1
|
Isah MB, Tajuddeen N, Yusuf A, Mohammed A, Ibrahim MA, Melzig M, Zhang X. The antidiabetic properties of lignans: a comprehensive review. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156717. [PMID: 40220408 DOI: 10.1016/j.phymed.2025.156717] [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: 01/27/2025] [Revised: 03/23/2025] [Accepted: 04/01/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Diabetes mellitus (DM) is a chronic metabolic disease with a high global prevalence. Lignans, a class of plant natural compounds found in commonly consumed foods, are well-tolerated by humans and have demonstrated promising potential in the management of DM. Consumption of lignan-rich foods has been associated with improved overall health and quality of life. PURPOSE The clinical and preclinical evidence on the role of lignans in managing DM are critically examined. METHODS A thorough literature search was conducted across major scientific databases, focusing on studies that reported the effects of individual lignans on key diabetes indicators, such as glucose utilisation and insulin sensitivity, in both human and animal models, as well as in cell-based studies. RESULTS A total of 180 lignans were included in the review. Out of these, only three were investigated in randomised clinical trials in humans and 31 in animal models. The reviewed evidence suggests some beneficial effects of lignans in preventing the development of obesity-related diabetes. Their therapeutic benefits in preventing diabetes-related complications, particularly diabetic nephropathy, in both type 1 and type 2 diabetes, are also supported. Metabolites of various lignans, produced by microbial metabolism in the gut and serum enzymes, appear to be key bioactive forms, highlighting the need for detailed pharmacodynamic studies, optimised dosage designs, and the use of the appropriate lignan molecules for cell-based screening. CONCLUSION Lignans and their microbial metabolites show promise in preventing obesity-related diabetes and mitigating diabetes-related complications such as diabetic nephropathy, though further clinical studies are needed to optimize their therapeutic potential.
Collapse
Affiliation(s)
- Murtala Bindawa Isah
- Chinese-German Joint Laboratory for Natural Product Research, Shaanxi International Cooperation Demonstration Base, Shaanxi University of Technology, Hanzhong, 723000, Shaanxi, China; Department of Biochemistry, Umaru Musa Yar'adua University Katsina, Nigeria.
| | - Nasir Tajuddeen
- Department of Chemistry, Ahmadu Bello University Zaria, Nigeria
| | - Anas Yusuf
- Chinese-German Joint Laboratory for Natural Product Research, Shaanxi International Cooperation Demonstration Base, Shaanxi University of Technology, Hanzhong, 723000, Shaanxi, China
| | - Aminu Mohammed
- Department of Biochemistry, Ahmadu Bello University Zaria, Nigeria
| | | | - Matthias Melzig
- Chinese-German Joint Laboratory for Natural Product Research, Shaanxi International Cooperation Demonstration Base, Shaanxi University of Technology, Hanzhong, 723000, Shaanxi, China; Freie Universitaet Berlin, Institute of Pharmacy, Berlin, Germany.
| | - Xiaoying Zhang
- Chinese-German Joint Laboratory for Natural Product Research, Shaanxi International Cooperation Demonstration Base, Shaanxi University of Technology, Hanzhong, 723000, Shaanxi, China; Centre of Molecular and Environmental Biology (CBMA), Department of Biology, University of Minho, Campus de Gualtar, Braga, 4710-057, Portugal; Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada.
| |
Collapse
|
2
|
Phungphong S, Suthivanich P, Boonhoh W, Punsawad C, Cheng Z, Bupha-Intr T. Targeting NLRP3 inflammasome attenuates cardiac pyroptosis and fibrosis in estrogen-deficient diabetic rats. Pflugers Arch 2025:10.1007/s00424-025-03092-6. [PMID: 40383837 DOI: 10.1007/s00424-025-03092-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 04/15/2025] [Accepted: 05/04/2025] [Indexed: 05/20/2025]
Abstract
Cardiac diastolic dysfunction is a hallmark of diabetic cardiomyopathy (DCM), particularly in postmenopausal women where estrogen deficiency exacerbates cardiac remodeling. This study investigated the roles of NLRP3 inflammasome activation and cardiac mast cell (CMC) behavior in diabetic ovariectomized (OVX) rat models. Female Wistar rats were divided into five groups: sham-operated, OVX, diabetic (Sham-DM), OVX-diabetic (OVX-DM), and OVX-DM treated with the NLRP3 inhibitor MCC950. Diabetes was induced using a high-calorie quick fat diet (13.8% crude fat, 24.35% crude protein, 25% sucrose; 406.80 kcal/100 g) followed by a single intraperitoneal injection of streptozotocin (30 mg/kg). MCC950 (10 mg/kg BW, intraperitoneally) was administered daily for 4 weeks. Echocardiography revealed significant diastolic dysfunction in OVX-DM rats, with increased left ventricular internal diameter (LVIDd) and reduced mitral valve E/A ratio, while MCC950 treatment partially restored diastolic function (p < 0.05). Masson's trichrome staining showed increased myocardial fibrosis in OVX-DM rats (2.59 ± 0.20%) compared to Sham-DM (1.94 ± 0.16%, p < 0.05), which was reduced with MCC950 treatment (0.88 ± 0.13%, p < 0.05). Western blot analysis demonstrated elevated expression of NLRP3, cleaved caspase-1, IL-1β, and GSDMD-N in OVX-DM hearts. MCC950 significantly reduced cleaved caspase-1, IL-1β, and GSDMD-N expression without altering NLRP3 protein levels. Additionally, mast cell degranulation was markedly increased in OVX-DM rats (62.14%) compared to controls (P < 0.05) and was attenuated by MCC950 (31.06%, P < 0.05). These findings suggest that NLRP3 inflammasome activation under conditions of estrogen deficiency and diabetes contributes to myocardial pyroptosis and mast cell degranulation, driving cardiac remodeling in postmenopausal DCM. Targeting NLRP3 pathways may provide an effective therapeutic strategy to mitigate diastolic dysfunction, fibrosis, and inflammation in diabetic hearts.
Collapse
Affiliation(s)
- Sukanya Phungphong
- Department of Medical Science, School of Medicine, Walailak University, Nakhon Si Thammarat, 80160, Thailand.
- Center of Excellence in Tropical Pathobiology, Walailak University, Nakhon Si Thammarat, 80160, Thailand.
| | - Phichaya Suthivanich
- Doctor of Philosophy Program in Physiology, Graduate School, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Worakan Boonhoh
- Akkhraratchakumari Veterinary College, Walailak University, Nakhon Si Thammarat, 80160, Thailand
| | - Chuchard Punsawad
- Department of Medical Science, School of Medicine, Walailak University, Nakhon Si Thammarat, 80160, Thailand
- Center of Excellence in Tropical Pathobiology, Walailak University, Nakhon Si Thammarat, 80160, Thailand
| | - Zhaokang Cheng
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA, 99202-2131, USA
| | - Tepmanas Bupha-Intr
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| |
Collapse
|
3
|
Parks S, John NO, Morris A, Shah K, Eves E, Adams D, Barry E, Causer V, Churm R, Ahmed-Evans S, Haggett AV, Patel R, Forde R, Reynolds R. Supporting women with diabetes experiencing menopause: A workshop to co-design research recommendations for improving the understanding of and support for women with diabetes experiencing menopause. Diabet Med 2025:e70039. [PMID: 40344580 DOI: 10.1111/dme.70039] [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: 03/25/2025] [Accepted: 03/28/2025] [Indexed: 05/11/2025]
Abstract
AIMS Based on direct feedback on the challenges faced by women with diabetes navigating menopause, the Diabetes Research Steering Groups (DRSGs), organised by Diabetes UK, conducted a workshop aimed at outlining key research priorities in this area. The statement will provide actionable recommendations for researchers and funders, ensuring future studies address the unique needs and lived experiences of women living with diabetes during this life phase. The DRSGs also highlighted the need to bring researchers and healthcare professionals in the field of diabetes together with those working in women's health to collaborate in this field. The goal is to bridge the gap in understanding and support, driving research that informs more effective care strategies and improves quality of life during this critical and often challenging life stage. METHODS In November 2023, Diabetes UK convened a workshop on diabetes and menopause, bringing together 49 participants, including people with lived experience, researchers, healthcare professionals, and representatives from charities and funding bodies. The workshop began with presentations from experts and case studies, followed by small-group discussions facilitated by Diabetes UK staff. Participants were asked to identify key evidence gaps and refine research questions related to diabetes and menopause. Groups focused on thematic topics, discussing barriers and potential solutions. Insights from these discussions were collated and thematically analysed, resulting in key recommendations to address health inequalities in diabetes research. RESULTS The following key areas were identified for increased future research focus: Improving the understanding of the mechanisms underlying the impact of menopause on blood glucose control in women with diabetes Understanding the safety and efficacy of treatments, technology, and exercise in the management of diabetes and menopause Understanding how to best support women from underserved communities in their management of diabetes and menopause Understanding the impact of menopause on the mental health of women with diabetes and tailoring psychosocial interventions that support women during this period. Supporting healthcare professional education in how to effectively support women with diabetes experiencing menopause. Exploring how innovative models of care can support women with diabetes experiencing menopause more effectively. CONCLUSIONS This position statement presents recommendations to address the unmet needs of women with diabetes experiencing menopause. The diabetes research community is to act upon these recommendations to ensure the views and needs of this group are well-represented.
Collapse
Affiliation(s)
| | - Nia Otake John
- Wolfson Institute of Population Health, Queen Mary University of London, London, UK
| | | | | | | | - Dawn Adams
- Expert by Lived Experience, Diabetes UK, London, UK
| | - Eleanor Barry
- Nuffield Department of Primary Care Health Sciences, Radcliffe Observatory Quarter, University of Oxford, Oxford, UK
| | - Vicki Causer
- Expert by Lived Experience, Diabetes UK, London, UK
| | - Rachel Churm
- Applied Sports Technology, Exercise and Medicine (A-STEM) Research Centre, Faculty of Science and Engineering, Swansea University, Swansea, UK
| | | | | | | | - Rita Forde
- School of Nursing and Midwifery, University College Cork, Cork, Ireland
- Care in Long Term Conditions Research Division, Florence Nightingale Faculty of Nursing, Midwifery and Palliative Care, King's College London, London, UK
| | - Rebecca Reynolds
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
4
|
Li J, Chen C, Dong X, Cao Y, Bao Z, Liu Y, Yan J, Zhang P, Guo Y, Zeng X. Associations of acrylamide biomarkers with reproductive lifespan: A cross-sectional study in U.S. women. Food Chem Toxicol 2025; 202:115500. [PMID: 40334970 DOI: 10.1016/j.fct.2025.115500] [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: 02/16/2025] [Revised: 04/23/2025] [Accepted: 04/29/2025] [Indexed: 05/09/2025]
Abstract
Acrylamide (AA), a potential carcinogen mainly present in thermally processed carbohydrate-rich foods, and cigarette smoke, forms acrylamide-hemoglobin adducts (HbAA) and its metabolite glycidamide-hemoglobin adducts (HbGA). Female reproductive lifespan (from menarche to menopause) is the key to women's health management and disease prevention. The reproductive lifespan of US women has shifted, but epidemiological research on its causes is lacking. This cross-sectional study included 1275 naturally postmenopausal women from four cycles of the US National Health and Nutrition Examination Survey, with a mean age of 61 ± 12 years. We examined the associations between HbAA, HbGA, the ratio of HbGA to HbAA (HbGA/HbAA) with female reproductive lifespan using linear regressions, restricted cubic splines (RCS) models, and threshold effect analyses. Results showed that HbAA and HbGA levels below 36.6 and 44.7 pmol/gHb showed no significant association with reproductive lifespan, but above these levels, the negative associations with reproductive lifespan strengthened. After adjusting for all covariates, each 2.7 - fold increase (one ln unit) in HbAA and HbGA was associated with a reduction in reproductive lifespan of 1.4 years (P = 0.003) and 2 years (P = 0.007) respectively. Higher levels of HbAA and HbGA shorten reproductive lifespan, while HbGA/HbAA showed no significant association. CLINICAL TRIAL: not applicable.
Collapse
Affiliation(s)
- Junying Li
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Zhengzhou Key Laboratory of Gynecological Disease's Early Diagnosis, Zhengzhou, China
| | - Chen Chen
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Zhengzhou Key Laboratory of Gynecological Disease's Early Diagnosis, Zhengzhou, China
| | - Xing Dong
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Zhengzhou Key Laboratory of Gynecological Disease's Early Diagnosis, Zhengzhou, China
| | - Yushan Cao
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Zhengzhou Key Laboratory of Gynecological Disease's Early Diagnosis, Zhengzhou, China
| | - Zhuo Bao
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Zhengzhou Key Laboratory of Gynecological Disease's Early Diagnosis, Zhengzhou, China
| | - Yixuan Liu
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Zhengzhou Key Laboratory of Gynecological Disease's Early Diagnosis, Zhengzhou, China
| | - Jinxiang Yan
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Zhengzhou Key Laboratory of Gynecological Disease's Early Diagnosis, Zhengzhou, China
| | - Pei Zhang
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Zhengzhou Key Laboratory of Gynecological Disease's Early Diagnosis, Zhengzhou, China
| | - Yongzhen Guo
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Zhengzhou Key Laboratory of Gynecological Disease's Early Diagnosis, Zhengzhou, China.
| | - Xianxu Zeng
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Zhengzhou Key Laboratory of Gynecological Disease's Early Diagnosis, Zhengzhou, China.
| |
Collapse
|
5
|
Yao J, Wu F, Xiu M. Interrelationships between glucose metabolism and gonadal hormones in female first-episode patients with schizophrenia. J Psychosom Res 2025; 192:112087. [PMID: 40068565 DOI: 10.1016/j.jpsychores.2025.112087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 02/20/2025] [Accepted: 03/02/2025] [Indexed: 04/26/2025]
Abstract
BACKGROUND Schizophrenia (SCZ) is a complex mental disorder that typically emerges in late adolescence or early adulthood. The underlying molecular mechanisms of SCZ remain unclear. There is growing evidence supporting the involvement of abnormalities in metabolic and endocrine systems in the pathophysiology of SCZ. Our study was designed to explore the interrelationship between gonadal hormones and glucose metabolism in patients with SCZ. METHODS One hundred and twenty-four female first-episode patients diagnosed with SCZ according to DSM-IV were recruited. The glucose metabolism parameters, including fasting glucose, insulin, and insulin resistance index were measured. In addition, gonadal hormones, including follicle-stimulating hormone, testosterone, estradiol, progesterone, and luteinizing hormone were also determined in the same group of patients. RESULTS Abnormal glucose metabolism and sex hormones were observed in female DNFES patients, with a particularly high rate of insulin resistance (49.6 %). Correlation analysis revealed significant associations between glucose metabolic disturbance and sex hormones (all p < 0.05). Moreover, regression analysis adjusted for age, waist circumference, and BMI revealed that estradiol was negatively correlated with fasting insulin in female patients (β = -0.37, t = -4.0, p < 0.001). CONCLUSION These results provide evidence supporting the presence of disturbance in glucose metabolism and gonadal hormones at the onset of SCZ in female patients, suggesting that these dysregulations interact with each other to be involved in the disease's pathophysiological process.
