical College of Zhejiang, University of Traditional Chinese Medicine, Hangzhou, Zhejiang, China
| | - Yingying Xu
- The Third Affiliated Hospital of Zhejiang, University of Traditional Chinese Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
15
|
Ihalainen JK, Mikkonen RS, Ackerman KE, Heikura IA, Mjøsund K, Valtonen M, Hackney AC. Beyond Menstrual Dysfunction: Does Altered Endocrine Function Caused by Problematic Low Energy Availability Impair Health and Sports Performance in Female Athletes? Sports Med 2024; 54:2267-2289. [PMID: 38995599 PMCID: PMC11393114 DOI: 10.1007/s40279-024-02065-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/04/2024] [Indexed: 07/13/2024]
Abstract
Low energy availability, particularly when problematic (i.e., prolonged and/or severe), has numerous negative consequences for health and sports performance as characterized in relative energy deficiency in sport. These consequences may be driven by disturbances in endocrine function, although scientific evidence clearly linking endocrine dysfunction to decreased sports performance and blunted or diminished training adaptations is limited. We describe how low energy availability-induced changes in sex hormones manifest as menstrual dysfunction and accompanying hormonal dysfunction in other endocrine axes that lead to adverse health outcomes, including negative bone health, impaired metabolic activity, undesired outcomes for body composition, altered immune response, problematic cardiovascular outcomes, iron deficiency, as well as impaired endurance performance and force production, all of which ultimately may influence athlete health and performance. Where identifiable menstrual dysfunction indicates hypothalamic-pituitary-ovarian axis dysfunction, concomitant disturbances in other hormonal axes and their impact on the athlete's health and sports performance must be recognized as well. Given that the margin between podium positions and "losing" in competitive sports can be very small, several important questions regarding low energy availability, endocrinology, and the mechanisms behind impaired training adaptations and sports performance have yet to be explored.
Collapse
Affiliation(s)
- Johanna K Ihalainen
- Biology of Physical Activity, Faculty of Sport and Health Sciences, University of Jyväskylä, PO Box 35, 40014, Jyväskylä, Finland.
- Finnish Institute of High Performance Sport KIHU, Jyväskylä, Finland.
| | - Ritva S Mikkonen
- Biology of Physical Activity, Faculty of Sport and Health Sciences, University of Jyväskylä, PO Box 35, 40014, Jyväskylä, Finland
- Sports Technology Unit, Faculty of Sport and Health Sciences, University of Jyväskylä, Vuokatti, Finland
| | - Kathryn E Ackerman
- Wu Tsai Female Athlete Program, Division of Sports Medicine, Boston Children's Hospital, Boston, MA, USA
- Neuroendocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ida A Heikura
- Canadian Sport Institute-Pacific, Victoria, BC, Canada
- Exercise Science, Physical and Health Education, University of Victoria, Victoria, BC, Canada
| | - Katja Mjøsund
- Paavo Nurmi Centre and Unit for Health and Physical Activity, University of Turku, Turku, Finland
- National Olympic Training Centre Helsinki, Helsinki, Finland
| | - Maarit Valtonen
- Finnish Institute of High Performance Sport KIHU, Jyväskylä, Finland
| | - Anthony C Hackney
- Department of Exercise and Sport Science, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
16
|
Ojedo-Martín C, Rodríguez-López ES, Acevedo-Gómez MB, Úbeda-D’Ocasar E, de-Diego MV, Lara B. At What Point in the Menstrual Cycle Are the Pelvic Floor Muscles at Their Weakest? J Funct Morphol Kinesiol 2024; 9:135. [PMID: 39189220 PMCID: PMC11348105 DOI: 10.3390/jfmk9030135] [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: 07/15/2024] [Revised: 08/02/2024] [Accepted: 08/06/2024] [Indexed: 08/28/2024] Open
Abstract
Pelvic floor muscle (PFM) strength is a critical factor for optimal pelvic floor function. Fluctuations in strength values based on different phases of the menstrual cycle (MC) could signify a need for a paradigm shift in evaluating, approaching, and planning training. This research aims to examine and contrast the pelvic floor muscle strength during different phases of the menstrual cycle. A prospective observational study employing digital assessment with the modified Oxford scale and vaginal dynamometry measurements was performed, in order to assess the baseline strength and the contraction strength of the PFMs in eumenorrheic females at three different phases of the MC: the early follicular phase (EFP), the late follicular phase (LFP), and the mid-luteal phase (MLP). During two complete cycles, tympanic temperature and body weight were measured and the urinary luteinizing hormone concentration was tested to determine the time of ovulation. In total, 216 dynamometric measurements of PFM strength were obtained from eighteen nulliparous women (25.72 ± 5.03 years). There were no differences between the baseline strength (p = 0.886) and the contraction strength (p = 0.756) with the dynamometric speculum. In the post hoc analysis, the baseline strength, contraction strength, and strength showed no significant differences between MC phases. As no differences in PFM strength in women were found, the PFMs do not seem to be weaker at any time during the menstrual cycle. It appears that the assessment, establishment, and monitoring of a PFM training program could be initiated at any point in the cycle.
Collapse
Affiliation(s)
- Cristina Ojedo-Martín
- Physiotherapy and Health Research Group (FYSA), Department of Physiotherapy, Faculty of Health Sciences-HM Hospitals, University Camilo José Cela, 28014 Madrid, Spain; (C.O.-M.); (M.B.A.-G.); (E.Ú.-D.)
- Department of Physiotherapy, Faculty of Health Sciences-HM Hospitals, University Camilo José Cela, 28014 Madrid, Spain
| | - Elena Sonsoles Rodríguez-López
- Physiotherapy and Health Research Group (FYSA), Department of Physiotherapy, Faculty of Health Sciences-HM Hospitals, University Camilo José Cela, 28014 Madrid, Spain; (C.O.-M.); (M.B.A.-G.); (E.Ú.-D.)
- Department of Physiotherapy, Faculty of Health Sciences-HM Hospitals, University Camilo José Cela, 28014 Madrid, Spain
| | - María Barbaño Acevedo-Gómez
- Physiotherapy and Health Research Group (FYSA), Department of Physiotherapy, Faculty of Health Sciences-HM Hospitals, University Camilo José Cela, 28014 Madrid, Spain; (C.O.-M.); (M.B.A.-G.); (E.Ú.-D.)
| | - Edurne Úbeda-D’Ocasar
- Physiotherapy and Health Research Group (FYSA), Department of Physiotherapy, Faculty of Health Sciences-HM Hospitals, University Camilo José Cela, 28014 Madrid, Spain; (C.O.-M.); (M.B.A.-G.); (E.Ú.-D.)
- Department of Physiotherapy, Faculty of Health Sciences-HM Hospitals, University Camilo José Cela, 28014 Madrid, Spain
| | | | - Beatriz Lara
- Exercise Physiology Laboratory, Faculty of Health Sciences-HM Hospitals, University Camilo José Cela, 28014 Madrid, Spain;
| |
Collapse
|
17
|
Osmancevic A, Allison M, Miljkovic I, Vella CA, Ouyang P, Trimpou P, Daka B. Levels of Sex Hormones and Abdominal Muscle Composition in Men from The Multi-Ethnic Study of Atherosclerosis. Sci Rep 2024; 14:16114. [PMID: 38997435 PMCID: PMC11245501 DOI: 10.1038/s41598-024-66948-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 07/05/2024] [Indexed: 07/14/2024] Open
Abstract
Information on the associations of testosterone levels with abdominal muscle volume and density in men is limited, while the role of estradiol and SHBG on these muscle characteristics are unclear. Therefore, this study aimed to investigate the association between fasting serum sex hormones and CT-derived abdominal muscle area and radiodensity in adult men. Conducted as a cross sectional observational study using data from the Multi-Ethnic Study of Atherosclerosis, our analyses focused on a community-based sample of 907 men aged 45-84 years, with 878 men having complete data. CT scans of the abdomen were interrogated for muscle characteristics, and multivariable linear regressions were used to test the associations. After adjustment for relevant factors, higher levels of both total testosterone and estradiol were associated with higher abdominal muscle area (1.74, 0.1-3.4, and 1.84, 0.4-3.3, respectively). In the final analyses, levels of total testosterone showed a positive association, while an inverse relationship was observed for SHBG with abdominal muscle radiodensity (0.3, 0.0-0.6, and - 0.33, - 0.6 to - 0.1, respectively). Our results indicate a complex association between sex hormones and abdominal muscle characteristics in men. Specifically, total testosterone and estradiol were associated with abdominal muscle area, while only total testosterone was associated with muscle radiodensity and SHBG was inversely associated with muscle radiodensity.Clinical Trial: NCT00005487.
Collapse
Grants
- 75N92020D00005 NHLBI NIH HHS
- N01HC95160 NHLBI NIH HHS
- N01HC95163 NHLBI NIH HHS
- UL1-TR-000040, UL1-TR-001079, and UL1-TR-001420 NCATS NIH HHS
- N01HC95168 NHLBI NIH HHS
- N01HC95165 NHLBI NIH HHS
- N01HC95159 NHLBI NIH HHS
- 75N92020D00007 NHLBI NIH HHS
- HHSN268201500003I NHLBI NIH HHS
- N01HC95167 NHLBI NIH HHS
- UL1 TR000040 NCATS NIH HHS
- 75N92020D00002 NHLBI NIH HHS
- HHSN268201500003C NHLBI NIH HHS
- UL1 TR001079 NCATS NIH HHS
- ALFGBG-966255 VGR Regional Research and Development Council Grants
- 75N92020D00001 NHLBI NIH HHS
- N01HC95169 NHLBI NIH HHS
- 75N92020D00001, HHSN268201500003I, N01-HC-95159, 75N92020D00005, N01-HC-95160, 75N92020D00002, N01-HC-95161, 75N92020D00003, N01-HC-95162, 75N92020D00006, N01-HC-95163, 75N92020D00004, N01-HC-95164, 75N92020D00007, N01-HC-95165, N01-HC-95166, N01-HC-95167, N01-HC-95168 and N01-HC-95169 NHLBI NIH HHS
- N01HC95164 NHLBI NIH HHS
- N01HC95162 NHLBI NIH HHS
- 75N92020D00003 NHLBI NIH HHS
- N01HC95161 NHLBI NIH HHS
- UL1 TR001420 NCATS NIH HHS
- 75N92020D00004 NHLBI NIH HHS
- 75N92020D00006 NHLBI NIH HHS
- N01HC95166 NHLBI NIH HHS
- The Local Research and Development Council Göteborg och Södra Bohuslän
- National Heart, Lung, and Blood Institute
- National Center for Advancing Translational Sciences
- University of Gothenburg
Collapse
Affiliation(s)
- Amar Osmancevic
- General Practice / Family Medicine, School of Public Health and Community Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Matthew Allison
- Division of Preventive Medicine, School of Medicine, UC San Diego, San Diego, CA, USA
| | - Iva Miljkovic
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Chantal A Vella
- Department of Movement Sciences, University of Idaho, Moscow, ID, USA
| | - Pamela Ouyang
- Institute for Clinical and Translational Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Penelope Trimpou
- Department of Endocrinology, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Bledar Daka
- General Practice / Family Medicine, School of Public Health and Community Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
18
|
Clontz AD, Gan E, Hursting SD, Bae-Jump VL. Effects of Weight Loss on Key Obesity-Related Biomarkers Linked to the Risk of Endometrial Cancer: A Systematic Review and Meta-Analysis. Cancers (Basel) 2024; 16:2197. [PMID: 38927903 PMCID: PMC11201950 DOI: 10.3390/cancers16122197] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 05/26/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
Endometrial cancer (EC) includes various histologic types, with estrogen-dependent endometrioid carcinoma being the most common. Obesity significantly increases the risk of developing this type, especially in postmenopausal women, due to elevated estrogen production by adipocytes. This review examines the impact of weight loss from different interventions on reducing obesity-related risk factors for endometrioid EC. A systematic review and meta-analysis were conducted on three weight loss interventions: bariatric surgery, pharmacotherapy, and lifestyle changes. The effects of these interventions on inflammatory biomarkers (CRP, TNF-α, IL-6) and hormones (leptin, estrogen) were analyzed. Data from controlled studies were pooled to assess the significance of weight loss in reducing these biomarkers. Despite heterogeneity, bariatric surgery resulted in an overall 25.8% weight reduction, outperforming lifestyle and pharmacotherapy interventions. Weight loss reduced CRP levels by 33.5% and IL-6 levels by 41.9%. TNF-α levels decreased by 13% with percent weight loss over 7%. Leptin levels also decreased significantly, although the exact weight loss percentage was not statistically significant. Weight loss effectively reduces proinflammatory markers and hormones associated with increased risk of endometrioid EC. The strengths of this review include a comprehensive examination of different weight-loss interventions and a large pool of participants. However, limitations include high heterogeneity among studies and only 43% of the participants being postmenopausal. Limited data on sex hormones and racial disparities underscore the need for further research.
Collapse
Affiliation(s)
- Angela D. Clontz
- Department of Nutrition and Nutrition Research Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (A.D.C.); (S.D.H.)
| | - Emma Gan
- Department of Medicine, Imperial College London, London SW7 2AZ, UK;
| | - Stephen D. Hursting
- Department of Nutrition and Nutrition Research Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (A.D.C.); (S.D.H.)
| | - Victoria L. Bae-Jump
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
19
|
Jiang Z, Huang B, Cui Z, Lu Z, Ma H. Synergistic effect of genistein and adiponectin reduces fat deposition in chicken hepatocytes by activating the ERβ-mediated SIRT1-AMPK signaling pathway. Poult Sci 2024; 103:103734. [PMID: 38636201 PMCID: PMC11040169 DOI: 10.1016/j.psj.2024.103734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 03/31/2024] [Accepted: 04/02/2024] [Indexed: 04/20/2024] Open
Abstract
Dietary supplementation with bioactive substances that can regulate lipid metabolism is an effective approach for reducing excessive fat deposition in chickens. Genistein (GEN) has the potential to alleviate fat deposition; however, the underlying mechanism of GEN's fat-reduction action in chickens remains unclear. Therefore, the present study aimed to explore the underlying mechanism of GEN on the reduction of fat deposition from a novel perspective: intercellular transmission of adipokine between adipocytes and hepatocytes. The findings showed that GEN enhanced the secretion of adiponectin (APN) in chicken adipocytes, and the enhancement effect of GEN was completely blocked when the cells were pretreated with inhibitors targeting estrogen receptor β (ERβ) or proliferator-activated receptor γ (PPARγ) signals, respectively. Furthermore, the results demonstrated that both co-treatment with GEN and APN or treatment with the medium supernatant (Med SUP) derived from chicken adipocytes treated with GEN significantly decreased the content of triglyceride and increased the protein levels of ERβ, Sirtuin 1 (SIRT1) and phosphor-AMP-activated protein kinase (p-AMPK) in chicken hepatocytes compared to the cells treated with GEN or APN alone. Moreover, the increase in the protein levels of SIRT1 and p-AMPK induced by GEN and APN co-treatment or Med SUP treatment were blocked in chicken hepatocytes pretreated with the inhibitor of ERβ signals. Importantly, the up-regulatory effect of GEN and APN co-treatment or Med SUP treatment on the protein level of p-AMPK was also blocked in chicken hepatocytes pretreated with a SIRT1 inhibitor; however, the increase in the protein level of SIRT1 induced by GEN and APN co-treatment or Med SUP treatment was not reversed when the hepatocytes were pretreated with an AMPK inhibitor. In conclusion, the present study demonstrated that GEN enhanced APN secretion by activating the ERβ-Erk-PPARγ signaling pathway in chicken adipocytes. Subsequently, adipocyte-derived APN synergized with GEN to activate the ERβ-mediated SIRT1-AMPK signaling pathway in chicken hepatocytes, ultimately reducing fat deposition. These findings provide substantial evidence from a novel perspective, supporting the potential use of GEN as a dietary supplement to prevent excessive fat deposition in poultry.
Collapse
Affiliation(s)
- Zhihao Jiang
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Benzeng Huang
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Ziyi Cui
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Ze Lu
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Haitian Ma
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
20
|
Shay D, Welly R, Mao J, Kinkade J, Brown JK, Rosenfeld CS, Vieira-Potter VJ. Knockdown of Esr1 from DRD1-Rich Brain Regions Affects Adipose Tissue Metabolism: Potential Crosstalk between Nucleus Accumbens and Adipose Tissue. Int J Mol Sci 2024; 25:6130. [PMID: 38892320 PMCID: PMC11172510 DOI: 10.3390/ijms25116130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 05/20/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
Declining estrogen (E2) leads to physical inactivity and adipose tissue (AT) dysfunction. Mechanisms are not fully understood, but E2's effects on dopamine (DA) activity in the nucleus accumbens (NAc) brain region may mediate changes in mood and voluntary physical activity (PA). Our prior work revealed that loss of E2 robustly affected NAc DA-related gene expression, and the pattern correlated with sedentary behavior and visceral fat. The current study used a new transgenic mouse model (D1ERKO) to determine whether the abolishment of E2 receptor alpha (ERα) signaling within DA-rich brain regions affects PA and AT metabolism. Adult male and female wild-type (WT) and D1ERKO (KD) mice were assessed for body composition, energy intake (EE), spontaneous PA (SPA), and energy expenditure (EE); underwent glucose tolerance testing; and were assessed for blood biochemistry. Perigonadal white AT (PGAT), brown AT (BAT), and NAc brain regions were assessed for genes and proteins associated with DA, E2 signaling, and metabolism; AT sections were also assessed for uncoupling protein (UCP1). KD mice had greater lean mass and EE (genotype effects) and a visible change in BAT phenotype characterized by increased UCP1 staining and lipid depletion, an effect seen only among females. Female KD had higher NAc Oprm1 transcript levels and greater PGAT UCP1. This group tended to have improved glucose tolerance (p = 0.07). NAc suppression of Esr1 does not appear to affect PA, yet it may directly affect metabolism. This work may lead to novel targets to improve metabolic dysfunction following E2 loss, possibly by targeting the NAc.
Collapse
Affiliation(s)
- Dusti Shay
- Department of Nutrition and Exercise Physiology, Division of Food, Nutrition and Exercise Sciences, CAFNR, University of Missouri, Columbia, MO 65211, USA; (D.S.)
| | - Rebecca Welly
- Department of Nutrition and Exercise Physiology, Division of Food, Nutrition and Exercise Sciences, CAFNR, University of Missouri, Columbia, MO 65211, USA; (D.S.)
| | - Jiude Mao
- Biomedical Sciences, University of Missouri, E102 Veterinary Medicine Building, Columbia, MO 65211, USA (C.S.R.)
| | - Jessica Kinkade
- Biomedical Sciences, University of Missouri, E102 Veterinary Medicine Building, Columbia, MO 65211, USA (C.S.R.)
| | - Joshua K. Brown
- Department of Nutrition and Exercise Physiology, Division of Food, Nutrition and Exercise Sciences, CAFNR, University of Missouri, Columbia, MO 65211, USA; (D.S.)
| | - Cheryl S. Rosenfeld
- Biomedical Sciences, University of Missouri, E102 Veterinary Medicine Building, Columbia, MO 65211, USA (C.S.R.)
- MU Institute of Data Science and Informatics, University of Missouri, E102 Veterinary Medicine Building, Columbia, MO 65211, USA
- Genetics Area Program, University of Missouri, E102 Veterinary Medicine Building, Columbia, MO 65211, USA
- Thompson Center for Autism and Neurobehavioral Disorders, University of Missouri, E102 Veterinary Medicine Building, Columbia, MO 65211, USA
| | - Victoria J. Vieira-Potter
- Department of Nutrition and Exercise Physiology, Division of Food, Nutrition and Exercise Sciences, CAFNR, University of Missouri, Columbia, MO 65211, USA; (D.S.)
| |
Collapse
|
21
|
Libby AE, Solt CM, Jackman MR, Sherk VD, Foright RM, Johnson GC, Nguyen TT, Breit MJ, Hulett N, Rudolph MC, Roberson PA, Wellberg EA, Jambal P, Scalzo RL, Higgins J, Kumar TR, Wierman ME, Pan Z, Shankar K, Klemm DJ, Moreau KL, Kohrt WM, MacLean PS. Effects of follicle-stimulating hormone on energy balance and tissue metabolic health after loss of ovarian function. Am J Physiol Endocrinol Metab 2024; 326:E626-E639. [PMID: 38536037 PMCID: PMC11208003 DOI: 10.1152/ajpendo.00400.2023] [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: 11/29/2023] [Revised: 02/29/2024] [Accepted: 03/20/2024] [Indexed: 05/01/2024]
Abstract
Loss of ovarian function imparts increased susceptibility to obesity and metabolic disease. These effects are largely attributed to decreased estradiol (E2), but the role of increased follicle-stimulating hormone (FSH) in modulating energy balance has not been fully investigated. Previous work that blocked FSH binding to its receptor in mice suggested this hormone may play a part in modulating body weight and energy expenditure after ovariectomy (OVX). We used an alternate approach to isolate the individual and combined contributions of FSH and E2 in mediating energy imbalance and changes in tissue-level metabolic health. Female Wistar rats were ovariectomized and given the gonadotropin releasing hormone (GnRH) antagonist degarelix to suppress FSH production. E2 and FSH were then added back individually and in combination for a period of 3 wk. Energy balance, body mass composition, and transcriptomic profiles of individual tissues were obtained. In contrast to previous studies, suppression and replacement of FSH in our paradigm had no effect on body weight, body composition, food intake, or energy expenditure. We did, however, observe organ-specific effects of FSH that produced unique transcriptomic signatures of FSH in retroperitoneal white adipose tissue. These included reductions in biological processes related to lipogenesis and carbohydrate transport. In addition, rats administered FSH had reduced liver triglyceride concentration (P < 0.001), which correlated with FSH-induced changes at the transcriptomic level. Although not appearing to modulate energy balance after loss of ovarian function in rats, FSH may still impart tissue-specific effects in the liver and white adipose tissue that might affect the metabolic health of those organs.NEW & NOTEWORTHY We find no effect of follicle-stimulating hormone (FSH) on energy balance using a novel model in which rats are ovariectomized, subjected to gonadotropin-releasing hormone antagonism, and systematically given back FSH by osmotic pump. However, tissue-specific effects of FSH on adipose tissue and liver were observed in this study. These include unique transcriptomic signatures induced by the hormone and a stark reduction in hepatic triglyceride accumulation.