Collapse
Affiliation(s)
- Jing Yao
- Peking University HuiLongGuan Clinical Medical School, Beijing Huilongguan Hospital, Beijing, China
| | - Fengchun Wu
- Department of Psychiatry, The Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, China.
| | - Meihong Xiu
- Peking University HuiLongGuan Clinical Medical School, Beijing Huilongguan Hospital, Beijing, China
| |
Collapse
|
6
|
Panwar A, Malik SO, Adib M, Lopaschuk GD. Cardiac energy metabolism in diabetes: emerging therapeutic targets and clinical implications. Am J Physiol Heart Circ Physiol 2025; 328:H1089-H1112. [PMID: 40192025 DOI: 10.1152/ajpheart.00615.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/15/2024] [Accepted: 04/01/2025] [Indexed: 04/25/2025]
Abstract
Patients with diabetes are at an increased risk for developing diabetic cardiomyopathy and other cardiovascular complications. Alterations in cardiac energy metabolism in patients with diabetes, including an increase in mitochondrial fatty acid oxidation and a decrease in glucose oxidation, are important contributing factors to this increase in cardiovascular disease. A switch from glucose oxidation to fatty acid oxidation not only decreases cardiac efficiency due to increased oxygen consumption but it can also increase reactive oxygen species production, increase lipotoxicity, and redirect glucose into other metabolic pathways that, combined, can lead to heart dysfunction. Currently, there is a lack of therapeutics available to treat diabetes-induced heart failure that specifically target cardiac energy metabolism. However, it is becoming apparent that part of the benefit of existing agents such as GLP-1 receptor agonists and sodium-glucose cotransporter 2 inhibitors may be related to their effects on cardiac energy metabolism. In addition, direct approaches aimed at inhibiting cardiac fatty acid oxidation or increasing glucose oxidation hold future promise as potential therapeutic approaches to treat diabetes-induced cardiovascular disease.
Collapse
Affiliation(s)
- Archee Panwar
- Cardiovascular Research Centre, Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Sufyan O Malik
- Cardiovascular Research Centre, Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Muhtasim Adib
- Cardiovascular Research Centre, Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Gary D Lopaschuk
- Cardiovascular Research Centre, Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
7
|
Wu F, Zheng F, Li X, Wu D, Li H, Zeng Y, Tang Y, Liu S, Li A. Association between non-skimmed milk consumption and metabolic dysfunction-associated fatty liver disease in US adults: insights from NHANES data. BMC Gastroenterol 2025; 25:270. [PMID: 40251472 PMCID: PMC12007357 DOI: 10.1186/s12876-025-03834-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 04/01/2025] [Indexed: 04/20/2025] Open
Abstract
PURPOSE Previous studies on the association between non-skimmed milk consumption and non-alcoholic fatty liver disease (NAFLD) have reported inconsistent findings, with some suggesting an increased risk and others indicating a protective effect. Moreover, as the research focus has shifted globally from NAFLD to metabolic dysfunction-associated fatty liver disease (MAFLD), there remains limited evidence on the relationship between non-skimmed milk consumption and MAFLD. The objective of this cross-sectional study was to investigate this association using data from the National Health and Nutrition Examination Survey (NHANES). METHODS In this U.S. population-based study, adults with complete information on non-skimmed milk consumption and MAFLD diagnosis from the 2017-March 2020 Pre-Pandemic NHANES were included. MAFLD was defined using the controlled attenuation parameter (CAP). The association between non-skimmed milk consumption and MAFLD was assessed using weighted multivariable logistic regression, adjusting for potential confounders. Subgroup and sensitivity analyses were conducted to evaluate effect modifications and robustness. RESULTS The study involved 3,758 participants in total, 1,423 (37.87%) of whom had MAFLD according to the diagnosis. Frequent non-skimmed milk consumption was independently associated with higher MAFLD risk. Compared to the "Rarely" group (< 1 time/week), the "Sometimes" group (≥ 1 time/week but < 1 time/day) had an odds ratio (OR) of 1.67 (95% CI: 1.32-2.12, P = 0.004), and the "Often" group (≥ 1 time/day) had an OR of 1.36 (95% CI: 1.06-1.75, P = 0.046). Stratified analysis revealed that the association was significantly modified by education level (P for interaction = 0.010), with a stronger association observed among participants with higher education levels. Sensitivity analysis yielded consistent results, further supporting the robustness of the association. CONCLUSION Our findings suggest a significant association between frequent non-skimmed milk consumption and risk of MAFLD, particularly in highly educated individuals. These results highlight the importance of dietary modifications, specifically reducing non-skimmed milk intake, as a potential preventive strategy for MAFLD, especially in high-risk populations.
Collapse
Affiliation(s)
- Futao Wu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fuying Zheng
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xue Li
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Danzhu Wu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Honghao Li
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Gastroenterology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, China
| | - Yingyi Zeng
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yan Tang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Side Liu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
- Department of Gastroenterology, Zhuhai Peoples Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, China.
| | - Aimin Li
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
8
|
Kong X, Wang W. Prediabetes Phenotypes and All-Cause or Cardiovascular Mortality: Evidence From a Population-Based Study. Endocr Pract 2025; 31:486-493. [PMID: 39837477 DOI: 10.1016/j.eprac.2025.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 01/07/2025] [Accepted: 01/13/2025] [Indexed: 01/23/2025]
Abstract
OBJECTIVE Fasting plasma glucose (FPG), glycated hemoglobin A1C (HbA1C), and 2-hour postload plasma glucose (2h PG) are all currently used to define prediabetes. We aimed to determine whether a higher number of prediabetes defects correspond to an increased all-cause and cardiovascular disease (CVD) mortality. METHODS Individuals with prediabetes and available information on FPG, HbA1C, 2h PG, and mortality data were derived from the 2005-2016 National Health and Nutrition Examination Survey. Kaplan-Meier survival curves, Cox proportional hazards regression analysis, and stratified analysis were used to compare all-cause and CVD mortality among participants with one, two, and all three defects. RESULTS Among the 4511 individuals included, 76.31%, 30.89%, and 41.65% met the FPG-, 2h PG-, and HbA1C-defined criteria for prediabetes, respectively. There were 2609 (60.78%), 1420 (29.60%), and 482 (9.62%) adults meeting one, two, and all three criteria for prediabetes, respectively. During a median follow-up of 100 months, a total of 534 (180 CVD-related) deaths occurred. The multivariable-adjusted hazard ratios and 95% confidence intervals in those meeting two and three criteria were 1.341 (1.042-1.727) and 1.369 (1.027-1.824), respectively, for all-cause mortality (P for trend = 0.006), and 1.836 (1.228-2.744) and 2.037 (1.092-3.801), respectively, for CVD mortality (P for trend = 0.002), with those meeting only one criterion as the reference. In subgroup analysis, the association between the number of diagnostic criteria for prediabetes and CVD mortality was observed only in men. CONCLUSIONS A higher number of diagnostic criteria for prediabetes was associated with increased all-cause and CVD mortality.
Collapse
Affiliation(s)
- Xiufang Kong
- Department of Rheumatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wei Wang
- Department of Nephrology, Shanghai Tenth People's Hospital, Shanghai, China.
| |
Collapse
|
9
|
Bapat P, Budhram DR, Bakhsh A, Abuabat MI, Verhoeff NJ, Mumford D, Cheema W, Falappa C, Orszag A, Jain A, Cherney DZI, Fralick M, Weisman A, Tomlinson G, Lovblom LE, Perkins BA. Longitudinal Determination of Diabetes Complications and Other Clinical Variables as Risk Factors for Diabetic Ketoacidosis in Type 1 Diabetes. Diabetes Care 2025; 48:614-622. [PMID: 39950992 DOI: 10.2337/dc24-2385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 01/22/2025] [Indexed: 03/23/2025]
Abstract
OBJECTIVE We aimed to determine whether diabetes complications, such as kidney disease that may impair acid-base buffering capacity, independently predict the risk of subsequent diabetic ketoacidosis (DKA). RESEARCH DESIGN AND METHODS We accessed previously collected 34-year data from the Diabetes Control and Complications Trial and Epidemiology of Diabetes Interventions and Complications study through public data access. Multivariable Cox proportional hazards models with time-varying exposures and covariates were used to examine the associations of macrovascular disease and early and late stages of neuropathy, nephropathy, and retinopathy, with subsequent DKA occurrence as the outcome. RESULTS Of 1,441 participants, 297 experienced 488 DKA events over follow-up. Major adverse cardiovascular events [hazard ratio (HR) 3.16, 95% CI 1.57-6.35, P = 0.001] and late-stage neuropathy, which comprised serious foot ulcer or amputation (HR 1.59, 95% CI 1.04-2.45, P = 0.03) were independently associated with higher DKA risk. Higher risk was also associated with shorter diabetes duration (HR 0.76, 95% CI 0.64-0.91, P = 0.002), female sex (HR 2.04, 95% CI 1.56-2.67, P < 0.001), current insulin pump use (HR 3.04, 95% CI 2.29-4.02, P < 0.001), higher time-updated HbA1c (per additional 1%: HR 1.39, 95% CI 1.29-1.50, P < 0.001), and higher current insulin dose (per 1 additional unit/kg/day: HR 2.32, 95% CI 1.62-3.33, P < 0.001). CONCLUSIONS A major cardiovascular event, foot ulcer, or amputation confers the greatest risk of future DKA independent of previously recognized risk factors, implying a need to target patients with these events for DKA prevention interventions, such as self-management skills for metabolic control, management of depression, and DKA education.
Collapse
Affiliation(s)
- Priya Bapat
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
- Division of Endocrinology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Dalton R Budhram
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
- Division of General Internal Medicine, Department of Medicine, University Health Network and Sinai Health, University of Toronto, Toronto, Ontario, Canada
| | - Abdulmohsen Bakhsh
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
- Division of Endocrinology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Kidney & Pancreas Health Centre, Organ Transplant Centre of Excellence, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - Mohammad I Abuabat
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
- Division of Endocrinology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Internal Medicine and Critical Care Department, King Abdullah bin Abdulaziz University Hospital, Princess Norah University, Riyadh, Saudi Arabia
| | - Natasha J Verhoeff
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
- Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Doug Mumford
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
- Patient-partner
| | - Wajeeha Cheema
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
- Patient-partner
| | - Cesar Falappa
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
- Division of General Internal Medicine, Department of Medicine, University Health Network and Sinai Health, University of Toronto, Toronto, Ontario, Canada
| | - Andrej Orszag
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
- Division of General Internal Medicine, Department of Medicine, University Health Network and Sinai Health, University of Toronto, Toronto, Ontario, Canada
| | - Akshay Jain
- TLC Diabetes and Endocrinology, Surrey, British Columbia, Canada
- Division of Endocrinology, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - David Z I Cherney
- Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Division of Nephrology, Department of Medicine, University Health Network, Toronto, Ontario, Canada
| | - Michael Fralick
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
- Division of General Internal Medicine, Department of Medicine, University Health Network and Sinai Health, University of Toronto, Toronto, Ontario, Canada
| | - Alanna Weisman
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
- Division of Endocrinology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Institute of Health Policy, Management, and Evaluation, University of Toronto, Toronto, Ontario, Canada
| | - George Tomlinson
- Institute of Health Policy, Management, and Evaluation, University of Toronto, Toronto, Ontario, Canada
- Biostatistics Division, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
- Department of Medicine, University Health Network and Sinai Health, University of Toronto, Toronto, Ontario, Canada
| | - Leif Erik Lovblom
- Institute of Health Policy, Management, and Evaluation, University of Toronto, Toronto, Ontario, Canada
- Biostatistics Department, University Health Network, Toronto, Ontario, Canada
| | - Bruce A Perkins
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
- Division of Endocrinology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Patient-partner
- Institute of Health Policy, Management, and Evaluation, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
10
|
Jin Z, Zheng L, Sun C, Xu B, Guo X, Zhang Y, Li L, Wei X. More comprehensive relationship between eGDR and sarcopenia in China: a nationwide cohort study with national representation. Diabetol Metab Syndr 2025; 17:97. [PMID: 40122882 PMCID: PMC11931793 DOI: 10.1186/s13098-025-01657-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 03/04/2025] [Indexed: 03/25/2025] Open
Abstract
INTRODUCTION Although studies had shown that Insulin resistance (IR) was correlated with the occurrence of sarcopenia, there were still many controversial conclusions. Therefore, we conducted a more comprehensive study on the relationship between the estimated glucose disposal rate (eGDR), an alternative indicator of IR, and the risk of sarcopenia, muscle mass, and muscle strength to clarify their interactions. METHODS The Study included individuals from The China Health and Retirement Longitudinal Study (CHARLS) who had complete eGDR data at baseline and did not develop low muscle mass and low muscle strength. The individuals were divided into four subgroups based on the quartile (Q) of the eGDR. The lowest quartile (Q1) of the eGDR was used as a reference. Logistic regression and linear regression were used to evaluate the relationship between eGDR and sarcopenia (low muscle mass, low muscle strength, possible sarcopenia, and sarcopenia) and sarcopenia related features (ASM/Ht2, grip, and RMS), respectively. In addition, we further evaluated the nonlinear relationship using smooth curve fitting and threshold effect analysis. RESULTS The results showed that after adjusting for confounders, eGDR was negatively associated with the risk of sarcopenia and positively associated with sarcopenia related characteristics. In addition, men showed a more significant reduction in the likelihood of low muscle mass compared to women. But as eGDR levels rise, women gain more ASM/Ht2. Further nonlinear analysis revealed an inverse correlation between eGDR and ASM/Ht2 at the inflection point of 15.3893. Besides that, eGDR was positively correlated with grip (7.1862) and RMS (11.1042) before the inflection point. CONCLUSIONS The study found that higher levels of eGDR were associated with a lower risk of developing sarcopenia. However, the effects of eGDR on muscle mass and muscle strength need to be considered comprehensively. For muscle mass, it is recommended to maintain eGDR below 15.3893, and for muscle strength, it is recommended to maintain eGDR below 7.1862, with more potential benefits for early warning of sarcopenia.