Collapse
Affiliation(s)
- Andrew E Libby
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Claudia M Solt
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Matthew R Jackman
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Vanessa D Sherk
- Department of Orthopedics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Center for Scientific Review, National Institutes of Health, Bethesda, Maryland, United States
| | - Rebecca M Foright
- Department of Anatomy and Cell Biology, University of Kansas Medical Campus, Kansas City, Kansas, United States
| | - Ginger C Johnson
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Thi-Tina Nguyen
- Section of Nutrition, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Matthew J Breit
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Nicholas Hulett
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Michael C Rudolph
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Campus, Oklahoma City, Oklahoma, United States
| | - Paul A Roberson
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Elizabeth A Wellberg
- Stephenson Cancer Center, University of Oklahoma Health Sciences Campus, Oklahoma City, Oklahoma, United States
| | - Purevsuren Jambal
- Section of Nutrition, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Rebecca L Scalzo
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Janine Higgins
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - T Rajendra Kumar
- Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Margaret E Wierman
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Zhaoxing Pan
- Section of Endocrinology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Kartik Shankar
- Section of Nutrition, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Dwight J Klemm
- Cardiovascular Pulmonary Research Laboratory, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Kerrie L Moreau
- Division of Geriatric Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Wendy M Kohrt
- Division of Geriatric Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Paul S MacLean
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| |
Collapse
|
22
|
Dongre P, Majumdar A. Network pharmacology analysis of Chandraprabha Vati: A new hope for the treatment of Metabolic Syndrome. J Ayurveda Integr Med 2024; 15:100902. [PMID: 38821011 PMCID: PMC11177199 DOI: 10.1016/j.jaim.2024.100902] [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: 10/02/2023] [Revised: 12/25/2023] [Accepted: 02/01/2024] [Indexed: 06/02/2024] Open
Abstract
BACKGROUND Drug research is increasingly using Network Pharmacology (NP) to tackle complex conditions like Metabolic Syndrome (MetS), which is characterized by obesity, hyperglycemia, and dyslipidemia. Single-action drugs are inadequate to treat MetS, which is marked by a range of complications including glucose intolerance, hyperlipidemia, mitochondrial dysfunction, and inflammation. OBJECTIVES To analyze Chandraprabha vati using Network Pharmacology to assess its potential in alleviating MetS-related complications. MATERIAL AND METHODS The genes related to MetS, inflammation, and the target genes of the CPV components were identified using network pharmacology tools like DisgNET and BindingDB. Followed by mapping of the CPV target genes with the genes implicated in MetS and inflammation to identify putative potential targets. Gene ontology, pathway enrichment analysis, and STRING database were employed for further exploration. Furthermore, drug-target-protein interactions network were visualized using Cytoscape 3.9.1. RESULTS The results showed that out of the 225 target genes of the CPV components, 33 overlapping and 19 non-overlapping genes could be potential targets for MetS. Similarly, 14 overlapping and 7 non-overlapping genes could be potential targets for inflammation. The CPV bioactives target genes were found to be involved in lipid and insulin homeostasis via several pathways revealed by the pathway analysis. The importance of CPV in treating MetS was supported by GO enrichment data; this could be due to its potential to influence pathways linked to metabolism, ER stress, mitochondrial dysfunction, oxidative stress, and inflammation. CONCLUSIONS These results offer a promising approach to developing treatment and repurposing CPV for complex conditions such as MetS.
Collapse
Affiliation(s)
- Prashant Dongre
- Department of Pharmacology, Bombay College of Pharmacy, Kalina, Santacruz (E), Mumbai, 400098, India
| | - Anuradha Majumdar
- Department of Pharmacology, Bombay College of Pharmacy, Kalina, Santacruz (E), Mumbai, 400098, India.
| |
Collapse
|
23
|
Zahr T, Boda VK, Ge J, Yu L, Wu Z, Que J, Li W, Qiang L. Small molecule conjugates with selective estrogen receptor β agonism promote anti-aging benefits in metabolism and skin recovery. Acta Pharm Sin B 2024; 14:2137-2152. [PMID: 38799642 PMCID: PMC11119546 DOI: 10.1016/j.apsb.2024.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 12/13/2023] [Accepted: 01/05/2024] [Indexed: 05/29/2024] Open
Abstract
Estrogen is imperative to mammalian reproductivity, metabolism, and aging. However, the hormone activating estrogen receptor (ERs) α can cause major safety concerns due to the enrichment of ERα in female tissues and certain malignancies. In contrast, ERβ is more broadly expressed in metabolic tissues and the skin. Thus, it is desirable to generate selective ERβ agonist conjugates for maximizing the therapeutic effects of ERs while minimizing the risks of ERα activation. Here, we report the design and production of small molecule conjugates containing selective non-steroid ERβ agonists Gtx878 or genistein. Treatment of aged mice with our synthesized conjugates improved aging-associated declines in insulin sensitivity, visceral adipose integrity, skeletal muscle function, and skin health, with validation in vitro. We further uncovered the benefits of ERβ conjugates in the skin using two inducible skin injury mouse models, showing increased skin basal cell proliferation, epidermal thickness, and wound healing. Therefore, our ERβ-selective agonist conjugates offer novel therapeutic potential to improve aging-associated conditions and aid in rejuvenating skin health.
Collapse
Affiliation(s)
- Tarik Zahr
- Naomi Berrie Diabetes Center, Columbia University, New York, NY 10032, USA
- Department of Molecular Pharmacology and Therapeutics, Columbia University, New York, NY 10032, USA
| | - Vijay K. Boda
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Drug Discovery Center, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Jian Ge
- Division of Digestive and Liver Diseases, Columbia University, New York, NY 10032, USA
- Center for Human Development, Columbia University, New York, NY 10027, USA
| | - Lexiang Yu
- Naomi Berrie Diabetes Center, Columbia University, New York, NY 10032, USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Zhongzhi Wu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Drug Discovery Center, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Jianwen Que
- Division of Digestive and Liver Diseases, Columbia University, New York, NY 10032, USA
- Center for Human Development, Columbia University, New York, NY 10027, USA
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Wei Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Drug Discovery Center, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Li Qiang
- Naomi Berrie Diabetes Center, Columbia University, New York, NY 10032, USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| |
Collapse
|
24
|
Kor A, Sungu N, Karabulut Ş, Erten Ş. Renal tubular estrogen ß receptors are expressed at high levels in small vessel vasculitis and are primarily localized in the distal tubule. FASEB J 2024; 38:e23497. [PMID: 38376916 DOI: 10.1096/fj.202302362rr] [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: 11/16/2023] [Revised: 01/29/2024] [Accepted: 02/05/2024] [Indexed: 02/21/2024]
Abstract
This study investigated the possible roles of renal estrogen receptors (ER) in glomerulonephritis associated with small vessel vasculitis. The relationships of ERs were investigated in antineutrophilic cytoplasmic antibody (ANCA)-associated glomerulonephritis and immunoglobulin A (IgA) nephropathy groups, which are small vessel vasculitis subtypes with two different glomerulonephritis development pathophysiologies. The design of this study was prepared as a retrospective cohort study. The study included 42 patients with ANCA-associated vasculitis and 18 with IgA nephropathy in the small vessel vasculitis group. For the control group, intact renal tissues of 28 patients who underwent nephrectomy due to renal cell carcinoma were used. Renal biopsy samples of the groups were stained with ER beta (ß) and ER alpha (α). Tubular ER ß expression score (TERßES) median values were found to be significantly higher in ANCA- associated vasculitis (B = 0.724, OR [95%CI]: 2.064 [1.141-3.731], p = .016) and IgA nephropathy (B = 0.898, OR [95%CI]: 2.454 [1.307-4.609], p = .005) than in intact kidney tissue. It was determined that tubular ERß was most frequently localized in the distal tubule at 57.9% and the second most common in the proximal tubule at 20.4%. The expression of tubular ERß is increased in glomerulonephritis due to small vessel vasculitis. Tubular ERßs are most commonly localized in the distal tubule. Further studies are needed to understand the physiological and pathophysiological effects of altered renal ER levels in small vessel vasculitis.
Collapse
Affiliation(s)
- Ahmet Kor
- Department of Rheumatology, Ministry of Health, Aksaray University Training and Research Hospital, Aksaray, Turkey
| | - Nuran Sungu
- Department of Medical Pathology, Faculty of Medicine Ankara Bilkent City Hospital, Ankara Yıldırım Beyazıt University, Ankara, Turkey
| | - Şefika Karabulut
- Gulhane Department of Microbiology, Virology, Health Sciences University, Ankara, Turkey
| | - Şükran Erten
- Department of Rheumatology, Faculty of Medicine Ankara Bilkent City Hospital, Ankara Yıldırım Beyazıt University, Ankara, Turkey
| |
Collapse
|
25
|
Wu T, Ding K, Wang C, Lin G, Xie C, Chen X, Li Q, Yu F, Mao Y, Hong W, Lu L, Li S. G-protein-coupled estrogen receptor 1 promotes peritoneal metastasis of gastric cancer through nicotinamide adenine dinucleotide kinase 1-mediated redox modulation. FASEB J 2024; 38:e23449. [PMID: 38315451 DOI: 10.1096/fj.202301172rrrr] [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: 06/12/2023] [Revised: 12/20/2023] [Accepted: 01/10/2024] [Indexed: 02/07/2024]
Abstract
Adipose tissue is the second most important site of estrogen production, where androgens are converted into estrogen by aromatase. While gastric cancer patients often develop adipocyte-rich peritoneal metastasis, the underlying mechanism remains unclear. In this study, we identified the G-protein-coupled estrogen receptor (GPER1) as a promoter of gastric cancer peritoneal metastasis. Functional in vitro studies revealed that β-Estradiol (E2) or the GPER1 agonist G1 inhibited anoikis in gastric cancer cells. Additionally, genetic overexpression or knockout of GPER1 significantly inhibited or enhanced gastric cancer cell anoikis in vitro and peritoneal metastasis in vivo, respectively. Mechanically, GPER1 knockout disrupted the NADPH pool and increased reactive oxygen species (ROS) generation. Conversely, overexpression of GPER1 had the opposite effects. GPER1 suppressed nicotinamide adenine dinucleotide kinase 1(NADK1) ubiquitination and promoted its phosphorylation, which were responsible for the elevated expression of NADK1 at protein levels and activity, respectively. Moreover, genetic inhibition of NADK1 disrupted NADPH and redox homeostasis, leading to high levels of ROS and significant anoikis, which inhibited lung and peritoneal metastasis in cell-based xenograft models. In summary, our study suggests that inhibiting GPER1-mediated NADK1 activity and its ubiquitination may be a promising therapeutic strategy for peritoneal metastasis of gastric cancer.
Collapse
Affiliation(s)
- Teng Wu
- GMU-GIBH Joint School of Life Sciences, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, P.R. China
- Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, P.R. China
| | - Ke Ding
- GMU-GIBH Joint School of Life Sciences, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, P.R. China
| | - Chun Wang
- GMU-GIBH Joint School of Life Sciences, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, P.R. China
| | - Guoliang Lin
- GMU-GIBH Joint School of Life Sciences, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, P.R. China
| | - Chengjie Xie
- GMU-GIBH Joint School of Life Sciences, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, P.R. China
| | - Xianying Chen
- GMU-GIBH Joint School of Life Sciences, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, P.R. China
| | - Quanxin Li
- GMU-GIBH Joint School of Life Sciences, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, P.R. China
| | - Fenghai Yu
- GMU-GIBH Joint School of Life Sciences, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, P.R. China
| | - Yuling Mao
- Center for Reproductive Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, P.R. China
| | - Wei Hong
- GMU-GIBH Joint School of Life Sciences, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, P.R. China
| | - Lei Lu
- GMU-GIBH Joint School of Life Sciences, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, P.R. China
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Medical University, Guangzhou, P.R. China
| | - Shuai Li
- GMU-GIBH Joint School of Life Sciences, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, P.R. China
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Medical University, Guangzhou, P.R. China
| |
Collapse
|
26
|
Yao H, Hu Y, Tong H, Shi S. Dimethylglycine Alleviates Metabolic Dysfunction-Associated Fatty Liver Disease by Improving the Circulating Estrogen Level via Gut Staphylococcus. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:2708-2717. [PMID: 38131116 DOI: 10.1021/acs.jafc.3c07075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
Our previous study screened out dietary 0.1% dimethylglycine (DMG), which had beneficial effects on egg production and fat deposition in laying hens during the late laying period. In this paper, it was further found that dietary DMG alleviated fatty liver disease and enhanced lipid deposited into the yolk while promoting hepatic lipid transport. There are intestinal estrogen-metabolizing bacteria (EBM) having β-glucuronase (GUS) activity that regulates the content of circulating estrogen (E2) in mammals. There were 39 related bacteria found in laying hens, and DMG increased E2 in blood, Staphylococcus abundance among EBM and GUS activity in cecum chyme. Interfered in situ, Staphylococcus with GUS activity was proved the target bacteria for DMG. Furthermore, E2 could modify hepatic lipid deposition through promoting lipid transport by the steatosis LMH model. These perspectives confirm that DMG, mediated by Staphylococcus, alleviates the restriction of hepatic lipid transport due to reduced levels of E2 in laying hens.
Collapse
Affiliation(s)
- Hong Yao
- Jiangsu Institute of Poultry Science, Yangzhou, Jiangsu 225125, China
| | - Yan Hu
- Jiangsu Institute of Poultry Science, Yangzhou, Jiangsu 225125, China
| | - Haibing Tong
- Jiangsu Institute of Poultry Science, Yangzhou, Jiangsu 225125, China
| | - Shourong Shi
- Jiangsu Institute of Poultry Science, Yangzhou, Jiangsu 225125, China
| |
Collapse
|
27
|
Abbott DA, Mancini MG, Bolt MJ, Szafran AT, Neugebauer KA, Stossi F, Gorelick DA, Mancini MA. A novel ERβ high throughput microscopy platform for testing endocrine disrupting chemicals. Heliyon 2024; 10:e23119. [PMID: 38169792 PMCID: PMC10758781 DOI: 10.1016/j.heliyon.2023.e23119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 11/24/2023] [Accepted: 11/27/2023] [Indexed: 01/05/2024] Open
Abstract
In this study we present an inducible biosensor model for the Estrogen Receptor Beta (ERβ), GFP-ERβ:PRL-HeLa, a single-cell-based high throughput (HT) in vitro assay that allows direct visualization and measurement of GFP-tagged ERβ binding to ER-specific DNA response elements (EREs), ERβ-induced chromatin remodeling, and monitor transcriptional alterations via mRNA fluorescence in situ hybridization for a prolactin (PRL)-dsRED2 reporter gene. The model was used to accurately (Z' = 0.58-0.8) differentiate ERβ-selective ligands from ERα ligands when treated with a panel of selective agonists and antagonists. Next, we tested an Environmental Protection Agency (EPA)-provided set of 45 estrogenic reference chemicals with known ERα in vivo activity and identified several that activated ERβ as well, with varying sensitivity, including a subset that is completely novel. We then used an orthogonal ERE-containing transgenic zebrafish (ZF) model to cross validate ERβ and ERα selective activities at the organism level. Using this environmentally relevant ZF assay, some compounds were confirmed to have ERβ activity, validating the GFP-ERβ:PRL-HeLa assay as a screening tool for potential ERβ active endocrine disruptors (EDCs). These data demonstrate the value of sensitive multiplex mechanistic data gathered by the GFP-ERβ:PRL-HeLa assay coupled with an orthogonal zebrafish model to rapidly identify environmentally relevant ERβ EDCs and improve upon currently available screening tools for this understudied nuclear receptor.
Collapse
Affiliation(s)
- Derek A. Abbott
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Maureen G. Mancini
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- GCC Center for Advanced Microscopy and Image Informatics, Houston, TX, USA
| | - Michael J. Bolt
- GCC Center for Advanced Microscopy and Image Informatics, Houston, TX, USA
- Center for Translational Cancer Research, Institute of Biosciences & Technology, Texas A&M University, Houston, TX, USA
| | - Adam T. Szafran
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- GCC Center for Advanced Microscopy and Image Informatics, Houston, TX, USA
| | - Kaley A. Neugebauer
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA
| | - Fabio Stossi
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- GCC Center for Advanced Microscopy and Image Informatics, Houston, TX, USA
| | - Daniel A. Gorelick
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA
| | - Michael A. Mancini
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- GCC Center for Advanced Microscopy and Image Informatics, Houston, TX, USA
- Center for Translational Cancer Research, Institute of Biosciences & Technology, Texas A&M University, Houston, TX, USA
- Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
28
|
Lopes PKF, Costa SDO, Simino LADP, Chaves WF, Silva FA, Costa CL, Milanski M, Ignacio-Souza LM, Torsoni AS, Torsoni MA. Hypothalamic inflammation and the development of an obese phenotype induced by high-fat diet consumption is exacerbated in alpha7 nicotinic cholinergic receptor knockout mice. Food Res Int 2024; 176:113808. [PMID: 38163714 DOI: 10.1016/j.foodres.2023.113808] [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: 09/14/2023] [Revised: 11/27/2023] [Accepted: 12/02/2023] [Indexed: 01/03/2024]
Abstract
Hypothalamic inflammation and metabolic changes resulting from the consumption of high-fat diets have been linked to low grade inflammation and obesity. Inflammation impairs the hypothalamic expression of α7 nicotinic acetylcholine receptor (α7nAChR). The α7nAChR is described as the main component of the anti-inflammatory cholinergic pathway in different inflammation models. To assess whether the reduction in α7nAChR expression exacerbates hypothalamic inflammation induced by a high-fat diet (HFD), were used male and female global α7nAChR knockout mouse line in normal or high-fat diet for 4 weeks. Body weight gain, adiposity, glucose homeostasis, hypothalamic inflammation, food intake, and energy expenditure were evaluated. Insulin sensitivity was evaluated in neuronal cell culture. Consumption of an HFD for 4 weeks resulted in body weight gain and adiposity in male Chrna7-/- mice and the hypothalamus of male Chrna7-/- mice showed neuroinflammatory markers, with increased gene expression of pro-inflammatory cytokines and dysregulation in the nuclear factor kappa B pathway. Moreover, male Chrna7-/- mice consuming an HFD showed alterations in glucose homeostasis and serum of Chrna7-/- mice that consumed an HFD impaired insulin signalling in neuronal cell culture experiments. In general, female Chrna7-/- mice that consumed an HFD did not show the phenotypic and molecular changes found in male mice, indicating that there is sexual dimorphism in the analysed parameters. Thus, receptor deletion resulted in increased susceptibility to hypothalamic inflammation and metabolic damage associated with HFD consumption in male mice.
Collapse
Affiliation(s)
| | - Suleyma de Oliveira Costa
- Laboratory of Metabolic Disorders (Labdime), School of Applied Sciences, University of Campinas (UNICAMP), Brazil
| | - Laís A de Paula Simino
- Laboratory of Metabolic Disorders (Labdime), School of Applied Sciences, University of Campinas (UNICAMP), Brazil
| | - Wenicios Ferreira Chaves
- Laboratory of Metabolic Disorders (Labdime), School of Applied Sciences, University of Campinas (UNICAMP), Brazil
| | - Franciely Alves Silva
- Laboratory of Metabolic Disorders (Labdime), School of Applied Sciences, University of Campinas (UNICAMP), Brazil
| | - Caroline Lobo Costa
- Laboratory of Metabolic Disorders (Labdime), School of Applied Sciences, University of Campinas (UNICAMP), Brazil
| | - Marciane Milanski
- Laboratory of Metabolic Disorders (Labdime), School of Applied Sciences, University of Campinas (UNICAMP), Brazil; Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Brazil
| | - Leticia Martins Ignacio-Souza
- Laboratory of Metabolic Disorders (Labdime), School of Applied Sciences, University of Campinas (UNICAMP), Brazil; Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Brazil
| | - Adriana Souza Torsoni
- Laboratory of Metabolic Disorders (Labdime), School of Applied Sciences, University of Campinas (UNICAMP), Brazil; Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Brazil
| | - Marcio Alberto Torsoni
- Laboratory of Metabolic Disorders (Labdime), School of Applied Sciences, University of Campinas (UNICAMP), Brazil; Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Brazil.
| |
Collapse
|
29
|
Su Z, Yao B, Liu G, Fang J. Polyphenols as potential preventers of osteoporosis: A comprehensive review on antioxidant and anti-inflammatory effects, molecular mechanisms, and signal pathways in bone metabolism. J Nutr Biochem 2024; 123:109488. [PMID: 37865383 DOI: 10.1016/j.jnutbio.2023.109488] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 10/03/2023] [Accepted: 10/17/2023] [Indexed: 10/23/2023]
Abstract
Osteoporosis (OP) is a skeletal disorder characterized by decreased bone density, alterations in bone microstructure, and increased damage to the bones. As the population ages and life expectancy increases, OP has become a global epidemic, drawing attention from scientists and doctors. Because of polyphenols have favorable antioxidant and anti-allergy effects, which are regarded as potential methods to prevent angiocardipathy and OP. Polyphenols offer a promising approach to preventing and treating OP by affecting bone metabolism, reducing bone resolution, maintaining bone density, and lowering the differentiation level of osteoclasts (OC). There are multiple ways in which polyphenols affect bone metabolism. This article provides an overview of how polyphenols inhibit oxidative stress, exert antibacterial effects, and prevent the occurrence of OP. Furthermore, we will explore the regulatory mechanisms and signaling pathways implicated in this process.
Collapse
Affiliation(s)
- Zhan Su
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, Hunan, China
| | - Bin Yao
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, Hunan, China
| | - Gang Liu
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, Hunan, China
| | - Jun Fang
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, Hunan, China.
| |
Collapse
|
30
|
Reghupaty SC, Dall NR, Svensson KJ. Hallmarks of the metabolic secretome. Trends Endocrinol Metab 2024; 35:49-61. [PMID: 37845120 PMCID: PMC10841501 DOI: 10.1016/j.tem.2023.09.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 09/24/2023] [Accepted: 09/25/2023] [Indexed: 10/18/2023]
Abstract
The identification of novel secreted factors is advancing at an unprecedented pace. However, there is a critical need to consolidate and integrate this knowledge to provide a framework of their diverse mechanisms, functional significance, and inter-relationships. Complicating this effort are challenges related to nonstandardized methods, discrepancies in sample handling, and inconsistencies in the annotation of unknown molecules. This Review aims to synthesize the rapidly expanding field of the metabolic secretome, encompassing the five major types of secreted factors: proteins, peptides, metabolites, lipids, and extracellular vesicles. By systematically defining the functions and detection of the components within the metabolic secretome, this Review provides a primer into the advances of the field, and how integration of the techniques discussed can provide a deeper understanding of the mechanisms underlying metabolic homeostasis and its disorders.
Collapse
Affiliation(s)
- Saranya C Reghupaty
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA, USA; Stanford Cardiovascular Institute, Stanford University School of Medicine, CA, USA
| | - Nicholas R Dall
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA, USA; Stanford Cardiovascular Institute, Stanford University School of Medicine, CA, USA
| | - Katrin J Svensson
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA, USA; Stanford Cardiovascular Institute, Stanford University School of Medicine, CA, USA.
| |
Collapse
|
31
|
Yang C, Zhao J, Lin C, Gao Y, Luo J, He F, Fang Z, Li Z, Ran Q, Yang Z. Inhibition of integrin receptors reduces extracellular matrix levels, ameliorating benign prostate hyperplasia. Int J Biol Macromol 2023; 253:126499. [PMID: 37659484 DOI: 10.1016/j.ijbiomac.2023.126499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 07/26/2023] [Accepted: 08/22/2023] [Indexed: 09/04/2023]
Abstract
Although a high prevalence of benign prostate hyperplasia (BPH) has been documented, the risk factors are poorly understood. Metabolic syndrome increases the risk of BPH. Succinylation, a type of posttranslational modification, mostly targets metabolic processes. The level of succinylation was investigated in 4 BPH patients and 4 healthy controls. Additionally, 176 patients with BPH were analyzed by using pan-antisuccinyllysine antibody blotting. TMT-labeling proteomic and sc-RNAseq Cellchat analyses were employed to identify key signaling factors involved in the development of BPH. In vivo and in vitro experiments were used to confirm the role of integrin receptors. The global succinylation level in BPH was higher than that in the healthy prostate. Positive correlations of prostate volume with IHC score sand urodynamics testing were found in large clinical cohorts. The extracellular matrix (ECM), metabolic processes and immune signaling were involved in succinylation in BPH, as indicated by using TMT-labeling proteomic analysis, and this finding was also confirmed by sc-RNAseq CellChat analysis. The proteins upregulated in SIRT5 knockout WPMY-1 cells were also enriched in the extracellular matrix and metabolic processes. More importantly, integrin receptor inhibition in a mouse model of BPH significantly ameliorated prostate hyperplasia. High levels of succinylation modifications were found in BPH, and succinylated proteins influenced the activation of the ECM. Inhibition of ECM signaling further ameliorated prostate hyperplasia in mice.