Collapse
Affiliation(s)
- Zikai Jin
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, 100102, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Liming Zheng
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, 100102, China
| | - Chuanrui Sun
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, 100102, China
| | - Bo Xu
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, 100102, China
| | - Xiangyun Guo
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Yili Zhang
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Linghui Li
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, 100102, China.
| | - Xu Wei
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, 100102, China.
| |
Collapse
|
11
|
Mittal R, Prasad K, Lemos JRN, Arevalo G, Hirani K. Unveiling Gestational Diabetes: An Overview of Pathophysiology and Management. Int J Mol Sci 2025; 26:2320. [PMID: 40076938 PMCID: PMC11900321 DOI: 10.3390/ijms26052320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 02/14/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
Gestational diabetes mellitus (GDM) is characterized by an inadequate pancreatic β-cell response to pregnancy-induced insulin resistance, resulting in hyperglycemia. The pathophysiology involves reduced incretin hormone secretion and signaling, specifically decreased glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP), impairing insulinotropic effects. Pro-inflammatory cytokines, including tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6), impair insulin receptor substrate-1 (IRS-1) phosphorylation, disrupting insulin-mediated glucose uptake. β-cell dysfunction in GDM is associated with decreased pancreatic duodenal homeobox 1 (PDX1) expression, increased endoplasmic reticulum stress markers (CHOP, GRP78), and mitochondrial dysfunction leading to impaired ATP production and reduced glucose-stimulated insulin secretion. Excessive gestational weight gain exacerbates insulin resistance through hyperleptinemia, which downregulates insulin receptor expression via JAK/STAT signaling. Additionally, hypoadiponectinemia decreases AMP-activated protein kinase (AMPK) activation in skeletal muscle, impairing GLUT4 translocation. Placental hormones such as human placental lactogen (hPL) induce lipolysis, increasing circulating free fatty acids which activate protein kinase C, inhibiting insulin signaling. Placental 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) overactivity elevates cortisol levels, which activate glucocorticoid receptors to further reduce insulin sensitivity. GDM diagnostic thresholds (≥92 mg/dL fasting, ≥153 mg/dL post-load) are lower than type 2 diabetes to prevent fetal hyperinsulinemia and macrosomia. Management strategies focus on lifestyle modifications, including dietary carbohydrate restriction and exercise. Pharmacological interventions, such as insulin or metformin, aim to restore AMPK signaling and reduce hepatic glucose output. Emerging therapies, such as glucagon-like peptide-1 receptor (GLP-1R) agonists, show potential in improving glycemic control and reducing inflammation. A mechanistic understanding of GDM pathophysiology is essential for developing targeted therapeutic strategies to prevent both adverse pregnancy outcomes and the progression to overt diabetes in affected women.
Collapse
Affiliation(s)
| | | | | | | | - Khemraj Hirani
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (K.P.); (J.R.N.L.); (G.A.)
| |
Collapse
|
12
|
Feng Y, Jin X, Zhu J, Yuan M, Zhu L, Ye D, Shen Y. Association between endogenous estradiol, testosterone, and long-term mortality in adults with prediabetes and diabetes: Evidence from NHANES database. J Diabetes Investig 2025; 16:481-491. [PMID: 39705158 PMCID: PMC11871383 DOI: 10.1111/jdi.14367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/28/2024] [Accepted: 11/17/2024] [Indexed: 12/22/2024] Open
Abstract
AIM AND INTRODUCTION Diabetes and prediabetes pose significant global public health challenges. Sex steroids, particularly testosterone and estradiol, play crucial roles in various metabolic processes. This study investigates the relationship between sex hormone levels and long-term mortality in adults with prediabetes and diabetes, as well as those without glucose intolerance. MATERIAL AND METHODS This retrospective cohort study utilized data from the NHANES 2013-2016, including adults aged 50-79 across prediabetic, diabetic, and non-diabetic groups. Serum testosterone, estradiol, and their ratios (T/E) were analyzed. The primary outcomes were all-cause mortality and CVD mortality tracked until December 2019. Cox regression models estimated the associations between hormone levels and mortality risks. RESULTS The study included 3,665 participants (male: 2,140; female: 1,775). In males with prediabetes, higher estradiol (adjusted hazard ratio [aHR] = 0.17, 95% confidence interval [CI]: 0.07-0.43) or testosterone (aHR = 0.39, 95% CI: 0.31-0.50) was significantly associated with lower risk of all-cause mortality. Higher estradiol (aHR = 0.12, 95% CI: 0.04-0.32) or testosterone (aHR = 0.36, 95% CI: 0.27-0.48) was significantly associated with lower CVD mortality risk. In females with diabetes, there was a significant association between higher estradiol levels (aHR = 0.22, 95% CI: 0.06-0.83) or T/E ratio (aHR = 0.18, 95% CI: 0.04-0.73) with a reduced all-cause mortality risk. CONCLUSIONS This study identifies some novel associations between estradiol, testosterone, and their ratios with long-term mortality in men and women across different glycemic statuses. These findings suggest a potential protective role of sex hormones in individuals with altered glucose metabolism, with gender difference, warranting further investigation.
Collapse
Affiliation(s)
- Ye Feng
- Department of Endocrinology and Metabolic Disease, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Xi Jin
- Department of Endocrinology and Metabolic Disease, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Jing Zhu
- Department of Endocrinology and Metabolic Disease, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Meng Yuan
- Department of Endocrinology and Metabolic Disease, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Liang Zhu
- Department of Clinical Pharmacy, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Dan Ye
- Department of Endocrinology and Metabolic Disease, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Yuqing Shen
- Department of Endocrinology and Metabolic Disease, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| |
Collapse
|
13
|
Buscemi C, Randazzo C, Barile AM, Caldarella R, Murro I, Caruso R, Colombrita P, Lombardo M, De Pergola G, Buscemi S. The impact of breakfast skipping on plasma glucose levels in non-diabetic individuals: gender-based differences and implications. Int J Food Sci Nutr 2025; 76:203-208. [PMID: 39743858 DOI: 10.1080/09637486.2024.2446886] [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: 08/28/2024] [Revised: 12/04/2024] [Accepted: 12/19/2024] [Indexed: 01/04/2025]
Abstract
Chrono-nutritional factors may play a significant role in glucose regulation and the risk of type 2 diabetes (T2D). We investigated the association between breakfast skipping (BS) and fasting plasma glucose (FPG) levels in a non-diabetic cohort of the ABCD project (nutrition, cardiovascular wellness and diabetes) representing the general population of Palermo, Italy Among the 623 participants included in the analysis, 118 individuals were identified as BS. In men, BS was associated with higher values of FPG (p < .05), glycated haemoglobin (p < .01), triglycerides (p < .05) and insulin (p < .005). Among women, no significant differences were observed between BS and non-skippers. Multivariate regression analysis confirmed that BS was an independent predictor of higher FPG levels in men (p < .01); waist circumference emerged as the primary predictor of FPG in women (p < .001). This study supports the importance of dietary habits in glycaemic control and T2D risk.
Collapse
Affiliation(s)
- Carola Buscemi
- Dipartimento di Promozione della Salute, Materno-Infantile, Medicina Interna e Specialistica di Eccellenza (PROMISE), University of Palermo, Palermo, Italy
- Unit of Clinical Nutrition, Obesity and Metabolic Diseases, AOU Policlinico "P. Giaccone", Palermo, Italy
| | - Cristiana Randazzo
- Dipartimento di Promozione della Salute, Materno-Infantile, Medicina Interna e Specialistica di Eccellenza (PROMISE), University of Palermo, Palermo, Italy
- Unit of Clinical Nutrition, Obesity and Metabolic Diseases, AOU Policlinico "P. Giaccone", Palermo, Italy
| | - Anna Maria Barile
- Dipartimento di Promozione della Salute, Materno-Infantile, Medicina Interna e Specialistica di Eccellenza (PROMISE), University of Palermo, Palermo, Italy
- Unit of Clinical Nutrition, Obesity and Metabolic Diseases, AOU Policlinico "P. Giaccone", Palermo, Italy
| | - Rosalia Caldarella
- Dipartimento di Promozione della Salute, Materno-Infantile, Medicina Interna e Specialistica di Eccellenza (PROMISE), University of Palermo, Palermo, Italy
- Unit of Laboratory Medicine, AOU Policlinico "P. Giaccone", Palermo, Italy
| | - Isanna Murro
- Center of Nutrition for the Research and the Care of Obesity and Metabolic Diseases, National Institute of Gastroenterology IRCCS "Saverio de Bellis", Castellana Grotte, Bari, Italy
| | - Roberta Caruso
- Dipartimento di Promozione della Salute, Materno-Infantile, Medicina Interna e Specialistica di Eccellenza (PROMISE), University of Palermo, Palermo, Italy
- Unit of Clinical Nutrition, Obesity and Metabolic Diseases, AOU Policlinico "P. Giaccone", Palermo, Italy
| | - Piero Colombrita
- Dipartimento di Promozione della Salute, Materno-Infantile, Medicina Interna e Specialistica di Eccellenza (PROMISE), University of Palermo, Palermo, Italy
- Unit of Clinical Nutrition, Obesity and Metabolic Diseases, AOU Policlinico "P. Giaccone", Palermo, Italy
| | - Martina Lombardo
- Dipartimento di Promozione della Salute, Materno-Infantile, Medicina Interna e Specialistica di Eccellenza (PROMISE), University of Palermo, Palermo, Italy
- Unit of Clinical Nutrition, Obesity and Metabolic Diseases, AOU Policlinico "P. Giaccone", Palermo, Italy
| | - Giovanni De Pergola
- Center of Nutrition for the Research and the Care of Obesity and Metabolic Diseases, National Institute of Gastroenterology IRCCS "Saverio de Bellis", Castellana Grotte, Bari, Italy
| | - Silvio Buscemi
- Dipartimento di Promozione della Salute, Materno-Infantile, Medicina Interna e Specialistica di Eccellenza (PROMISE), University of Palermo, Palermo, Italy
- Unit of Clinical Nutrition, Obesity and Metabolic Diseases, AOU Policlinico "P. Giaccone", Palermo, Italy
| |
Collapse
|
14
|
Gupta S, Afzal M, Agrawal N, Almalki WH, Rana M, Gangola S, Chinni SV, Kumar K B, Ali H, Singh SK, Jha SK, Gupta G. Harnessing the FOXO-SIRT1 axis: insights into cellular stress, metabolism, and aging. Biogerontology 2025; 26:65. [PMID: 40011269 DOI: 10.1007/s10522-025-10207-0] [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: 02/02/2025] [Accepted: 02/15/2025] [Indexed: 02/28/2025]
Abstract
Aging and metabolic disorders share intricate molecular pathways, with the Forkhead box O (FOXO)- Sirtuin 1 (SIRT1) axis emerging as a pivotal regulator of cellular stress adaptation, metabolic homeostasis, and longevity. This axis integrates nutrient signaling with oxidative stress defence, modulating glucose and lipid metabolism, mitochondrial function, and autophagy to maintain cellular stability. FOXO transcription factors, regulated by SIRT1 deacetylation, enhance antioxidant defence mechanisms, activating genes such as superoxide dismutase (SOD) and catalase, thereby counteracting oxidative stress and metabolic dysregulation. Recent evidence highlights the dynamic role of reactive oxygen species (ROS) as secondary messengers in redox signaling, influencing FOXO-SIRT1 activity in metabolic adaptation. Additionally, key redox-sensitive regulators such as nuclear factor erythroid 2-related factor 2 (Nrf2) and Peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α) interact with this pathway, orchestrating mitochondrial biogenesis and adaptive stress responses. Pharmacological interventions, including alpha-lipoic acid (ALA), resveratrol, curcumin and NAD+ precursors, exhibit therapeutic potential by enhancing insulin sensitivity, reducing oxidative burden, and restoring metabolic balance. This review synthesizes current advancements in FOXO-SIRT1 regulation, its emerging role in redox homeostasis, and its therapeutic relevance, offering insights into future strategies for combating metabolic dysfunction and aging-related diseases.