Collapse
Affiliation(s)
- Chengfei Yang
- Department of Urology, Second Affiliated Hospital, Army Medical University, Chongqing, China; Department of Thoracic Surgery, Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Jiang Zhao
- Department of Urology, Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Chuanchuan Lin
- Department of Blood Transfusion, Irradiation Biology Laboratory, Army Medical University, Chongqing, China
| | - Ye Gao
- Department of Urology, Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Jing Luo
- Department of Urology, Second Affiliated Hospital, Army Medical University, Chongqing, China; Department of urology, General Hospital of Xinjiang Military Command, Urumqi, Xinjiang, PR China
| | - Fan He
- Department of Urology, Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Zhenqiang Fang
- Department of Urology, Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Zhongjun Li
- Department of Blood Transfusion, Irradiation Biology Laboratory, Army Medical University, Chongqing, China.
| | - Qian Ran
- Department of Blood Transfusion, Irradiation Biology Laboratory, Army Medical University, Chongqing, China.
| | - Zhenxing Yang
- Department of Blood Transfusion, Irradiation Biology Laboratory, Army Medical University, Chongqing, China; Department of Urology, Second Affiliated Hospital, Army Medical University, Chongqing, China.
| |
Collapse
|
32
|
Azizian H, Farhadi Z, Bader M, Alizadeh Ghalenoei J, Ghafari MA, Mahmoodzadeh S. GPER activation attenuates cardiac dysfunction by upregulating the SIRT1/3-AMPK-UCP2 pathway in postmenopausal diabetic rats. PLoS One 2023; 18:e0293630. [PMID: 38134189 PMCID: PMC10745199 DOI: 10.1371/journal.pone.0293630] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 10/16/2023] [Indexed: 12/24/2023] Open
Abstract
Postmenopausal diabetic women are at higher risk to develop cardiovascular diseases (CVD) compared with nondiabetic women. Alterations in cardiac cellular metabolism caused by changes in sirtuins are one of the main causes of CVD in postmenopausal diabetic women. Several studies have demonstrated the beneficial actions of the G protein-coupled estrogen receptor (GPER) in postmenopausal diabetic CVD. However, the molecular mechanisms by which GPER has a cardioprotective effect are still not well understood. In this study, we used an ovariectomized (OVX) type-two diabetic (T2D) rat model induced by high-fat diet/streptozotocin to investigate the effect of G-1 (GPER-agonist) on sirtuins, and their downstream pathways involved in regulation of cardiac metabolism and function. Animals were divided into five groups: Sham-Control, T2D, OVX+T2D, OVX+T2D+Vehicle, and OVX+T2D+G-1. G-1 was administrated for six weeks. At the end, hemodynamic factors were measured, and protein levels of sirtuins, AMP-activated protein kinase (AMPK), and uncoupling protein 2 (UCP2) were determined by Western blot analysis. In addition, cardiac levels of oxidative stress biomarkers were measured. The findings showed that T2D led to left ventricular dysfunction and signs of oxidative stress in the myocardium, which were accompanied by decreased protein levels of Sirt1/2/3/6, p-AMPK, and UCP2 in the heart. Moreover, the induction of the menopausal state exacerbated these changes. In contrast, treatment with G-1 ameliorated the hemodynamic changes associated with ovariectomy by increasing Sirt1/3, p-AMPK, UCP2, and improving oxidative status. The results provide evidence of the cardioprotective effects of GPER operating through Sirt1/3, p-AMPK, and UCP2, thereby improving cardiac function. Our results suggest that increasing Sirt1/3 levels may offer new therapeutic approaches for postmenopausal diabetic CVD.
Collapse
Affiliation(s)
- Hossein Azizian
- Yazd Neuroendocrine Research Center, School of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Zeinab Farhadi
- Yazd Neuroendocrine Research Center, School of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Michael Bader
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu Berlin, Berlin, Germany
- University of Lübeck, Institute for Biology, Lübeck, Germany
| | - Jalil Alizadeh Ghalenoei
- Yazd Neuroendocrine Research Center, School of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Mohammad Amin Ghafari
- Yazd Neuroendocrine Research Center, School of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Shokoufeh Mahmoodzadeh
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| |
Collapse
|
33
|
Umbayev B, Saliev T, Safarova (Yantsen) Y, Yermekova A, Olzhayev F, Bulanin D, Tsoy A, Askarova S. The Role of Cdc42 in the Insulin and Leptin Pathways Contributing to the Development of Age-Related Obesity. Nutrients 2023; 15:4964. [PMID: 38068822 PMCID: PMC10707920 DOI: 10.3390/nu15234964] [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: 10/25/2023] [Revised: 11/22/2023] [Accepted: 11/26/2023] [Indexed: 12/18/2023] Open
Abstract
Age-related obesity significantly increases the risk of chronic diseases such as type 2 diabetes, cardiovascular diseases, hypertension, and certain cancers. The insulin-leptin axis is crucial in understanding metabolic disturbances associated with age-related obesity. Rho GTPase Cdc42 is a member of the Rho family of GTPases that participates in many cellular processes including, but not limited to, regulation of actin cytoskeleton, vesicle trafficking, cell polarity, morphology, proliferation, motility, and migration. Cdc42 functions as an integral part of regulating insulin secretion and aging. Some novel roles for Cdc42 have also been recently identified in maintaining glucose metabolism, where Cdc42 is involved in controlling blood glucose levels in metabolically active tissues, including skeletal muscle, adipose tissue, pancreas, etc., which puts this protein in line with other critical regulators of glucose metabolism. Importantly, Cdc42 plays a vital role in cellular processes associated with the insulin and leptin signaling pathways, which are integral elements involved in obesity development if misregulated. Additionally, a change in Cdc42 activity may affect senescence, thus contributing to disorders associated with aging. This review explores the complex relationships among age-associated obesity, the insulin-leptin axis, and the Cdc42 signaling pathway. This article sheds light on the vast molecular web that supports metabolic dysregulation in aging people. In addition, it also discusses the potential therapeutic implications of the Cdc42 pathway to mitigate obesity since some new data suggest that inhibition of Cdc42 using antidiabetic drugs or antioxidants may promote weight loss in overweight or obese patients.
Collapse
Affiliation(s)
- Bauyrzhan Umbayev
- National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan; (Y.S.); (A.Y.); (F.O.); (A.T.); (S.A.)
| | - Timur Saliev
- S.D. Asfendiyarov Kazakh National Medical University, Almaty 050012, Kazakhstan;
| | - Yuliya Safarova (Yantsen)
- National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan; (Y.S.); (A.Y.); (F.O.); (A.T.); (S.A.)
| | - Aislu Yermekova
- National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan; (Y.S.); (A.Y.); (F.O.); (A.T.); (S.A.)
| | - Farkhad Olzhayev
- National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan; (Y.S.); (A.Y.); (F.O.); (A.T.); (S.A.)
| | - Denis Bulanin
- Department of Biomedical Sciences, School of Medicine, Nazarbayev University, Astana 010000, Kazakhstan;
| | - Andrey Tsoy
- National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan; (Y.S.); (A.Y.); (F.O.); (A.T.); (S.A.)
| | - Sholpan Askarova
- National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan; (Y.S.); (A.Y.); (F.O.); (A.T.); (S.A.)
| |
Collapse
|
34
|
Li J, Xu A, Xue J, Qian W, Xu P, Hu Z, Chen C, Wu C. Development and validation of a deep eutectic solvent-assisted liquid-liquid extraction method for simultaneous quantification of six steroid hormones in serum by liquid chromatography-tandem mass spectrometry. J Chromatogr A 2023; 1710:464413. [PMID: 37806044 DOI: 10.1016/j.chroma.2023.464413] [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: 07/05/2023] [Revised: 09/13/2023] [Accepted: 09/24/2023] [Indexed: 10/10/2023]
Abstract
Steroid hormones have been reported to be associated with endocrine system diseases. This paper proposes a novel procedure of deep eutectic solvent (DES)-assisted liquid-liquid extraction (LLE) to extract six steroid hormones (including cortisone, cortisol, androstenedione, testosterone, 17-hydroxyprogesterone, and progesterone) from serum coupled with liquid chromatography-tandem mass spectrometry (LC-MS/MS). A total of five types of L-proline, choline chloride, and citric acid-based DESs were tailored; the DES from L-proline and ethylene glycol at a molar ratio of 1:4 with 20 % acetonitrile was selected as the best-fit assisted solvent for the six steroid hormones compared with other DESs. The parameters for extraction by selected DES were optimized using Box-Behnken design (BBD), and the optimal extraction conditions are 200 µL of acetonitrile, 100 µL of the sample, and 80 µL of DES. Under optimum conditions, the method has good linear calibration ranges (between 0.07 ng mL-1 and 600 ng mL-1), correlation coefficients of determination (r2>0.99), and low limits of quantification (between 0.02 and 0.60 ng mL-1). The extraction recoveries were in the range of 81.84-114.43 %, and the intra-day and inter-day relative standard deviations (RSDs) were less than 10 %.In general, the DES-LC-MS/MS method is a simple and environmentally-friendly method, which can be complementary to the presently available methods for determining steroid hormones in serum.
Collapse
Affiliation(s)
- Jufang Li
- Zhejiang Biozon Medical Technology Co., Ltd, Hangzhou, Zhejiang 310000, China
| | - Aiping Xu
- Laprode (Zhejiang) Analysis Co., Ltd, Hangzhou, Zhejiang 310000, China
| | - Jianyou Xue
- Zhejiang Biozon Medical Technology Co., Ltd, Hangzhou, Zhejiang 310000, China
| | - Wei Qian
- Zhejiang Biozon Medical Technology Co., Ltd, Hangzhou, Zhejiang 310000, China
| | - Ping Xu
- Zhejiang Biozon Medical Technology Co., Ltd, Hangzhou, Zhejiang 310000, China
| | - Zhineng Hu
- Zhejiang Biozon Medical Technology Co., Ltd, Hangzhou, Zhejiang 310000, China
| | - Chen Chen
- Department of Clinical Laboratory, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu 210004, China.
| | - Chaochao Wu
- Zhejiang Biozon Medical Technology Co., Ltd, Hangzhou, Zhejiang 310000, China.
| |
Collapse
|
35
|
Wang Y, Yu T, Chen J, Zhao R, Diao M, Mei P, He S, Qiu W, Ye G, Jiang L, Xiao H, Liao Y. Immune characteristics analysis and construction of a four-gene prognostic signature for lung adenocarcinoma based on estrogen reactivity. BMC Cancer 2023; 23:1047. [PMID: 37907850 PMCID: PMC10619241 DOI: 10.1186/s12885-023-11415-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 09/17/2023] [Indexed: 11/02/2023] Open
Abstract
Lung adenocarcinoma (LUAD) is a common type of malignant tumor with poor prognosis and high mortality. In our previous studies, we found that estrogen is an important risk factor for LUAD, and different estrogen statuses can predict different prognoses. Therefore, in this study, we constructed a prognostic signature related to estrogen reactivity to determine the relationship between different estrogen reactivities and prognosis. We downloaded the LUAD dataset from The Cancer Genome Atlas (TCGA) database, calculated the estrogen reactivity of each sample, and divided them into a high-estrogen reactivity group and a low-estrogen reactivity group. The difference in overall survival between the groups was significant. We also analyzed the status of immune cell infiltration and immune checkpoint expression between the groups. We analyzed the differential gene expression between the groups and screened four key prognostic factors by the least absolute shrinkage and selection operator (LASSO) regression and univariable and multivariable Cox regression. Based on the four genes, a risk signature was established. To a certain extent, the receiver operating characteristic (ROC) curve showed the predictive ability of the risk signature, which was further verified using the GSE31210 dataset. We also determined the role of estrogen in LUAD using an orthotopic mouse model. Additionally, we developed a predictive nomogram combining the risk signature with other clinical characteristics. In conclusion, our four-gene prognostic signature based on estrogen reactivity had prognostic value and can provide new insights into the development of treatment strategies for LUAD.