Collapse
Affiliation(s)
- Saurabh Gupta
- Department of Pharmacology, Chameli Devi Institute of Pharmacy, Khandwa Road, Village Umrikheda, Near Tollbooth, Indore, Madhya Pradesh, 452020, India
| | - Muhammad Afzal
- Pharmacy Program, Department of Pharmaceutical Sciences, Batterjee Medical College, P.O. Box 6231, 21442, Jeddah, Saudi Arabia
| | - Neetu Agrawal
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Mohit Rana
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Saurabh Gangola
- Department of Microbiology, Graphic Era Deemed to be University, Dehradun, 248002, India
| | - Suresh V Chinni
- Department of Biochemistry, Faculty of Medicine, Bioscience, and Nursing, MAHSA University, 42610, Jenjarom, Selangor, Malaysia
| | - Benod Kumar K
- Department of General Surgery, Consultant Head and Neck Surgical Oncology, Dr.D.Y.Patil Medical College, Hospital and Research Centre, Pimpri, Pune, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T Road, Phagwara, Punjab, India
- Sunway Biofunctional Molecules Discovery Centre (SBMDC), School of Medical and Life Sciences, Sunway University, Sunway, Malaysia
| | - Saurabh Kumar Jha
- Department of Zoology, Kalindi College, University of Delhi, 110008, New Delhi, India
- Centre for Himalayan Studies, University of Delhi, Delhi, 110007, India
| | - Gaurav Gupta
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India.
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates.
| |
Collapse
|
15
|
Yan X, Li L, Gao J, Wang L, Ai K, Lei X, Tang M, Zhang X, Zhang D. Sex differences in intracranial plaque burden in patients with type 2 diabetes mellitus with acute ischemic cerebrovascular disease: a pilot study based on high-resolution MRI. Front Endocrinol (Lausanne) 2025; 15:1417240. [PMID: 39926392 PMCID: PMC11802420 DOI: 10.3389/fendo.2024.1417240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 12/30/2024] [Indexed: 02/11/2025] Open
Abstract
Background Atherosclerosis (AS) is the main cause of macrovascular disease. Previous studies have found sex differences in the prevalence of type 2 diabetes mellitus (T2DM) and its associated macrovascular disease outcomes. However, the relationship between sex differences, T2DM, and AS is not fully understood. This study attempts to explore possible associations between sex, treatment, and the burden of intracranial atherosclerosis (ICAS) in patients with T2DM who have experienced an acute ischemic cerebrovascular disease. Methods We focused on patients with T2DM with acute ischemic stroke or transient ischemic attack due to intracranial atherosclerotic stenosis. ICAS was assessed by 3T cardiovascular magnetic resonance vascular wall imaging. Plaque counts of the total, proximal, and distal intracranial arteries were used to assess plaque burden. Patients with a history of T2DM and currently taking hypoglycemic drugs were defined as being treated. Poisson regression models or negative binomial regression models were used to analyze the interaction between sex and treatment in relation to plaque burden. Results A total of 495 plaques were detected in 120 patients (75 male; mean age, 60.77 ± 11.01 years), including 311 proximal and 184 distal plaques. The intracranial culprit plaque was located proximal to the artery in both male (85.3%) and female (88.9%) patients. The adjusted total and proximal intracranial plaque burdens were 1.261 times (95% confidence interval [CI], 1.050-1.515, P=0.013) and 1.322 times (95%CI, 1.055-1.682, P=0.016) higher in male than in female patients. The risk ratio for proximal plaque burden in untreated male versus female patients was 0.966 (95%CI, 0.704-1.769). However, the proximal plaque risk ratio for treated male versus female patients was 1.530 (95%CI, 1.076-2.174). The interaction of sex and treatment significantly affected the proximal plaque burden. Conclusion Male patients with T2DM and acute cerebrovascular disease have a significantly higher adjusted risk of total and proximal intracranial plaque burden compared to female patients. Female patients undergoing antidiabetic treatment have a significantly reduced risk of proximal plaque to males. Considering that culprit plaques tend to accumulate in the proximal arteries, understanding how to reduce the burden of proximal plaques may help reduce the risk of adverse cerebrovascular events.
Collapse
Affiliation(s)
- Xuejiao Yan
- Department of Magnetic Resonance Imaging (MRI), Shaanxi Provincial People’s Hospital, Xi’an, Shaanxi, China
| | - Ling Li
- Department of Magnetic Resonance Imaging (MRI), Shaanxi Provincial People’s Hospital, Xi’an, Shaanxi, China
| | - Jie Gao
- Department of Magnetic Resonance Imaging (MRI), Shaanxi Provincial People’s Hospital, Xi’an, Shaanxi, China
| | - Lihui Wang
- Department of Radiology, Xi‘an International Medical Center Hospital, Xi’an, Shaanxi, China
| | - Kai Ai
- Department of Clinical Science, Philips Healthcare, Xi’an, China
| | - Xiaoyan Lei
- Department of Magnetic Resonance Imaging (MRI), Shaanxi Provincial People’s Hospital, Xi’an, Shaanxi, China
| | - Min Tang
- Department of Magnetic Resonance Imaging (MRI), Shaanxi Provincial People’s Hospital, Xi’an, Shaanxi, China
| | - Xiaoling Zhang
- Department of Magnetic Resonance Imaging (MRI), Shaanxi Provincial People’s Hospital, Xi’an, Shaanxi, China
| | - Dongsheng Zhang
- Department of Magnetic Resonance Imaging (MRI), Shaanxi Provincial People’s Hospital, Xi’an, Shaanxi, China
| |
Collapse
|
16
|
Maleki Sedgi F, Mozaffari N, Pashaei MR, Hajizadeh-Sharafabad F. Effect of fermented soybean on metabolic outcomes, anthropometric indices, and body composition: a systematic review and meta-analysis of clinical trials. Food Funct 2025; 16:389-405. [PMID: 39763426 DOI: 10.1039/d4fo02668c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2025]
Abstract
The aim of the current study was to systematically review and quantify the findings of randomized controlled trials (RCTs) assessing the effect of fermented soy products on anthropometric indices, body composition, and metabolic outcomes. PubMed, Scopus, and Web of Science were searched to identify the relevant articles from inception until March 2024. The weighted mean differences (WMD) and corresponding 95% confidence intervals (CI) were calculated as effect sizes and analyzed using the random-effects method. A total of 2205 records were found, of which 15 RCTs were eligible. Results demonstrated significant beneficial effects of fermented soy on body mass index (WMD = -0.14 kg m-2, 95% CI: -0.28, -0.01, P = 0.039), waist circumference (WMD = -1.50 cm, 95% CI: -2.94, -0.07, P = 0.04), visceral fat (WMD = -692.17 mm2, 95% CI: -1011.58, -372.77, P < 0.001), fasting plasma glucose (WMD = -6.39 mg dL-1, 95% CI: -10.38, -2.40, P = 0.002), and total cholesterol (WMD = -5.0 mg dL-1, 95% CI: -6.60, -3.39, P < 0.001) compared with controls. However, the responses of other parameters to fermented soy were not significant. Overall, fermented soy may confer health benefits on certain metabolic outcomes, anthropometric indices, and body composition.
Collapse
Affiliation(s)
- Fatemeh Maleki Sedgi
- Department of Nutrition, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Nazanin Mozaffari
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Mohammad Reza Pashaei
- Patient Safety Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia, Iran.
- Department of Internal Medicine, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | | |
Collapse
|
17
|
Oviedo JM, Cortes-Selva D, Marchetti M, Gordon L, Gibbs L, Maschek JA, Cox J, Frietze S, Amiel E, Fairfax KC. Schistosoma mansoni antigen induced innate immune memory features mitochondrial biogenesis and can be inhibited by ovarian produced hormones. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.14.632838. [PMID: 39868249 PMCID: PMC11761400 DOI: 10.1101/2025.01.14.632838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
We have previously identified that S. mansoni infection induces a unique form of myeloid training that protects male but not female mice from high fat diet induced disease. Here we demonstrate that ovarian derived hormones account for this sex specific difference. Ovariectomy of females prior to infection permits metabolic reprogramming of the myeloid lineage, with BMDM exhibiting carbon source flexibility for cellular respiration, and mice protected from systemic metabolic disease. The innate training phenotype of infection can be replicated by in vivo injection of SEA, and by exposure of bone marrow to SEA in culture prior to macrophage differentiation (Day 0). This protective phenotype is linked to increased chromatin accessibility of lipid and mitochondrial pathways in BMDM including Nrf1 and Tfam, as well as mitochondrial biogenesis. This work provides evidence that S. mansoni antigens induce a unique form of innate training inhibited by ovarian-derived hormones in females.
Collapse
Affiliation(s)
- Juan Marcos Oviedo
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City UT, 84112
| | - Diana Cortes-Selva
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City UT, 84112
| | - Marco Marchetti
- Department of Human Genetics, Utah Center for Genetic Discovery, University of Utah, Salt Lake City, UT, 84112, USA
| | | | - Lisa Gibbs
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City UT, 84112
| | - J. Alan Maschek
- Metabolomics, Proteomics and Mass Spectrometry Cores, University of Utah, Salt Lake City, UT, 84112
- Department of Nutrition and Integrative Physiology and the Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, UT 84112
| | - James Cox
- Department of Biochemistry, University of Utah, Salt Lake City UT, 84112
- Metabolomics, Proteomics and Mass Spectrometry Cores, University of Utah, Salt Lake City, UT, 84112
| | - Seth Frietze
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT 05405
| | - Eyal Amiel
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT 05405
| | - Keke C. Fairfax
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City UT, 84112
| |
Collapse
|
18
|
Yang Y, Chen Y, Liu C, Wang S, Zhao Y, Cao W, Wang K. Analysis of Exosomal miRNA and Hepatic mRNA Expression in the Dysregulation of Insulin Action in Perimenopausal Mice. J Diabetes Res 2025; 2025:6251747. [PMID: 39823117 PMCID: PMC11737908 DOI: 10.1155/jdr/6251747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 12/04/2024] [Accepted: 12/19/2024] [Indexed: 01/19/2025] Open
Abstract
Introduction: The aim of present study was to evaluate the impact of perimenopause on insulin resistance. Specifically, insulin sensitivity was assessed in a perimenopausal mouse model treated with 4-vinylcyclohexene diepoxide (VCD), together with the changes in exosomal miRNA and hepatic mRNA expression profiles. Methods: Homeostasis model assessment of insulin resistance (HOMA-IR) was utilized to assess the status of insulin resistance, and insulin action was evaluated during menopausal transition. RNA sequencing (RNA-seq) analysis was used to identify altered expression profiles of exosomal miRNAs and hepatic mRNAs. Differentially expressed miRNA (DEM)-differentially expressed gene (DEG) network analyses were also conducted. Furthermore, altered expression levels of these exosomal miRNAs and genes were validated in plasma exosomes and liver tissue of perimenopausal mice. Results: HOMA-IR in VCD-treated mice was significantly increased, and hepatic glycogen was significantly decreased. Key exosomal miRNAs (miR-17-3p, miR-134-5p, miR-700-5p, and miR-6899-3p) and hepatic genes (G6pdx, Ptpn2, Lepr, Kras, and Braf) may be associated with impaired insulin signaling during perimenopause. Conclusion: The perimenopausal period acts as a potential factor in introducing insulin resistance as evidenced by impaired insulin action and altered expression profiles of exosomal miRNAs and hepatic genes. The present study contributes to the understanding that abnormal cargos carried by plasma exosomes, such as miRNAs, may be related to altered expression of the corresponding genes in the liver and abnormal insulin response.
Collapse
Affiliation(s)
- Yu Yang
- Department of Endocrinology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yu Chen
- Department of Endocrinology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Traditional Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Changju Liu
- Department of Endocrinology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Su Wang
- Department of Endocrinology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yijing Zhao
- Department of Endocrinology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Wen Cao
- Department of Endocrinology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Kun Wang
- Department of Endocrinology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| |
Collapse
|
19
|
Florencio-Silva R, Sasso GRDS, Sasso-Cerri E, Cerri PS, Gil CD, de Jesus Simões M. Relationship between autophagy and NLRP3 inflammasome during articular cartilage degradation in oestrogen-deficient rats with streptozotocin-induced diabetes. Ann Anat 2025; 257:152318. [PMID: 39216675 DOI: 10.1016/j.aanat.2024.152318] [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: 05/14/2024] [Revised: 07/23/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Estrogen deficiency and Diabetes mellitus (DM) cause joint tissue deterioration, although the mechanisms are uncertain. This study evaluated the immunoexpression of autophagy and NLRP3-inflammasome markers, in rat articular cartilage with estrogen deficiency and DM. METHODS Twenty rats were sham-operated (SHAM) or ovariectomized (OVX) and equally allocated into four groups: SHAM and OVX groups administered with vehicle solution; SHAM and OVX groups treated with 60 mg/kg/body weight of streptozotocin, intraperitoneally, to induce DM (SHAM-DM and OVX-DM groups). After seven weeks, the rats were euthanized, and their joint knees were processed for paraffin embedding. Sections were stained with haematoxylin-eosin, toluidine blue, safranin-O/fast-green or subjected to picrosirius-red-polarisation method; immunohistochemistry to detect beclin-1 and microtubule-associated protein 1B-light chain 3 (autophagy markers), NLRP3 and interleukin-1β (IL-1β) (inflammasome activation markers), along with matrix metalloproteinase-9 (MMP-9), Nuclear factor-kappa B (NFκB), and Vascular endothelial growth factor A (VEGF-A) were performed. RESULTS Deterioration of articular cartilage and subchondral bone were greater in SHAM-DM and OVX-DM groups. Higher percentages of immunolabeled chondrocytes to NLRP3, IL-1β, MMP-9, NFκB, and VEGF-A, as well as lower percentages of chondrocytes immunolabeled to autophagy markers, were noticed in estrogen-deficient and diabetic groups. These differences were greater in the OVX-DM group. Percentages of immunolabeled chondrocytes showed negative correlation between autophagy markers v.s IL-1β, NLRP-3, MMP-9, NFκB, and VEGF-A, along with positive correlation between VEGF-A vs. MMP-9, NFκB, IL-1β, and NLRP3, and MMP-9 vs. NFκB. CONCLUSIONS In conclusion, autophagy reduction and NLRP3 inflammasome activation in chondrocytes may be implicated in articular cartilage degradation, under estrogen-deficient and DM conditions. Moreover, the combination of estrogen deficiency and DM may potentiate those effects.
Collapse
Affiliation(s)
- Rinaldo Florencio-Silva
- Universidade Federal de São Paulo - UNIFESP, Escola Paulista de Medicina - EPM, Departamento de Ginecologia, São Paulo, SP, Brazil; Universidade Federal de São Paulo - UNIFESP, Escola Paulista de Medicina - EPM, Departamento de Morfologia e Genética, Disciplina de Histologia e Biologia Estrutural, São Paulo, SP, Brazil.
| | - Gisela Rodrigues da Silva Sasso
- Universidade Federal de São Paulo - UNIFESP, Escola Paulista de Medicina - EPM, Departamento de Morfologia e Genética, Disciplina de Histologia e Biologia Estrutural, São Paulo, SP, Brazil
| | - Estela Sasso-Cerri
- São Paulo State University (UNESP), School of Dentistry, Araraquara - Department of Morphology, Genetics, Orthodontics and Pediatric Dentistry - Laboratory of Histology and Embryology, Araraquara, SP, Brazil
| | - Paulo Sérgio Cerri
- São Paulo State University (UNESP), School of Dentistry, Araraquara - Department of Morphology, Genetics, Orthodontics and Pediatric Dentistry - Laboratory of Histology and Embryology, Araraquara, SP, Brazil
| | - Cristiane Damas Gil
- Universidade Federal de São Paulo - UNIFESP, Escola Paulista de Medicina - EPM, Departamento de Morfologia e Genética, Disciplina de Histologia e Biologia Estrutural, São Paulo, SP, Brazil
| | - Manuel de Jesus Simões
- Universidade Federal de São Paulo - UNIFESP, Escola Paulista de Medicina - EPM, Departamento de Ginecologia, São Paulo, SP, Brazil; Universidade Federal de São Paulo - UNIFESP, Escola Paulista de Medicina - EPM, Departamento de Morfologia e Genética, Disciplina de Histologia e Biologia Estrutural, São Paulo, SP, Brazil
| |
Collapse
|
20
|
Sumida KD, Lordan VM, Donovan CM. Enhanced glucose production in norepinephrine and palmitate stimulated hepatocytes following endurance training. Front Physiol 2024; 15:1514082. [PMID: 39742157 PMCID: PMC11685187 DOI: 10.3389/fphys.2024.1514082] [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: 10/19/2024] [Accepted: 11/29/2024] [Indexed: 01/03/2025] Open
Abstract
Enhanced hepatic gluconeogenesis plays an important role in exercise glucose homeostasis when hepatic glycogen stores are depleted. Livers from trained animals demonstrate greater rates of gluconeogenesis in the presence of elevated substrate with and without hormonal stimulation. Training has been reported to have a particularly profound impact on norepinephrine-stimulated gluconeogenesis, but this was only demonstrated in the presence of other gluconeogenic hormones. Here we reexamine the impact of endurance training on norepinephrine-stimulated gluconeogenesis in the absence of any other hormones. Isolated hepatocytes from trained and untrained rats were incubated in 6 mM lactate with various concentrations of norepinephrine (0 nM-20 nM). Absent norepinephrine, gluconeogenic rates were significantly greater from trained hepatocytes compared to controls (97.2 ± 6.7 vs 57.6 ± 8.7 nmol/mg protein; p < 0.01). In the presence of NE (0.5-20 nM), gluconeogenesis from trained liver cells was significantly greater at all NE concentrations compared to controls. The NE-stimulated increase in gluconeogenesis above basal (0 nM NE) was also greater for trained vs control (36% vs 19%, respectively). Concomitant with the max NE-stimulated increase in gluconeogenesis, lactate uptake was significantly elevated for trained vs. control hepatocytes (307.22 ± 44.5 vs 124.5 ± 23.9 nmol/mg protein; p < 0.01), with lactate uptake quantitatively accounting for the entire increase in gluconeogenesis for trained hepatocytes. Endurance training was also observed to significantly elevate glucose production in presence of 0.6 mM palmitate, both in the absence and presence of NE. These findings confirm that hepatocytes from endurance-trained animals demonstrate enhanced rates of NE-stimulated gluconeogenesis, as well as palmitate-stimulated glucose production.
Collapse
Affiliation(s)
- Ken D. Sumida
- Department of Health Sciences, Chapman University, Orange, CA, United States
| | - Vera M. Lordan
- Department of Biological Sciences, USC Dornsife, University of Southern California, Los Angeles, CA, United States
| | - Casey M. Donovan
- Department of Biological Sciences, USC Dornsife, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
21
|
Zanini BM, de Avila BM, Garcia DN, Hense JD, Veiga GB, Barreto MM, Ashiqueali S, Mason JB, Yadav H, Masternak M, Schneider A. Dynamics of serum exosome microRNA profile altered by chemically induced estropause and rescued by estrogen therapy in female mice. GeroScience 2024; 46:5891-5909. [PMID: 38499957 PMCID: PMC11493931 DOI: 10.1007/s11357-024-01129-9] [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] [Received: 01/10/2024] [Accepted: 03/09/2024] [Indexed: 03/20/2024] Open
Abstract
The decline in the ovarian reserve leads to menopause and reduced serum estrogens. MicroRNAs are small non-coding RNAs, which can regulate gene expression and be secreted by cells and trafficked in serum via exosomes. Serum miRNAs regulate tissue function and disease development. Therefore, the aim of this study was to identify miRNA profiles in serum exosomes of mice induced to estropause and treated with 17β-estradiol (E2). Female mice were divided into three groups including control (CTL), injected with 4-Vinylcyclohexene diepoxide (VCD), and injected with VCD plus E2 (VCD + E2). Estropause was confirmed by acyclicity and a significant reduction in the number of ovarian follicles (p < 0.05). Body mass gain during estropause was higher in VCD and VCD + E2 compared to CTL females (p = 0.02). Sequencing of miRNAs was performed from exosomes extracted from serum, and 402 miRNAs were detected. Eight miRNAs were differentially regulated between CTL and VCD groups, seven miRNAs regulated between CTL and VCD + E2 groups, and ten miRNAs regulated between VCD and VCD + E2 groups. Only miR-200a-3p and miR-200b-3p were up-regulated in both serum exosomes and ovarian tissue in both VCD groups, suggesting that these exosomal miRNAs could be associated with ovarian activity. In the hepatic tissue, only miR-370-3p (p = 0.02) was up-regulated in the VCD + E2 group, as observed in serum. Our results suggest that VCD-induced estropause and E2 replacement have an impact on the profile of serum exosomal miRNAs. The miR-200 family was increased in serum exosomes and ovarian tissue and may be a candidate biomarker of ovarian function.
Collapse
Affiliation(s)
| | | | | | - Jéssica Damé Hense
- Faculdade de Nutrição, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | | | | | - Sarah Ashiqueali
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, USA
| | - Jeffrey B Mason
- College of Veterinary Medicine, Department of Veterinary Clinical and Life Sciences, Center for Integrated BioSystems, Utah State University, Logan, UT, USA
| | - Hariom Yadav
- USF Center for Microbiome Research, and Department of Neurosurgery and Brain Repair, Microbiomes Institute, University of South Florida, Tampa, FL, USA
| | - Michal Masternak
- Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, Orlando, FL, USA
- Department of Head and Neck Surgery, Poznan University of Medical Sciences, Poznan, Poland
| | - Augusto Schneider
- Faculdade de Nutrição, Universidade Federal de Pelotas, Pelotas, RS, Brazil.
| |
Collapse
|
22
|
Bardhi O, Dubey P, Palmer BF, Clegg DJ. Oestrogens, adipose tissues and environmental exposures influence obesity and diabetes across the lifecycle. Proc Nutr Soc 2024; 83:263-270. [PMID: 38305136 DOI: 10.1017/s0029665124000119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2024]
Abstract
Endogenous oestrogens regulate essential functions to include menstrual cycles, energy balance, adipose tissue distribution, pancreatic β-cell function, insulin sensitivity and lipid homeostasis. Oestrogens are a family of hormones which include oestradiol (E2), oestrone (E1) and oestriol (E3). Oestrogens function by binding and activating oestrogen receptors (ERs). Phytoestrogens are plant-derived compounds which exhibit oestrogenic-like activity and can bind to ERs. Phytoestrogens exert potential oestrogenic-like benefits; however, their effects are context-dependent and require cautious consideration regarding generalised health benefits. Xenoestrogens are synthetic compounds which have been determined to disrupt endocrine function through binding to ERs. Xenoestrogens enter the body through various routes and given their chemical structure they can accumulate, posing long-term health risks. Xenoestrogens interfere with endogenous oestrogens and their functions contributing to conditions like cancer, infertility, and metabolic disorders. Understanding the interplay between endogenous and exogenous oestrogens is critical in order to determine their potential health consequences and requires further investigation. This manuscript provides a summary of the role endogenous oestrogens have in regulating metabolic functions. Additionally, we discuss the impact phytoestrogens and synthetic xenoestrogens have on biological systems across various life stages. We highlight their mechanisms of action, potential benefits, risks and discuss the need for further research to bridge gaps in understanding and mitigate exposure-related health risks.
Collapse
Affiliation(s)
- Olgert Bardhi
- Department of Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Pallavi Dubey
- Department of Obstetrics and Gynecology, Paul L Foster School of Medicine, El Paso, TX, USA
| | - Biff Franklin Palmer
- Department of Medicine, Division of Nephrology, University of Texas Southwestern Medical, Center, Dallas, TX, USA
| | - Deborah J Clegg
- Vice President for Research, Texas Tech Health Sciences Center, El Paso, TX, USA
| |
Collapse
|
23
|
Ruisch IH, Widomska J, De Witte W, Mota NR, Fanelli G, Van Gils V, Jansen WJ, Vos SJB, Fóthi A, Barta C, Berkel S, Alam KA, Martinez A, Haavik J, O'Leary A, Slattery D, Sullivan M, Glennon J, Buitelaar JK, Bralten J, Franke B, Poelmans G. Molecular landscape of the overlap between Alzheimer's disease and somatic insulin-related diseases. Alzheimers Res Ther 2024; 16:239. [PMID: 39465382 PMCID: PMC11514822 DOI: 10.1186/s13195-024-01609-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 10/21/2024] [Indexed: 10/29/2024]
Abstract
Alzheimer's disease (AD) is a multifactorial disease with both genetic and environmental factors contributing to its etiology. Previous evidence has implicated disturbed insulin signaling as a key mechanism that plays a role in both neurodegenerative diseases such as AD and comorbid somatic diseases such as diabetes mellitus type 2 (DM2). In this study, we analysed available genome-wide association studies (GWASs) of AD and somatic insulin-related diseases and conditions (SID), i.e., DM2, metabolic syndrome and obesity, to identify genes associated with both AD and SID that could increase our insights into their molecular underpinnings. We then performed functional enrichment analyses of these genes. Subsequently, using (additional) GWAS data, we conducted shared genetic etiology analyses between AD and SID, on the one hand, and blood and cerebrospinal fluid (CSF) metabolite levels on the other hand. Further, integrating all these analysis results with elaborate literature searches, we built a molecular landscape of the overlap between AD and SID. From the landscape, multiple functional themes emerged, including insulin signaling, estrogen signaling, synaptic transmission, lipid metabolism and tau signaling. We also found shared genetic etiologies between AD/SID and the blood/CSF levels of multiple metabolites, pointing towards "energy metabolism" as a key metabolic pathway that is affected in both AD and SID. Lastly, the landscape provided leads for putative novel drug targets for AD (including MARK4, TMEM219, FKBP5, NDUFS3 and IL34) that could be further developed into new AD treatments.
Collapse
Affiliation(s)
- I Hyun Ruisch
- Department of Cognitive Neuroscience, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Joanna Widomska
- Department of Cognitive Neuroscience, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ward De Witte
- Department of Cognitive Neuroscience, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Nina R Mota
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Giuseppe Fanelli
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Veerle Van Gils
- Department of Psychiatry & Neuropsychology, Alzheimer Center Limburg, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Willemijn J Jansen
- Department of Psychiatry & Neuropsychology, Alzheimer Center Limburg, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Stephanie J B Vos
- Department of Psychiatry & Neuropsychology, Alzheimer Center Limburg, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Abel Fóthi
- Department of Molecular Biology, Institute of Biochemistry and Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Csaba Barta
- Department of Molecular Biology, Institute of Biochemistry and Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Simone Berkel
- Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Kazi A Alam
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Aurora Martinez
- Department of Biomedicine, University of Bergen, Bergen, Norway
- K.G. Jebsen Center for Translational Research in Parkinson's Disease, University of Bergen, Neuro-SysMed Center, Department of Neurology, Haukeland University Hospital, Bergen, Norway
| | - Jan Haavik
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Aet O'Leary
- Department of Psychiatry, University Hospital, Frankfurt, Germany
| | - David Slattery
- Department of Psychiatry, Psychosomatics and Psychotherapy, Goethe-Universität, Frankfurt, Germany
| | - Mairéad Sullivan
- Conway Institute of Biomedical and Biomolecular Research, School of Medicine, University College Dublin, Dublin, Ireland
| | - Jeffrey Glennon
- Conway Institute of Biomedical and Biomolecular Research, School of Medicine, University College Dublin, Dublin, Ireland
| | - Jan K Buitelaar
- Department of Cognitive Neuroscience, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Janita Bralten
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Barbara Franke
- Department of Cognitive Neuroscience, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Geert Poelmans
- Department of Cognitive Neuroscience, Radboud University Medical Center, Nijmegen, The Netherlands.