Collapse
Affiliation(s)
- Yangwei Wang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tong Yu
- Department of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiaping Chen
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rong Zhao
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mingxin Diao
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Peiyuan Mei
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shiwen He
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenlin Qiu
- Department of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guanchao Ye
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lijuan Jiang
- Department of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Han Xiao
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Yongde Liao
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
36
|
Sommer B, González-Ávila G, Flores-Soto E, Montaño LM, Solís-Chagoyán H, Romero-Martínez BS. Phytoestrogen-Based Hormonal Replacement Therapy Could Benefit Women Suffering Late-Onset Asthma. Int J Mol Sci 2023; 24:15335. [PMID: 37895016 PMCID: PMC10607548 DOI: 10.3390/ijms242015335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/10/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
It has been observed that plasmatic concentrations of estrogens, progesterone, or both correlate with symptoms in asthmatic women. Fluctuations in female sex steroid concentrations during menstrual periods are closely related to asthma symptoms, while menopause induces severe physiological changes that might require hormonal replacement therapy (HRT), that could influence asthma symptoms in these women. Late-onset asthma (LOA) has been categorized as a specific asthmatic phenotype that includes menopausal women and novel research regarding therapeutic alternatives that might provide relief to asthmatic women suffering LOA warrants more thorough and comprehensive analysis. Therefore, the present review proposes phytoestrogens as a promising HRT that might provide these females with relief for both their menopause and asthma symptoms. Besides their well-recognized anti-inflammatory and antioxidant capacities, phytoestrogens activate estrogen receptors and promote mild hormone-like responses that benefit postmenopausal women, particularly asthmatics, constituting therefore a very attractive potential therapy largely due to their low toxicity and scarce side effects.
Collapse
Affiliation(s)
- Bettina Sommer
- Departamento de Investigación en Hiperreactividad Bronquial, Instituto Nacional de Enfermedades Respiratorias ‘Ismael Cosio Villegas’, Calzada de Tlalpan 4502, Colonia Sección XVI, Mexico City CP 14080, Mexico
| | - Georgina González-Ávila
- Laboratorio de Oncología Biomédica, Departamento de Enfermedades Crónico Degenerativas, Instituto Nacional de Enfermedades Respiratorias ‘Ismael Cosio Villegas’, Mexico City CP 14080, Mexico;
| | - Edgar Flores-Soto
- Departmento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City CP 04510, Mexico; (E.F.-S.); (L.M.M.); (B.S.R.-M.)
| | - Luis M. Montaño
- Departmento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City CP 04510, Mexico; (E.F.-S.); (L.M.M.); (B.S.R.-M.)
| | - Héctor Solís-Chagoyán
- Neurociencia Cognitiva Evolutiva, Centro de Investigación en Ciencias Cognitivas, Universidad Autónoma del Estado de Morelos, Cuernavaca CP 62209, Morelos, Mexico;
| | - Bianca S. Romero-Martínez
- Departmento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City CP 04510, Mexico; (E.F.-S.); (L.M.M.); (B.S.R.-M.)
| |
Collapse
|
37
|
Miziak P, Baran M, Błaszczak E, Przybyszewska-Podstawka A, Kałafut J, Smok-Kalwat J, Dmoszyńska-Graniczka M, Kiełbus M, Stepulak A. Estrogen Receptor Signaling in Breast Cancer. Cancers (Basel) 2023; 15:4689. [PMID: 37835383 PMCID: PMC10572081 DOI: 10.3390/cancers15194689] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 09/19/2023] [Accepted: 09/20/2023] [Indexed: 10/15/2023] Open
Abstract
Estrogen receptor (ER) signaling is a critical regulator of cell proliferation, differentiation, and survival in breast cancer (BC) and other hormone-sensitive cancers. In this review, we explore the mechanism of ER-dependent downstream signaling in BC and the role of estrogens as growth factors necessary for cancer invasion and dissemination. The significance of the clinical implications of ER signaling in BC, including the potential of endocrine therapies that target estrogens' synthesis and ER-dependent signal transmission, such as aromatase inhibitors or selective estrogen receptor modulators, is discussed. As a consequence, the challenges associated with the resistance to these therapies resulting from acquired ER mutations and potential strategies to overcome them are the critical point for the new treatment strategies' development.
Collapse
Affiliation(s)
- Paulina Miziak
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 1 Chodzki Street, 20-093 Lublin, Poland; (M.B.); (E.B.); (A.P.-P.); (J.K.); (M.D.-G.)
| | - Marzena Baran
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 1 Chodzki Street, 20-093 Lublin, Poland; (M.B.); (E.B.); (A.P.-P.); (J.K.); (M.D.-G.)
| | - Ewa Błaszczak
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 1 Chodzki Street, 20-093 Lublin, Poland; (M.B.); (E.B.); (A.P.-P.); (J.K.); (M.D.-G.)
| | - Alicja Przybyszewska-Podstawka
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 1 Chodzki Street, 20-093 Lublin, Poland; (M.B.); (E.B.); (A.P.-P.); (J.K.); (M.D.-G.)
| | - Joanna Kałafut
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 1 Chodzki Street, 20-093 Lublin, Poland; (M.B.); (E.B.); (A.P.-P.); (J.K.); (M.D.-G.)
| | - Jolanta Smok-Kalwat
- Department of Clinical Oncology, Holy Cross Cancer Centre, 3 Artwinskiego Street, 25-734 Kielce, Poland;
| | - Magdalena Dmoszyńska-Graniczka
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 1 Chodzki Street, 20-093 Lublin, Poland; (M.B.); (E.B.); (A.P.-P.); (J.K.); (M.D.-G.)
| | - Michał Kiełbus
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 1 Chodzki Street, 20-093 Lublin, Poland; (M.B.); (E.B.); (A.P.-P.); (J.K.); (M.D.-G.)
| | - Andrzej Stepulak
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 1 Chodzki Street, 20-093 Lublin, Poland; (M.B.); (E.B.); (A.P.-P.); (J.K.); (M.D.-G.)
| |
Collapse
|
38
|
Chen X, Bing J, Lu S, Lin S, Li H, Du S, Liu J, Xi C, Zhang X, Zeng S. Notch1 is involved in cell proliferation and neuronal differentiation in the HVC of zebra finch (Taeniopygia guttata). Behav Brain Res 2023; 452:114564. [PMID: 37459956 DOI: 10.1016/j.bbr.2023.114564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 06/10/2023] [Accepted: 07/01/2023] [Indexed: 07/26/2023]
Abstract
Significant sex differences are found in songbirds' song control nuclei and their controlled song behaviors. To elucidate the underlying mechanisms, we explored the role of Notch1 during the development of the high vocal centre (HVC) and song learning in zebra finch. Our study first found that Notch1 positive cells were distributed in HVC with female-biased densities at posthatching day (PHD) 15, but male-biased at PHD 45 and adult. There were about 60 putative oestrogen-responsive elements within 2.5 kb upstream of Notch1, and Notch1 mRNA in the explants that contained the developing male HVC was significantly increased after estrogen addition into the cultured medium for 48 h. After injecting Notch1-interfering lentivirus into the male or female HVC at PHD 15, cell proliferation was significantly promoted in the ventricle zone overlying the HVC at PHD 23. In addition, neuronal differentiation towards Hu+ /BrdU+ at PHD 31, mature neurons (NeuN+/BrdU+) including those projecting to RA in HVC and the sizes of HVC and RA at adult increased significantly after Notch1-interfering lentiviruses were injected into the male HVC at PHD 15. However, the above measurements decreased, following the injection of the lentiviruses expressing Notch intracellular domain (NICD). Finally, the repeat numbers of syllables 'b' or 'c' of learned songs changed after the injection of Notch1-interfering or NICD-expressing lentiviruses into the HVC at PHD15. Our study suggests that Notch1 is related to the development of HVC and song learning in the zebra finch.
Collapse
Affiliation(s)
- Xiaoning Chen
- Beijing Key Laboratory of Gene Resource and Molecular Development, Beijing Normal University, Beijing 100875, China
| | - Jie Bing
- Beijing Key Laboratory of Gene Resource and Molecular Development, Beijing Normal University, Beijing 100875, China
| | - Shan Lu
- Beijing Key Laboratory of Gene Resource and Molecular Development, Beijing Normal University, Beijing 100875, China
| | - Shiying Lin
- Beijing Key Laboratory of Gene Resource and Molecular Development, Beijing Normal University, Beijing 100875, China
| | - Hongyang Li
- Beijing Key Laboratory of Gene Resource and Molecular Development, Beijing Normal University, Beijing 100875, China
| | - Sanyan Du
- Beijing Key Laboratory of Gene Resource and Molecular Development, Beijing Normal University, Beijing 100875, China
| | - Jin Liu
- Beijing Key Laboratory of Gene Resource and Molecular Development, Beijing Normal University, Beijing 100875, China
| | - Chao Xi
- Beijing Key Laboratory of Gene Resource and Molecular Development, Beijing Normal University, Beijing 100875, China
| | - Xinwen Zhang
- Hainan Instistute of Science and Technology, Haikou 571126, China; College of Life Sciences, Hainan Normal University, Haikou 571158, China.
| | - Shaoju Zeng
- Beijing Key Laboratory of Gene Resource and Molecular Development, Beijing Normal University, Beijing 100875, China.
| |
Collapse
|
39
|
Zhao Z, Yang LL, Wang QL, Du JF, Zheng ZG, Jiang Y, Li P, Li HJ. Baohuoside I inhibits FXR signaling pathway to interfere with bile acid homeostasis via targeting ER α degradation. Cell Biol Toxicol 2023; 39:1215-1235. [PMID: 35802278 DOI: 10.1007/s10565-022-09737-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 06/20/2022] [Indexed: 11/30/2022]
Abstract
Epimedii folium (EF) is an effective herbal medicine in osteoporosis treatment, but the clinical utilization of EF has been limited due to potential hepatotoxicity. The previous studies identified that baohuoside I (BI), the main active component of EF, was relevant to EF-induced liver injury. However, the mechanisms of BI causing direct injury to hepatocytes remain unclear. Here, we reveal that BI inhibits FXR-mediated signaling pathway via targeting estrogen receptor α (ER α), leading to the accumulation of bile acids (BAs). Targeted bile acid analyses show BI alters the BA composition and distribution, resulting in impaired BA homeostasis. Mechanistically, BI induces FXR-dependent hepatotoxicity at transcriptional level. Additionally, ER α is predicted to bind to the FXR promoter region based on transcription factor binding sites databases and we further demonstrate that ER α positively regulates FXR promoter activity and affects the expression of target genes involved in BA metabolism. Importantly, we discover that ER α and its mediated FXR transcription regulation might be involved in BI-induced liver injury via ligand-dependent ER α degradation. Collectively, our findings indicate that FXR is a newly discovered target gene of ER α mediated BI-induced liver injury, and suggest BI may be responsible for EF-induced liver injury.