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
24
|
Harvey BJ, Alvarez de la Rosa D. Sex Differences in Kidney Health and Disease. Nephron Clin Pract 2024; 149:77-103. [PMID: 39406203 DOI: 10.1159/000541352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 09/02/2024] [Indexed: 02/25/2025] Open
Abstract
BACKGROUND Sex differences exist in kidney physiology and disease which are underpinned by the biological actions of the sex hormones estrogen, progesterone and testosterone. In this review, we present an up-to-date discussion of the hormonal and molecular signalling pathways implicated in sex differences in kidney health and disease. SUMMARY Estrogen and progesterone have protective effects on renal blood flow, glomerular filtration rate and nephron ion and water reabsorptive processes, whereas testosterone tends to compromise these functions. The biological effects of estrogen appear to be the most important in reinforcing kidney function and protecting against kidney diseases in females. The actions of estrogen are myriad but all tend to bolster kidney physiology to maintain a steady-state and adaptable extracellular fluid volume (ECFV) and blood pressure. Estrogen safeguards ECFV homeostasis by stimulating renal epithelial sodium channel (ENaC) and water channel (AQP2) expression and transport function. Renal maintenance of ECFV within narrow physiological limits is a first-line of defense against hypertension and lowers the risk of cardiovascular disease in women. The estrogenic and XX chromosome basis for a female advantage are evident in a wide range of kidney diseases including acute kidney injury, chronic kidney disease, end-stage kidney disease, diabetic kidney disease, and polycystic kidney disease. The molecular mechanisms involve estrogen regulation of nephron ion and water transport, genetic immunogenic responses, activation of the protective arm of the renin angiotensin-aldosterone system and XX chromosome reinforcement of immune responses. Kidney disease can also predispose patients to cancer and women are protected in renal cancer with lower incidence, morbidity, and mortality than age-matched men with the disease. KEY MESSAGES This review underscores the importance of incorporating sex-specific considerations into clinical practice and basic research to bridge the gap in understanding and addressing biological sex disparities in kidney disease and renal cancer.
Collapse
Affiliation(s)
- Brian J Harvey
- Faculty of Medicine, Royal College of Surgeons in Ireland, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- Centro de Estudios Científicos, Valdivia, Chile
| | - Diego Alvarez de la Rosa
- Departmento de Ciencias Médicas Básicas and Instituto de Tecnologías Biomédicas, Universidad de La Laguna, La Laguna, Spain
| |
Collapse
|
25
|
Zhu H, Xiao H, Li L, Yang M, Lin Y, Zhou J, Zhang X, Zhou Y, Lan X, Liu J, Zeng J, Wang L, Zhong Y, Qian X, Cao Z, Liu P, Mei H, Cai M, Cai X, Tang Z, Hu L, Zhou R, Xu X, Yang H, Wang J, Jin X, Zhou A. Novel insights into the genetic architecture of pregnancy glycemic traits from 14,744 Chinese maternities. CELL GENOMICS 2024; 4:100631. [PMID: 39389014 PMCID: PMC11602577 DOI: 10.1016/j.xgen.2024.100631] [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: 03/08/2023] [Revised: 12/14/2023] [Accepted: 07/17/2024] [Indexed: 10/12/2024]
Abstract
Glycemic traits are critical indicators of maternal and fetal health during pregnancy. We performed genetic analysis for five glycemic traits in 14,744 Chinese pregnant women. Our genome-wide association study identified 25 locus-trait associations, including established links between gestational diabetes mellitus (GDM) and the genes CDKAL1 and MTNR1B. Notably, we discovered a novel association between fasting glucose during pregnancy and the ESR1 gene (estrogen receptor), which was validated by an independent study in pregnant women. The ESR1-GDM link was recently reported by the FinnGen project. Our work enhances the findings in East Asian populations and highlights the need for independent studies. Further analyses, including genetic correlation, Mendelian randomization, and transcriptome-wide association studies, provided genetic insights into the relationship between pregnancy glycemic traits and hypertension. Overall, our findings advance the understanding of genetic architecture of pregnancy glycemic traits, especially in East Asian populations.
Collapse
Affiliation(s)
- Huanhuan Zhu
- BGI Research, Shenzhen 518083, China; BGI Research, Wuhan 430074, China
| | - Han Xiao
- Institute of Maternal and Child Health, Wuhan Children's Hospital (Wuhan Maternal and Child Health Care Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430010, China
| | - Linxuan Li
- BGI Research, Shenzhen 518083, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Meng Yang
- Institute of Maternal and Child Health, Wuhan Children's Hospital (Wuhan Maternal and Child Health Care Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430010, China
| | - Ying Lin
- BGI Research, Shenzhen 518083, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jieqiong Zhou
- Department of Obstetrics, Wuhan Children's Hospital (Wuhan Maternal and Child Health Care Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430010, China
| | - Xinyi Zhang
- BGI Research, Shenzhen 518083, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yan Zhou
- Department of Obstetrics, Wuhan Children's Hospital (Wuhan Maternal and Child Health Care Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430010, China
| | - Xianmei Lan
- BGI Research, Shenzhen 518083, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiuying Liu
- Department of Obstetrics, Wuhan Children's Hospital (Wuhan Maternal and Child Health Care Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430010, China
| | - Jingyu Zeng
- BGI Research, Shenzhen 518083, China; College of Life Sciences, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Lin Wang
- BGI Research, Shenzhen 518083, China
| | - Yuanyuan Zhong
- Department of Obstetrics, Wuhan Children's Hospital (Wuhan Maternal and Child Health Care Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430010, China
| | - Xiaobo Qian
- BGI Research, Shenzhen 518083, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhongqiang Cao
- Institute of Maternal and Child Health, Wuhan Children's Hospital (Wuhan Maternal and Child Health Care Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430010, China
| | | | - Hong Mei
- Institute of Maternal and Child Health, Wuhan Children's Hospital (Wuhan Maternal and Child Health Care Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430010, China
| | | | - Xiaonan Cai
- Institute of Maternal and Child Health, Wuhan Children's Hospital (Wuhan Maternal and Child Health Care Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430010, China
| | | | - Liqin Hu
- Institute of Maternal and Child Health, Wuhan Children's Hospital (Wuhan Maternal and Child Health Care Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430010, China
| | | | - Xun Xu
- BGI Research, Shenzhen 518083, China; Guangdong Provincial Key Laboratory of Genome Read and Write, BGI Research, Shenzhen 518120, China
| | - Huanming Yang
- BGI Research, Shenzhen 518083, China; Guangdong Provincial Academician Workstation of BGI Synthetic Genomics, BGI, Shenzhen 518120, China; James D. Watson Institute of Genome Sciences, Hangzhou 310058, China
| | | | - Xin Jin
- BGI Research, Shenzhen 518083, China; BGI Research, Wuhan 430074, China; The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou 510006, China; Shanxi Medical University-BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan 030001, China; Shenzhen Key Laboratory of Transomics Biotechnologies, BGI Research, Shenzhen 518083, China.
| | - Aifen Zhou
- Institute of Maternal and Child Health, Wuhan Children's Hospital (Wuhan Maternal and Child Health Care Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430010, China; Department of Obstetrics, Wuhan Children's Hospital (Wuhan Maternal and Child Health Care Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430010, China.
| |
Collapse
|
26
|
Dragutinovic B, Moser F, Notbohm HL, Ihalainen JK, Bloch W, Schumann M. Influence of menstrual cycle and oral contraceptive phases on strength performance, neuromuscular fatigue, and perceived exertion. J Appl Physiol (1985) 2024; 137:919-933. [PMID: 39052822 DOI: 10.1152/japplphysiol.00198.2024] [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: 03/20/2024] [Revised: 07/23/2024] [Accepted: 07/23/2024] [Indexed: 07/27/2024] Open
Abstract
The primary aim of the study was to assess differences in strength performance, neuromuscular fatigue, and perceived exertion across phases of the menstrual cycle [MC; early follicular (eFP), late follicular (lFP), and mid-luteal phase (mLP)] and oral contraceptives [OCs; active pill phase (aPP) and nonactive pill phase (nPP)]. The secondary aim was to analyze the influence of fluctuating serum 17β-estradiol and progesterone concentrations on these parameters in naturally menstruating women. Thirty-four women (21 with a natural MC and 13 using OCs) completed three or two experimental sessions, respectively. Mean propulsive velocity (MPVmean) and total number of repetitions (REPtotal) were assessed during a power [3 × 8 at 60% 1RM (one-repetition maximum)] and hypertrophy squat loading (3 sets to failure at 70% 1RM), respectively. Changes in bench press and squat MPV at 60% 1RM in response to the loadings were used as surrogates for nonlocal and local fatigue, respectively. Total blood lactate accumulation (BLAA) and markers of perceived exertion were assessed in each session. No significant differences between any of the MC or OC phases were observed for MPVmean, REPtotal, nonlocal and local fatigue, and markers of perceived exertion (all P > 0.050). A higher intraindividual 17β-estradiol concentration was significantly associated with a lower MPVmean (P = 0.019). BLAA was significantly higher in the lFP than in the mLP (P = 0.019) and negatively associated with the intraindividual progesterone concentration (P = 0.005). Although 17β-estradiol may negatively influence the MPV, it appears that fluctuations of both sex hormones across the MC and OC phases are not prominent enough to induce significant or practically relevant changes in the assessed parameters.NEW & NOTEWORTHY Although a high intraindividual 17β-estradiol concentration was associated with a lower movement velocity, markers of strength performance and surrogates for nonlocal and local fatigue remained unaffected by MC and OC phases. Blood lactate accumulation was significantly reduced in the mLP. Furthermore, our findings suggest that the impact of the MC phases varies greatly among individuals. Individuals with high fluctuations in sex hormone concentrations may experience relevant changes in the assessed parameters.
Collapse
Affiliation(s)
- Boris Dragutinovic
- Department of Molecular and Cellular Sports Medicine, Institute of Cardiovascular Research and Sports Medicine, German Sport University, Cologne, Germany
- The German Research Centre of Elite Sport Cologne, German Sport University Cologne, Cologne, Germany
| | - Franziska Moser
- Department of Molecular and Cellular Sports Medicine, Institute of Cardiovascular Research and Sports Medicine, German Sport University, Cologne, Germany
| | - Hannah L Notbohm
- Department of Molecular and Cellular Sports Medicine, Institute of Cardiovascular Research and Sports Medicine, German Sport University, Cologne, Germany
| | - Johanna K Ihalainen
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
- Finnish Institute of High Performance Sport KIHU, Jyväskylä, Finland
| | - Wilhelm Bloch
- Department of Molecular and Cellular Sports Medicine, Institute of Cardiovascular Research and Sports Medicine, German Sport University, Cologne, Germany
| | - Moritz Schumann
- Department of Sports Medicine and Exercise Therapy, Chemnitz University of Technology, Chemnitz, Germany
| |
Collapse
|
27
|
Mentzinger J, Teixeira GF, Monnerat JADS, Velasco LL, Lucchetti BB, Martins MAC, Costa V, Andrade GPD, Magliano DC, Rocha HNM, da Nóbrega ACL, Medeiros RF, Rocha NG. Prenatal stress induces sex- and tissue-specific alterations in insulin pathway of Wistar rats offspring. Am J Physiol Heart Circ Physiol 2024; 327:H1055-H1066. [PMID: 39212771 DOI: 10.1152/ajpheart.00243.2024] [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: 04/18/2024] [Revised: 08/07/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND AND AIMS Prenatal stress may lead to tissue and sex-specific cardiometabolic disorders in the offspring through imbalances in the insulin signaling pathway. Therefore, we aimed to determine the sex-specific adaptations of prenatal stress on the insulin signaling pathway of cardiac and hepatic tissue of adult offspring Wistar rats. METHODS Wistar pregnant rats were divided into control and stress groups. Unpredictable stress protocol was performed from the 14th to the 21st day of pregnancy. After lactation, the dams were euthanized and blood was collected for corticosterone measurement and the offspring were separated into four groups according to sex and intervention (n=8/group). At 90 days old, the offspring were submitted to an oral glucose tolerance test (OGTT) and an insulin tolerance test (ITT). After euthanasia blood collection was used for biochemical analysis and the left ventricle and liver were used for protein expression and histological analysis. RESULTS Stress increased maternal corticosterone levels, and in the offspring, decreased glucose concentration in both OGTT and ITT, reduced insulin receptor (Irβ) and insulin receptor substrate-1 (IRS1) activation and reduced insulin receptor inhibition (PTP1B) in the liver of male offspring at 90 days old, without repercussions in cardiac tissue. Moreover, female offspring submitted to prenatal stress exhibited reduced fatty acid uptake, with lower hepatic CD36 expression, reduced high density lipoprotein (cHDL) and increased Castelli risk indexes I and II. CONCLUSIONS Unpredictable prenatal stress evoked reduced insulin sensitivity and liver-specific impairment in insulin signaling activation in male while increasing markers of cardiovascular risk in females.
Collapse
Affiliation(s)
- Juliana Mentzinger
- Department of Physiology and Pharmacology, Universidade Federal Fluminense, Niteroi, Brazil
| | | | | | | | | | | | - Viviane Costa
- Department of Physiology and Pharmacology, Fluminense Federal University, Brazil
| | | | | | | | | | | | | |
Collapse
|
28
|
Jang W, Kim S, Son Y, Kim S, Kim HJ, Jo H, Park J, Lee K, Lee H, Tully MA, Rahmati M, Smith L, Kang J, Woo S, Kim S, Hwang J, Rhee SY, Yon DK. Prevalence, Awareness, Treatment, and Control of Type 2 Diabetes in South Korea (1998 to 2022): Nationwide Cross-Sectional Study. JMIR Public Health Surveill 2024; 10:e59571. [PMID: 39190907 PMCID: PMC11387923 DOI: 10.2196/59571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/07/2024] [Accepted: 06/20/2024] [Indexed: 08/29/2024] Open
Abstract
BACKGROUND Type 2 diabetes poses an increasing disease burden in South Korea. The development and management of type 2 diabetes are closely related to lifestyle and socioeconomic factors, which have undergone substantial changes over the past few decades, including during the COVID-19 pandemic. OBJECTIVE This study aimed to investigate long-term trends in type 2 diabetes prevalence, awareness, treatment, and control. It also aimed to determine whether there were substantial alterations in the trends during the pandemic and whether these changes were more pronounced within specific demographic groups. METHODS This study examined the prevalence, awareness, treatment, and control of type 2 diabetes in a representative sample of 139,786 South Koreans aged >30 years, using data from the National Health and Nutrition Examination Survey and covering the period from 1998 to 2022. Weighted linear regression and binary logistic regression were performed to calculate weighted β coefficients or odds ratios. Stratified analyses were performed based on sex, age, region of residence, obesity status, educational background, household income, and smoking status. β (difference) was calculated to analyze the trend difference between the prepandemic period and the COVID-19 pandemic. To identify groups more susceptible to type 2 diabetes, we estimated interaction terms for each factor and calculated weighted odds ratios. RESULTS From 1998 to 2022, a consistent increase in the prevalence of type 2 diabetes was observed among South Koreans, with a notable rise to 15.61% (95% CI 14.83-16.38) during the pandemic. Awareness followed a U-shaped curve, bottoming out at 64.37% (95% CI 61.79-66.96) from 2013 to 2015 before increasing to 72.56% (95% CI 70.39-74.72) during the pandemic. Treatment also increased over time, peaking at 68.33% (95% CI 65.95-70.71) during the pandemic. Control among participants with diabetes showed no substantial change, maintaining a rate of 29.14% (95% CI 26.82-31.47) from 2020 to 2022, while control among treated participants improved to 30.68% (95% CI 27.88-33.48). During the pandemic, there was a steepening of the curves for awareness and treatment. However, while the slope of control among participants being treated increased, the slope of control among participants with diabetes showed no substantial change during the pandemic. Older populations and individuals with lower educational level exhibited less improvement in awareness and control trends than younger populations and more educated individuals. People with lower income experienced a deceleration in prevalence during the pandemic. CONCLUSIONS Over the recent decade, there has been an increase in type 2 diabetes prevalence, awareness, treatment, and control. During the pandemic, a steeper increase in awareness, treatment, and control among participants being treated was observed. However, there were heterogeneous changes across different population groups, underscoring the need for targeted interventions to address disparities and improve diabetes management for susceptible populations.
Collapse
Affiliation(s)
- Wonwoo Jang
- Department of Medicine, Kyung Hee University College of Medicine, Seoul, Republic of Korea
- Center for Digital Health, Medical Science Research Institute, Kyung Hee University College of Medicine, Seoul, Republic of Korea
| | - Seokjun Kim
- Department of Medicine, Kyung Hee University College of Medicine, Seoul, Republic of Korea
- Center for Digital Health, Medical Science Research Institute, Kyung Hee University College of Medicine, Seoul, Republic of Korea
| | - Yejun Son
- Center for Digital Health, Medical Science Research Institute, Kyung Hee University College of Medicine, Seoul, Republic of Korea
- Department of Precision Medicine, Kyung Hee University College of Medicine, Seoul, Republic of Korea
| | - Soeun Kim
- Center for Digital Health, Medical Science Research Institute, Kyung Hee University College of Medicine, Seoul, Republic of Korea
- Department of Precision Medicine, Kyung Hee University College of Medicine, Seoul, Republic of Korea
| | - Hyeon Jin Kim
- Center for Digital Health, Medical Science Research Institute, Kyung Hee University College of Medicine, Seoul, Republic of Korea
- Department of Regulatory Science, Kyung Hee University, Seoul, Republic of Korea
| | - Hyesu Jo
- Center for Digital Health, Medical Science Research Institute, Kyung Hee University College of Medicine, Seoul, Republic of Korea
- Department of Regulatory Science, Kyung Hee University, Seoul, Republic of Korea
| | - Jaeyu Park
- Center for Digital Health, Medical Science Research Institute, Kyung Hee University College of Medicine, Seoul, Republic of Korea
- Department of Regulatory Science, Kyung Hee University, Seoul, Republic of Korea
| | - Kyeongmin Lee
- Center for Digital Health, Medical Science Research Institute, Kyung Hee University College of Medicine, Seoul, Republic of Korea
- Department of Regulatory Science, Kyung Hee University, Seoul, Republic of Korea
| | - Hayeon Lee
- Center for Digital Health, Medical Science Research Institute, Kyung Hee University College of Medicine, Seoul, Republic of Korea
| | - Mark A Tully
- School of Medicine, Ulster University, Londonderry, United Kingdom
| | - Masoud Rahmati
- CEReSS-Health Service Research and Quality of Life Center, Aix-Marseille University, Marseille, France
- Department of Physical Education and Sport Sciences, Faculty of Literature and Human Sciences, Lorestan University, Khoramabad, Iran
- Department of Physical Education and Sport Sciences, Faculty of Literature and Humanities, Vali-E-Asr University of Rafsanjan, Rafsanjan, Iran
| | - Lee Smith
- Centre for Health, Performance and Wellbeing, Anglia Ruskin University, Cambridge, United Kingdom
| | - Jiseung Kang
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, United States
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, United States
| | - Selin Woo
- Center for Digital Health, Medical Science Research Institute, Kyung Hee University College of Medicine, Seoul, Republic of Korea
| | - Sunyoung Kim
- Department of Family Medicine, Kyung Hee University Medical Center, Kyung Hee University College of Medicine, Seoul, Republic of Korea
| | - Jiyoung Hwang
- Center for Digital Health, Medical Science Research Institute, Kyung Hee University College of Medicine, Seoul, Republic of Korea
| | - Sang Youl Rhee
- Department of Medicine, Kyung Hee University College of Medicine, Seoul, Republic of Korea
- Center for Digital Health, Medical Science Research Institute, Kyung Hee University College of Medicine, Seoul, Republic of Korea
- Department of Precision Medicine, Kyung Hee University College of Medicine, Seoul, Republic of Korea
- Department of Regulatory Science, Kyung Hee University, Seoul, Republic of Korea
- Department of Endocrinology and Metabolism, Kyung Hee University School of Medicine, Seoul, Republic of Korea
| | - Dong Keon Yon
- Department of Medicine, Kyung Hee University College of Medicine, Seoul, Republic of Korea
- Center for Digital Health, Medical Science Research Institute, Kyung Hee University College of Medicine, Seoul, Republic of Korea
- Department of Precision Medicine, Kyung Hee University College of Medicine, Seoul, Republic of Korea
- Department of Regulatory Science, Kyung Hee University, Seoul, Republic of Korea
- Department of Pediatrics, Kyung Hee University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
29
|
Tian X, Li C, Li T, Yu F, Shi R. Estrogen Status Influences Whole-Body Vibration Training-Induced Improvements on Muscle Mass and Strength in Female Ovariectomized Mice. Int J Med Sci 2024; 21:2149-2157. [PMID: 39239545 PMCID: PMC11373558 DOI: 10.7150/ijms.97770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 07/11/2024] [Indexed: 09/07/2024] Open
Abstract
Estradiol (E2) deficiency arising from menopause is closely related to changes in body composition and declines of muscle mass and strength in elderly women. Whole-body vibration training (WBV) is an emerging approach expected to improve muscle mass and strength of older person, but the underlying mechanisms remain unclear. The balance between protein synthesis and degradation is a determining factor for muscle mass and strength, which is regulated by Akt-mTOR and FoxO1 signal pathway, respectively. In the present study, we firstly determined whether the effects of WBV on muscle mass and strength in ovariectomized female mice was affected by estrogen level, then investigated whether this was associated with Akt-mTOR and FoxO1 signal pathways. We found that (1) WBV, E2 supplementation (E) and WBV combined with E2 supplementation (WBV+E) significantly increased serum estradiol content, quadriceps muscle mass and grip strength in ovariectomized mice, accompanied with alterations of body composition (reducing fat content, increasing lean body mass and lean percent), furthermore, the altered degrees of these indicators by WBV+E were greater than WBV alone; (2) WBV, E and WBV+E remarkably increased the activities of Akt and mTOR and decreased FoxO1 activity, and the changed degrees by WBV+E were greater than WBV alone; (3) Pearson correlation coefficient revealed that serum estradiol content was positively correlated with Akt and mTOR activities, while inversely associated with FoxO1 activity. We concluded that WBV could significantly increase muscle mass and strength in ovariectomized mice, which might achieve through activating Akt-mTOR and suppressing FoxO1 signal pathways, and the improving effect of WBV on muscle mass and strength was better when in the presence of estrogen.
Collapse
Affiliation(s)
- Xiangyang Tian
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Cong Li
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Tao Li
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Fangfang Yu
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Rengfei Shi
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| |
Collapse
|
30
|
Li X, Yang J, Blockeel C, Lin M, Tian X, Wu H, Cao Y, Deng L, Zhou X, Xie J, Hu Y, Chen X. Association of severity of menstrual dysfunction with cardiometabolic risk markers among women with polycystic ovary syndrome. Acta Obstet Gynecol Scand 2024; 103:1606-1614. [PMID: 38715377 PMCID: PMC11266638 DOI: 10.1111/aogs.14863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 02/23/2024] [Accepted: 04/13/2024] [Indexed: 07/25/2024]
Abstract
INTRODUCTION Polycystic ovary syndrome (PCOS) is associated with a wide range of unfavorable cardiometabolic risk factors, including obesity, hypertension, insulin resistance, impaired glucose metabolism, dyslipidemia, and metabolic syndrome. Compared with women with regular menstrual cycles, women with a history of irregular menstrual periods have an increased unfavorable cardiometabolic risk. Recently, the association between the severity of oligomenorrhea and hyperinsulinemia and insulin resistance has been demonstrated. However, evidence linking the severity of menstrual cyclicity with cardiometabolic risk in PCOS women is scarce. MATERIAL AND METHODS This work was a prospective cross-sectional study. A total of 154 women diagnosed with PCOS by the Rotterdam criteria were recruited from July 2021 to September 2022. PCOS women with eumenorrheic (eumeno group), oligomenorrhea (oligo group), and amenorrhea (ameno group) underwent history and physical examination, gonadal steroid hormone measurement, lipid profile, oral glucose tolerance test, and homeostasis model assessment of insulin resistance. RESULTS A trend toward an increase in unfavorable cardiometabolic risk markers including obesity, hypertension, prevalence of insulin resistance, prediabetes, dyslipidemia, and metabolic syndrome was observed in the ameno group (n = 57) as compared with the eumeno (n = 24) or oligo group (n = 73). A higher prevalence of insulin resistance (odds ratio [OR]: 3.02; 95% confidence interval [CI]: 1.03-8.81) and prediabetes (OR: 3.94; 95% CI: 1.01-15.40) was observed in the ameno group than in the eumeno group, and a higher proportion of dyslipidemia (OR: 2.44; 95% CI: 1.16-5.15) was observed in the ameno group than in the oligo group in the binary logistic regression analysis after adjusting for confounding factors. CONCLUSIONS PCOS women with amenorrhea show a higher prevalence of insulin resistance, prediabetes, and dyslipidemia compared with those with oligomenorrhea or eumenorrhea. The severity of menstrual dysfunction could be used as a readily obtainable marker for the identification of PCOS women at greatest risk of cardiometabolic diseases.
Collapse
Affiliation(s)
- Xuelan Li
- Reproductive Medicine CenterShunde Hospital of Southern Medical University (The First People's Hospital of Shunde)FoshanGuangdongChina
| | - Jie Yang
- Reproductive Medicine CenterShunde Hospital of Southern Medical University (The First People's Hospital of Shunde)FoshanGuangdongChina
| | - Christophe Blockeel
- Brussels, IVF, center for Reproductive MedicineUniversitair Ziekenhuis Brussel, Vrije Universiteit BrusselBrusselsBelgium
| | - Min Lin
- Reproductive Medicine CenterShunde Hospital of Southern Medical University (The First People's Hospital of Shunde)FoshanGuangdongChina
| | - Xiaoyan Tian
- Reproductive Medicine CenterShunde Hospital of Southern Medical University (The First People's Hospital of Shunde)FoshanGuangdongChina
| | - Haocun Wu
- Department of Clinical LaboratoryShunde Hospital of Southern Medical University (The First People's Hospital of Shunde)FoshanGuangdongChina
| | - Yaqi Cao
- College of ScienceMinzu University of ChinaBeijingChina
| | - Ling Deng
- Reproductive Medicine CenterShunde Hospital of Southern Medical University (The First People's Hospital of Shunde)FoshanGuangdongChina
| | - Xianli Zhou
- Reproductive Medicine CenterShunde Hospital of Southern Medical University (The First People's Hospital of Shunde)FoshanGuangdongChina
| | - Jinying Xie
- College of ScienceMinzu University of ChinaBeijingChina
| | - Yunzhao Hu
- Department of CardiologyShunde Hospital of Southern Medical University (The First People's Hospital of Shunde)FoshanGuangdongChina
| | - Xin Chen
- Reproductive Medicine CenterShunde Hospital of Southern Medical University (The First People's Hospital of Shunde)FoshanGuangdongChina
| |
Collapse
|
31
|
Xega V, Liu JL. Beyond reproduction: unraveling the impact of sex hormones on cardiometabolic health. MEDICAL REVIEW (2021) 2024; 4:284-300. [PMID: 39135604 PMCID: PMC11317208 DOI: 10.1515/mr-2024-0012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 05/07/2024] [Indexed: 08/15/2024]
Abstract
This review thoroughly explores the multifaceted roles of sexual hormones, emphasizing their impact beyond reproductive functions and underscoring their significant influence on cardiometabolic regulation. It analyzes the broader physiological implications of estrogen, testosterone, and progesterone, highlighting their effects on metabolic syndrome, lipid metabolism, glucose homeostasis, and cardiovascular health. Drawing from diverse molecular, clinical, and therapeutic studies, the paper delves into the intricate interplay between these hormones and cardiometabolic processes. By presenting a comprehensive analysis that goes beyond traditional perspectives, and recognizing sexual hormones as more than reproductive agents, the review sheds light on their broader significance in health and disease management, advocating for holistic and personalized medical approaches.