Collapse
Affiliation(s)
- Zhen Zhao
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, 210009, China
| | - Lu-Lu Yang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, 210009, China
| | - Qiao-Lei Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, 210009, China
| | - Jin-Fa Du
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, 210009, China
| | - Zu-Guo Zheng
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, 210009, China
| | - Yan Jiang
- College of Chemical Engineering, Nanjing Forestry University, 159 Longpan Road, Nanjing, 210037, China.
| | - Ping Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, 210009, China
| | - Hui-Jun Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, 210009, China.
| |
Collapse
|
40
|
Wei F, Gu Y, He L, Kapoor A, Lin X, Dong Y, Su Y, Neira SV, Tang D. HSD17B6 delays type 2 diabetes development via inhibiting SREBP activation. Metabolism 2023:155631. [PMID: 37330135 DOI: 10.1016/j.metabol.2023.155631] [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: 02/26/2023] [Revised: 06/09/2023] [Accepted: 06/11/2023] [Indexed: 06/19/2023]
Abstract
BACKGROUND The SREBP/SCAP/INSIG complex plays an essential role in SREBP activation and de novo lipogenesis. Whether the activation process is affected by hydroxysteroid 17-beta dehydrogenase 6 (HSD17B6) remains unknown. METHODS SREBP's transcriptional activities were analyzed using an SRE-luciferase (SRE-luc) reporter in 293T cells, Huh7 hepatoma cells, and primary human hepatocytes following a variety of conditions, including ectopic expression of HSD17B6, HSD17B6 mutants defective in its enzymatic activities, knockdown of HSD17B6, and cholesterol starvation. The interaction between HSD17B6 and SREBP/SCAP/INSIG complex was analyzed in 293T cells, Huh7 cells and mouse liver upon ectopic expression of HSD17B6 and its mutants; the interaction was also analyzed using endogenous proteins. The impacts of HSD17B6 on SREBP target expression, glucose tolerance, diet-induced obesity, and type 2 diabetes (T2D) were examined using Huh7 cells in vitro, and with C57BL/6 and NONcNZO10/LtJ T2D mice in vivo. RESULTS HSD17B6 binds to the SREBP/SCAP/INSIG complex and inhibits SREBP signaling in cultured hepatocytes and mouse liver. Although HSD17B6 plays a role in maintaining the equilibrium of 5α-dihydrotestosterone (DHT) in the prostate, a mutant defective in androgen metabolism was as effective as HSD17B6 in inhibiting SREBP signaling. Hepatic expression of both HSD17B6 and the defective mutant improved glucose intolerance and reduced hepatic triglyceride content in diet-induced obese C57BL/6 mice, while hepatic knockdown of HSD17B6 exacerbated glucose intolerance. Consistent with these results, liver-specific expression of HSD17B6 in a polygenic NONcNZO10/LtJ T2D mice reduced T2D development. CONCLUSIONS Our study unveils a novel role of HSD17B6 in inhibiting SREBP maturation via binding to the SREBP/SCAP/INSIG complex; this activity is independent of HSD17B6's sterol oxidase activity. Through this action, HSD17B6 improves glucose tolerance and attenuates the development of obesity-induced T2D. These findings position HSD17B6 as a potential therapeutic target for T2D therapy.
Collapse
Affiliation(s)
- Fengxiang Wei
- The Genetics Laboratory, Longgang District Maternity and Child Healthcare Hospital of Shenzhen City, Longgang District, Shenzhen, Guangdong, China; Urological Cancer Center for Research and Innovation (UCCRI), St Joseph's Hospital, Hamilton, ON L8N 4A6, Canada; The Research Institute of St Joe's Hamilton, St Joseph's Hospital, Hamilton, ON L8N 4A6, Canada; Department of Surgery, McMaster University, Hamilton, ON L8S 4K1, Canada.
| | - Yan Gu
- Urological Cancer Center for Research and Innovation (UCCRI), St Joseph's Hospital, Hamilton, ON L8N 4A6, Canada; The Research Institute of St Joe's Hamilton, St Joseph's Hospital, Hamilton, ON L8N 4A6, Canada; Department of Surgery, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Lizhi He
- Urological Cancer Center for Research and Innovation (UCCRI), St Joseph's Hospital, Hamilton, ON L8N 4A6, Canada; The Research Institute of St Joe's Hamilton, St Joseph's Hospital, Hamilton, ON L8N 4A6, Canada; Department of Surgery, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Anil Kapoor
- Urological Cancer Center for Research and Innovation (UCCRI), St Joseph's Hospital, Hamilton, ON L8N 4A6, Canada; The Research Institute of St Joe's Hamilton, St Joseph's Hospital, Hamilton, ON L8N 4A6, Canada; Department of Surgery, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Xiaozeng Lin
- Urological Cancer Center for Research and Innovation (UCCRI), St Joseph's Hospital, Hamilton, ON L8N 4A6, Canada; The Research Institute of St Joe's Hamilton, St Joseph's Hospital, Hamilton, ON L8N 4A6, Canada; Department of Surgery, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Ying Dong
- Urological Cancer Center for Research and Innovation (UCCRI), St Joseph's Hospital, Hamilton, ON L8N 4A6, Canada; The Research Institute of St Joe's Hamilton, St Joseph's Hospital, Hamilton, ON L8N 4A6, Canada; Department of Surgery, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Yingying Su
- Urological Cancer Center for Research and Innovation (UCCRI), St Joseph's Hospital, Hamilton, ON L8N 4A6, Canada; The Research Institute of St Joe's Hamilton, St Joseph's Hospital, Hamilton, ON L8N 4A6, Canada; Department of Surgery, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Sandra Vega Neira
- Urological Cancer Center for Research and Innovation (UCCRI), St Joseph's Hospital, Hamilton, ON L8N 4A6, Canada; The Research Institute of St Joe's Hamilton, St Joseph's Hospital, Hamilton, ON L8N 4A6, Canada; Department of Surgery, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Damu Tang
- Urological Cancer Center for Research and Innovation (UCCRI), St Joseph's Hospital, Hamilton, ON L8N 4A6, Canada; The Research Institute of St Joe's Hamilton, St Joseph's Hospital, Hamilton, ON L8N 4A6, Canada; Department of Surgery, McMaster University, Hamilton, ON L8S 4K1, Canada.
| |
Collapse
|
41
|
Abstract
Although sex differences have been noted in cellular function and behavior, therapy efficacy, and disease incidence and outcomes, the adoption of sex as a biological variable in tissue engineering and regenerative medicine remains limited. Furthering the development of personalized, precision medicine requires considering biological sex at the bench and in the clinic. This review provides the basis for considering biological sex when designing tissue-engineered constructs and regenerative therapies by contextualizing sex as a biological variable within the tissue engineering triad of cells, matrices, and signals. To achieve equity in biological sex within medicine requires a cultural shift in science and engineering research, with active engagement by researchers, clinicians, companies, policymakers, and funding agencies.
Collapse
Affiliation(s)
- Josephine B Allen
- Department of Materials Science and Engineering, University of Florida, Gainesville, Florida, USA;
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, USA;
| | - Christopher Ludtka
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, USA;
| | - Bryan D James
- Department of Marine Chemistry and Geochemistry, Woods Hole Oceanographic Institution, Woods Hole, Massachusetts, USA;
- Department of Biology, Woods Hole Oceanographic Institution, Woods Hole, Massachusetts, USA
| |
Collapse
|
42
|
Isola JVV, Ko S, Ocañas SR, Stout MB. Role of Estrogen Receptor α in Aging and Chronic Disease. ADVANCES IN GERIATRIC MEDICINE AND RESEARCH 2023; 5:e230005. [PMID: 37425648 PMCID: PMC10327608 DOI: 10.20900/agmr20230005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Estrogen receptor alpha (ERα) plays a crucial role in reproductive function in both sexes. It also mediates cellular responses to estrogens in multiple nonreproductive organ systems, many of which regulate systemic metabolic homeostasis and inflammatory processes in mammals. The loss of estrogens and/or ERα agonism during aging is associated with the emergence of several comorbid conditions, particularly in females undergoing the menopausal transition. Emerging data also suggests that male mammals likely benefit from ERα agonism if done in a way that circumvents feminizing characteristics. This has led us, and others, to speculate that tissue-specific ERα agonism may hold therapeutic potential for curtailing aging and chronic disease burden in males and females that are at high-risk of cancer and/or cardiovascular events with traditional estrogen replacement therapies. In this mini-review, we emphasize the role of ERα in the brain and liver, summarizing recent evidence that indicates these two organs systems mediate the beneficial effects of estrogens on metabolism and inflammation during aging. We also discuss how 17α-estradiol administration elicits health benefits in an ERα-dependent manner, which provides proof-of-concept that ERα may be a druggable target for attenuating aging and age-related disease burden.
Collapse
Affiliation(s)
- José V. V. Isola
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Sunghwan Ko
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Sarah R. Ocañas
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK 73104, USA
| | - Michael B. Stout
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
43
|
Zhuang Y, Sun X, Deng S, Wen Y, Xu Q, Guan Q. In vivo effects of low dose prenatal bisphenol A exposure on adiposity in male and female ICR offspring. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 257:114946. [PMID: 37105096 DOI: 10.1016/j.ecoenv.2023.114946] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 04/10/2023] [Accepted: 04/21/2023] [Indexed: 05/08/2023]
Abstract
BACKGROUND Bisphenol A (BPA) is known to exhibit endocrine disrupting activities and is associated with adiposity. We examined the obesogenic effect of prenatal BPA exposure in the present study. METHODS Pregnant ICR mice were exposed to vehicle or BPA via the drinking water at a dose of 0.5 μg/kg·d throughout the gestation. Obesity-related indexes were investigated in the 12-wk-old offspring. Primary mouse embryonic fibroblasts (MEFs) collected from treated embryos were used to test effects of BPA on adipocyte differentiation. RESULTS Offspring presented a significantly higher rate of weight gain than the control, with impaired insulin sensitivity and increased adipocyte size. Differentiation of MEFs from BPA-treated mice showed a higher propensity for the adipocyte commitment as well as up-regulation of genes enriched in lipid biosynthesis. TGF-β signaling pathway was found to modulate obesogenic effect of BPA in MEF model, but estrogen signaling pathway had no effect. CONCLUSIONS The present study provides strong evidence of the association between prenatal exposure to low dose of BPA and a significant increase in body weight in the offspring mice with a critical role played by TGF-β signaling pathway. The potential interactions modulating the binding of BPA and TGF-β that activate its obesogenic effects need to be examined.
Collapse
Affiliation(s)
- Yin Zhuang
- 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
| | - Xiangying Sun
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Siting Deng
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China; State Key Laboratory of Reproductive Medicine and Offspring Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Ya Wen
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China; State Key Laboratory of Reproductive Medicine and Offspring Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Qiujin Xu
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environment Science, Beijing 100012, China.
| | - Quanquan Guan
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China; State Key Laboratory of Reproductive Medicine and Offspring Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China.
| |
Collapse
|
44
|
Méndez-Álvarez D, Torres-Rojas MF, Lara-Ramirez EE, Marchat LA, Rivera G. Ligand-Based Virtual Screening, Molecular Docking, and Molecular Dynamic Simulations of New β-Estrogen Receptor Activators with Potential for Pharmacological Obesity Treatment. Molecules 2023; 28:molecules28114389. [PMID: 37298864 DOI: 10.3390/molecules28114389] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 05/24/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023] Open
Abstract
Obesity is a pandemic and a serious health problem in developed and undeveloped countries. Activation of estrogen receptor beta (ERβ) has been shown to promote weight loss without modifying caloric intake, making it an attractive target for developing new drugs against obesity. This work aimed to predict new small molecules as potential ERβ activators. A ligand-based virtual screening of the ZINC15, PubChem, and Molport databases by substructure and similarity was carried out using the three-dimensional organization of known ligands as a reference. A molecular docking screening of FDA-approved drugs was also conducted as a repositioning strategy. Finally, selected compounds were evaluated by molecular dynamic simulations. Compounds 1 (-24.27 ± 0.34 kcal/mol), 2 (-23.33 ± 0.3 kcal/mol), and 6 (-29.55 ± 0.51 kcal/mol) showed the best stability on the active site in complex with ERβ with an RMSD < 3.3 Å. RMSF analysis showed that these compounds do not affect the fluctuation of the Cα of ERβ nor the compactness according to the radius of gyration. Finally, an in silico evaluation of ADMET showed they are safe molecules. These results suggest that new ERβ ligands could be promising molecules for obesity control.
Collapse
Affiliation(s)
- Domingo Méndez-Álvarez
- Laboratorio de Biotecnología Farmacéutica, Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Reynosa 88710, Mexico
| | - Maria F Torres-Rojas
- Laboratorio de Biomedicina Molecular 2, Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, Ciudad de México 07320, Mexico
| | - Edgar E Lara-Ramirez
- Laboratorio de Biotecnología Farmacéutica, Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Reynosa 88710, Mexico
| | - Laurence A Marchat
- Laboratorio de Biomedicina Molecular 2, Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, Ciudad de México 07320, Mexico
| | - Gildardo Rivera
- Laboratorio de Biotecnología Farmacéutica, Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Reynosa 88710, Mexico
| |
Collapse
|
45
|
Dou J, Thangaraj SV, Puttabyatappa M, Elangovan VR, Bakulski K, Padmanabhan V. Developmental programming: Adipose depot-specific regulation of non-coding RNAs and their relation to coding RNA expression in prenatal testosterone and prenatal bisphenol-A -treated female sheep. Mol Cell Endocrinol 2023; 564:111868. [PMID: 36708980 PMCID: PMC10069610 DOI: 10.1016/j.mce.2023.111868] [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: 12/14/2022] [Revised: 01/19/2023] [Accepted: 01/22/2023] [Indexed: 01/27/2023]
Abstract
Inappropriate developmental exposure to steroids is linked to metabolic disorders. Prenatal testosterone excess or bisphenol A (BPA, an environmental estrogen mimic) leads to insulin resistance and adipocyte disruptions in female lambs. Adipocytes are key regulators of insulin sensitivity. Metabolic tissue-specific differences in insulin sensitivity coupled with adipose depot-specific changes in key mRNAs, were previously observed with prenatal steroid exposure. We hypothesized that depot-specific changes in the non-coding RNA (ncRNA) - regulators of gene expression would account for the direction of changes seen in mRNAs. Non-coding RNA (lncRNA, miRNA, snoRNA, snRNA) from various adipose depots of prenatal testosterone and BPA-treated animals were sequenced. Adipose depot-specific changes in the ncRNA that are consistent with the depot-specific mRNA expression in terms of directionality of changes and functional implications in insulin resistance, adipocyte differentiation and cardiac hypertrophy were found. Importantly, the adipose depot-specific ncRNA changes were model-specific and mutually exclusive, suggestive of different regulatory entry points in this regulation.
Collapse
Affiliation(s)
- John Dou
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | | | | | | | - Kelly Bakulski
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA.
| | | |
Collapse
|
46
|
MicroRNA and mRNA sequencing analyses reveal key hepatic metabolic and signaling pathways responsive to maternal undernutrition in full-term fetal pigs. J Nutr Biochem 2023; 116:109312. [PMID: 36871838 DOI: 10.1016/j.jnutbio.2023.109312] [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: 07/21/2022] [Revised: 01/03/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023]
Abstract
Maternal undernutrition is highly prevalent in developing countries, leading to severe fetus/infant mortality, intrauterine growth restriction, stunting, and severe wasting. However, the potential impairments of maternal undernutrition to metabolic pathways in offspring are not defined completely. In this study, two groups of pregnant domestic pigs received nutritionally balanced gestation diets with or without 50% feed intake restriction from 0 to 35 gestation days and 70% from 35 to 114 gestation days. Full-term fetuses were collected via C-section on day 113/114 of gestation. MicroRNA and mRNA deep sequencing were analyzed using the Illumina GAIIx system on fetal liver samples. The mRNA-miRNA correlation and associated signaling pathways were analyzed via CLC Genomics Workbench and Ingenuity Pathway Analysis Software. A total of 1189 and 34 differentially expressed mRNA and miRNAs were identified between full-nutrition (F) and restricted-nutrition (R) groups. The correlation analyses showed that metabolic and signaling pathways such as oxidative phosphorylation, death receptor signaling, neuroinflammation signaling pathway, and estrogen receptor signaling pathways were significantly modified, and the gene modifications in these pathways were associated with the miRNA changes induced by the maternal undernutrition. For example, the upregulated (p < 0.05) oxidative phosphorylation pathway in R group was validated using RT-qPCR, and the correlational analysis indicated that miR-221, 103, 107, 184, and 4497 correlate with their target genes NDUFA1, NDUFA11, NDUFB10 and NDUFS7 in this pathway. These results provide the framework for further understanding maternal malnutrition's negative impacts on hepatic metabolic pathways via miRNA-mRNA interactions in full-term fetal pigs.
Collapse
|
47
|
Braxas H, Musazadeh V, Zarezadeh M, Ostadrahimi A. Genistein effectiveness in improvement of glucose and lipid metabolism and homocysteine levels: A systematic review and meta-analysis. J Funct Foods 2023. [DOI: 10.1016/j.jff.2023.105433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023] Open
|
48
|
Hu Z, Li C, Wu T, Zhou J, Han L, Liu J, Qiang S, Zhao W, Li X, Liu X, Li J, Chen X. Sulfathiazole treats type 2 diabetes by restoring metabolism through activating CYP19A1. Biochim Biophys Acta Gen Subj 2023; 1867:130303. [PMID: 36627088 DOI: 10.1016/j.bbagen.2023.130303] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 12/24/2022] [Accepted: 01/03/2023] [Indexed: 01/09/2023]
Abstract
Globally, diabetes mellitus has been a major epidemic bringing metabolic and endocrine disorders. Currently, 1 in 11 adults suffers from diabetes mellitus, among the patients >90% contract type 2 diabetes mellitus (T2DM). Therefore, it is urgent to develop new drugs that effectively prevent and treat type 2 diabetes through new targets. With high-throughput screening, we found that sulfathiazole decreased the blood glucose and improved glucose metabolism in T2DM mice. Notably, we discovered that sulfathiazole treated T2DM by activating CYP19A1 protein to synthesize estrogen. Collectively, sulfathiazole along with CYP19A1 target bring new promise for the better therapy of T2DM.
Collapse
Affiliation(s)
- Zhuozhou Hu
- School of Pharmacy, Lanzhou University, 199 West Donggang Rd., Lanzhou, Gansu 730000, PR China
| | - Chun Li
- School of Pharmacy, Lanzhou University, 199 West Donggang Rd., Lanzhou, Gansu 730000, PR China
| | - Tongyu Wu
- School of Pharmacy, Lanzhou University, 199 West Donggang Rd., Lanzhou, Gansu 730000, PR China
| | - Jing Zhou
- School of Pharmacy, Lanzhou University, 199 West Donggang Rd., Lanzhou, Gansu 730000, PR China
| | - Liang Han
- School of Pharmacy, Lanzhou University, 199 West Donggang Rd., Lanzhou, Gansu 730000, PR China
| | - Jingjing Liu
- School of Pharmacy, Lanzhou University, 199 West Donggang Rd., Lanzhou, Gansu 730000, PR China
| | - Shaojia Qiang
- School of Pharmacy, Lanzhou University, 199 West Donggang Rd., Lanzhou, Gansu 730000, PR China
| | - Wenyang Zhao
- School of Pharmacy, Lanzhou University, 199 West Donggang Rd., Lanzhou, Gansu 730000, PR China
| | - Xiangxiang Li
- School of Pharmacy, Lanzhou University, 199 West Donggang Rd., Lanzhou, Gansu 730000, PR China
| | - Xiaohua Liu
- School of Pharmacy, Lanzhou University, 199 West Donggang Rd., Lanzhou, Gansu 730000, PR China
| | - Jiazhong Li
- School of Pharmacy, Lanzhou University, 199 West Donggang Rd., Lanzhou, Gansu 730000, PR China.
| | - Xinping Chen
- School of Pharmacy, Lanzhou University, 199 West Donggang Rd., Lanzhou, Gansu 730000, PR China; Southeast Research Institute of LZU, Putian, Fujian 351152, PR China; State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou 730000, PR China.
| |
Collapse
|
49
|
Protective Effects of Coumestrol on Metabolic Dysfunction and Its Estrogen Receptor-Mediated Action in Ovariectomized Mice. Nutrients 2023; 15:nu15040954. [PMID: 36839308 PMCID: PMC9966481 DOI: 10.3390/nu15040954] [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: 01/16/2023] [Revised: 02/10/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
Coumestrol, a phytoestrogen compound found in various plants, has been shown to act as a potent estrogen receptor (ER) agonist, with a higher binding affinity for ERβ than for ERα. However, there is currently limited information regarding its beneficial effects in postmenopausal disorders and its ER-mediated mechanisms. Herein, we investigated the effects of coumestrol (subcutaneous or oral treatment) on metabolic dysfunction in ovariectomized (OVX) mice fed a high-fat diet, in comparison with the effects of 17β-estradiol (E2) replacement. Coumestrol was administered daily at a dose of 5 mg/kg for 10 weeks. Coumestrol treatment through the subcutaneous route stimulated uterine growth in OVX mice at a level lower than that of E2. E2 and coumestrol prevented body fat accumulation, adipocyte hypertrophy, and hepatic steatosis, and enhanced voluntary physical activity. Coumestrol showed estrogen-mimetic effects in the regulation of the protein expressions involved in browning of white fat and insulin signaling, including increased hepatic expression of fibroblast growth factor 21. Importantly, the metabolic effects of coumestrol (oral administration at 10 mg/kg for 7 weeks) were mostly abolished following co-treatment with an ERβ-selective antagonist but not with an ERα-selective antagonist, indicating that the metabolic actions of coumestrol in OVX mice are primarily mediated by ERβ. These findings provide important insights into the beneficial effects of coumestrol as a phytoestrogen supplement for the prevention and treatment of postmenopausal symptoms.
Collapse
|
50
|
An S, Hwang SY, Gong J, Ahn S, Park IG, Oh S, Chin YW, Noh M. Computational Prediction of the Phenotypic Effect of Flavonoids on Adiponectin Biosynthesis. J Chem Inf Model 2023; 63:856-869. [PMID: 36716271 DOI: 10.1021/acs.jcim.3c00033] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
In silico machine learning applications for phenotype-based screening have primarily been limited due to the lack of machine-readable data related to disease phenotypes. Adiponectin, a nuclear receptor (NR)-regulated adipocytokine, is relatively downregulated in human metabolic diseases. Here, we present a machine-learning model to predict the adiponectin-secretion-promoting activity of flavonoid-associated phytochemicals (FAPs). We modeled a structure-activity relationship between the chemical similarity of FAPs and their bioactivities using a random forest-based classifier, which provided the NR activity of each FAP as a probability. To link the classifier-predicted NR activity to the phenotype, we next designed a single-cell transcriptomics-based multiple linear regression model to generate the relative adiponectin score (RAS) of FAPs. In experimental validation, estimated RAS values of FAPs isolated from Scutellaria baicalensis exhibited a significant correlation with their adiponectin-secretion-promoting activity. The combined cheminformatics and bioinformatics approach enables the computational reconstruction of phenotype-based screening systems.
Collapse
Affiliation(s)
- Seungchan An
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul08826, Republic of Korea
| | - Seok Young Hwang
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul08826, Republic of Korea
| | - Junpyo Gong
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul08826, Republic of Korea
| | - Sungjin Ahn
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul08826, Republic of Korea
| | - In Guk Park
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul08826, Republic of Korea
| | - Soyeon Oh
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul08826, Republic of Korea
| | - Young-Won Chin
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul08826, Republic of Korea
| | - Minsoo Noh
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul08826, Republic of Korea
| |
Collapse
|