Collapse
Affiliation(s)
- Viktoria Xega
- MeDiC Program, The Research Institute of McGill University Health Centre, Montreal, Canada
| | - Jun-Li Liu
- Division of Endocrinology and Metabolism, Department of Medicine, McGill University, Montreal, Canada
| |
Collapse
|
32
|
Rodriguez de Morales YA, Abramson BL. Cardiovascular and physiological risk factors in women at mid-life and beyond. Can J Physiol Pharmacol 2024; 102:442-451. [PMID: 38739947 DOI: 10.1139/cjpp-2023-0468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Cardiovascular disease (CVD) is the leading cause of death in women. After menopause, sex-specific and gender-specific factors may play an important role in increasing CVD risk, with changes in sex hormones, body fat distribution, lipid and metabolic profile, and structural and functional vascular modifications. Premature and early-onset menopause are detrimental to cardiovascular health due to the early cessation of the protective effect of endogenous estrogen. An independent association of menopause with an increased risk of CVD has been documented in early menopause (<45 years). Sex-related differences are relevant in pharmacokinetics and pharmacodynamics; different enzyme formations, drug compatibility, efficacy, and side effects vary for different sexes. Despite some progress in sex and gender research in CVD, disparities remain. Menopausal hormone therapy (MHT) is available at mid-life for symptoms of menopause and may impact cardiovascular risk. Taken early, MHT may reduce CVD morbimortality. However, this is balanced against the risk of increased thrombosis. This paper reviews physiologic changes that contribute to cardiovascular risk in postmenopausal women and discusses clinical implications. Specifically, it explores the atheroprotective effects of estrogen and MHT and the associations between menopause with lipid levels, hypertension, body composition, and diabetes for women at mid-life and beyond.
Collapse
Affiliation(s)
- Yenny A Rodriguez de Morales
- Fellow Cardiac Prevention and Ambulatory Care, St. Michael's Hospital, University of Toronto, Toronto, ON, Canada
| | - Beth L Abramson
- St. Michael's Hospital, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
33
|
Yang W, Arora M, Han HW, Jiang W, Kim DM, Ai W, Pan Q, Kumar MNVR, Brashear WA, Sun Y, Guo S. ZnPP-laden nanoparticles improve glucose homeostasis and chronic inflammation during obesity. Br J Pharmacol 2024; 181:2886-2904. [PMID: 38679457 DOI: 10.1111/bph.16356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 01/16/2024] [Accepted: 02/09/2024] [Indexed: 05/01/2024] Open
Abstract
BACKGROUND AND PURPOSE Chronic inflammation plays a pivotal role in the development of Type 2 diabetes mellitus (T2DM). Previous studies have shown that haem oxygenase-1 (HO-1) plays a proinflammatory role during metabolic stress, suggesting that HO-1 inhibition could be an effective strategy to treat T2DM. However, the application of HO-1 inhibitors is restricted due to solubility-limited bioavailability. In this study, we encapsulated the HO-1 inhibitor, zinc protoporphyrin IX (ZnPP), within nanoparticles and investigated their role in regulating glucose homeostasis and chronic inflammation during obesity. EXPERIMENTAL APPROACH We delivered DMSO-dissolved ZnPP (DMSO-ZnPP) and ZnPP-laden nanoparticles (Nano-ZnPP) to diet-induced obese male mice for 6 weeks. Glucose and insulin tolerance tests were carried out, liver and adipose tissue gene expression profiles analysed, and systemic inflammation analysed using flow cytometry. KEY RESULTS Nanoparticles significantly increased the delivery efficiency of ZnPP in both cells and mice. In mice with diet-induced obesity, inhibition of HO-1 by Nano-ZnPP significantly decreased adiposity, increased insulin sensitivity, and improved glucose tolerance. Moreover, Nano-ZnPP treatment attenuated both local and systemic inflammatory levels during obesity. Mechanistically, Nano-ZnPP significantly attenuated glucagon, TNF, and fatty acid synthesis signalling pathways in the liver. In white adipose tissue, the oxidative phosphorylation signalling pathway was enhanced and the inflammation signalling pathway diminished by Nano-ZnPP. Our results show that Nano-ZnPP has better effects on the improvement of glucose homeostasis and attenuation of chronic inflammation, than those of DMSO-dissolved ZnPP. CONCLUSIONS AND IMPLICATIONS These findings indicate that ZnPP-laden nanoparticles are potential therapeutic agents for treating T2DM.
Collapse
Affiliation(s)
- Wanbao Yang
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas, USA
| | - Meenakshi Arora
- College of Community Health Sciences, The University of Alabama, Tuscaloosa, Alabama, USA
- The Center for Convergent Bioscience and Medicine (CCBM), The University of Alabama, Tuscaloosa, Alabama, USA
| | - Hye Won Han
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas, USA
| | - Wen Jiang
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas, USA
| | - Da Mi Kim
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas, USA
| | - Weiqi Ai
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas, USA
| | - Quan Pan
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas, USA
| | - M N V Ravi Kumar
- College of Community Health Sciences, The University of Alabama, Tuscaloosa, Alabama, USA
- The Center for Convergent Bioscience and Medicine (CCBM), The University of Alabama, Tuscaloosa, Alabama, USA
| | - Wesley A Brashear
- High Performance Research Computing, Texas A&M University, College Station, Texas, USA
| | - Yuxiang Sun
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas, USA
| | - Shaodong Guo
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
34
|
Xiang X, Palasuberniam P, Pare R. The Role of Estrogen across Multiple Disease Mechanisms. Curr Issues Mol Biol 2024; 46:8170-8196. [PMID: 39194700 DOI: 10.3390/cimb46080483] [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: 06/13/2024] [Revised: 07/18/2024] [Accepted: 07/24/2024] [Indexed: 08/29/2024] Open
Abstract
Estrogen is a significant hormone that is involved in a multitude of physiological and pathological processes. In addition to its pivotal role in the reproductive system, estrogen is also implicated in the pathogenesis of a multitude of diseases. Nevertheless, previous research on the role of estrogen in a multitude of diseases, including Alzheimer's disease, depression, cardiovascular disease, diabetes, osteoporosis, gastrointestinal diseases, and estrogen-dependent cancers, has concentrated on a single disease area, resulting in a lack of comprehensive understanding of cross-disease mechanisms. This has brought some challenges to the current treatment methods for these diseases, because estrogen as a potential therapeutic tool has not yet fully developed its potential. Therefore, this review aims to comprehensively explore the mechanism of estrogen in these seven types of diseases. The objective of this study is to describe the relationship between each disease and estrogen, including the ways in which estrogen participates in regulating disease mechanisms, and to outline the efficacy of estrogen in treating these diseases in clinical practice. By studying the role of estrogen in a variety of disease mechanisms, it is hoped that a more accurate theoretical basis and clinical guidance for future treatment strategies will be provided, thus promoting the effective management and treatment of these diseases.
Collapse
Affiliation(s)
- Xiuting Xiang
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu 88400, Malaysia
| | - Praneetha Palasuberniam
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu 88400, Malaysia
| | - Rahmawati Pare
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu 88400, Malaysia
| |
Collapse
|
35
|
Parkin RA, Murray AJ. The therapeutic potential of irisin to mitigate the risk of metabolic syndrome in postmenopausal women. FRONTIERS IN REPRODUCTIVE HEALTH 2024; 6:1355922. [PMID: 39040132 PMCID: PMC11260725 DOI: 10.3389/frph.2024.1355922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 06/03/2024] [Indexed: 07/24/2024] Open
Abstract
Oestradiol withdrawal at menopause predisposes women to metabolic syndrome, a cluster of interrelated conditions including obesity, insulin resistance, dyslipidaemia and hypertension that together confer an increased risk of developing type 2 diabetes mellitus and cardiovascular disease. Hormone replacement therapies are commonly used to treat acute symptoms of the perimenopausal period, and whilst they have been associated with metabolic improvements in many studies, long-term use is considered unviable. Novel approaches are required to mitigate the risk of postmenopausal metabolic syndrome. In 2012, the exercise-inducible myokine irisin was isolated from the skeletal muscle of mice and identified to have anti-obesity and antidiabetic effects in vivo. Irisin is now recognised to exert pleiotropic action on cognitive, bone and metabolic health. There is accumulating evidence from in vitro and in vivo rodent studies that irisin can mitigate each component condition of metabolic syndrome. In postmenopausal women, independent associations have been observed between (a) exercise and plasma irisin concentration and (b) plasma irisin concentration and reduced incidence of metabolic syndrome. To date, however, no study has considered the mechanistic basis by which irisin, whether exercise-induced or exogenously administered, could reduce the incidence or severity of metabolic syndrome in postmenopausal women. This review aims to analyse the literature concerning the metabolic actions of irisin, with a focus on its therapeutic potential for metabolic syndrome driven by a state of oestradiol depletion. It evaluates the practicality of exercise as a therapy and discusses other irisin-based therapeutic strategies that may alleviate postmenopausal metabolic syndrome. Finally, it highlights areas where future research is required to advance knowledge of irisin's biological action such that it could be considered a viable candidate for clinical application.
Collapse
Affiliation(s)
| | - Andrew J. Murray
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
36
|
Liberty IA, Septadina IS, Mariana, Novita E, Amalia R, Ananingsih ES, Hasyim H, Hanifah L. The characteristics and risk of obesity central and concomitant impaired fasting glucose: Findings from a cross-sectional study. PLoS One 2024; 19:e0305604. [PMID: 38917085 PMCID: PMC11198825 DOI: 10.1371/journal.pone.0305604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 06/03/2024] [Indexed: 06/27/2024] Open
Abstract
INTRODUCTION Obesity is associated with concomitant chronic conditions. An early metabolic consequence of obesity is disruption of glucose and insulin homeostasis. One of the consequences is impaired fasting glucose (IFG). Visceral fat is metabolically more harmful than subcutaneous fat, but few information is available regarding the association between the risk of abnormal glucose in increased waist circumference. METHODS This study is based on a cross sectional of 1,381 population-based from Palembang, Indonesia. The eligibility requirements subject were to be older than 18 and consent to taking fasting glucose and lipid profile tests as well as physical exams measuring their body weight, height, blood pressure, abdominal circumference, and waist circumference. RESULTS The number of subjects consisting of 798 noncentral obesity with normoglycemia, 376 central obesity with normoglycemia, and 207 central obesity with concomitant IFG. The prevalence central obesity with concomitant IFG was 35.51%. In subjects with central obesity, there were significant differences in proportions based on sex, age, marital status, education, and occupation. In multivariate analysis show that the risk factors that contribute to having a significant association with central obesity with concomitant IFG are sex (female), age (>40 years), blood pressure (hypertension), and HDL-C <50 mg/dL (p<0.001). The analysis also founded that there was a significant difference in the dietary pattern of sweet foods (p = 0.018), sweet drinks (p = 0.002), soft drinks (p = 0.001) and smoking habit (p<0.001) between subjects with obesity central and concomitant IFG compared to subjects with noncentral obesity. The majority of subjects with obesity central and concomitant IFG had consuming these risky foods >6 times/week. CONCLUSION The prevalence of central obesity with IFG is quite high. There are significant differences in the characteristics, lipid profile, blood pressure, dietary pattern, and smoking habit of central obesity with concomitant IFG was confirmed in this population-based observational study.
Collapse
Affiliation(s)
- Iche Andriyani Liberty
- Department of Public Health and Community Medicine, Faculty of Medicine, Universitas Sriwijaya, Palembang, Indonesia
| | - Indri Seta Septadina
- Department of Anatomy, Faculty of Medicine, Universitas Sriwijaya, Palembang, Indonesia
| | - Mariana
- Department of Public Health and Community Medicine, Faculty of Medicine, Universitas Sriwijaya, Palembang, Indonesia
| | - Emma Novita
- Department of Anatomy, Faculty of Medicine, Universitas Sriwijaya, Palembang, Indonesia
| | - Resi Amalia
- Faculty of Medicine, Universitas Muhammadiyah, Palembang, Indonesia
| | | | - Hamzah Hasyim
- Faculty of Public Health, Universitas Sriwijaya, Palembang, Indonesia
| | - Laily Hanifah
- Universitas Pembangunan Nasional Veteran Jakarta, Jakarta, Indonesia
| |
Collapse
|
37
|
Ojaghi M, Pamenter ME. Hypoxia impairs blood glucose homeostasis in naked mole-rat adult subordinates but not queens. J Exp Biol 2024; 227:jeb247537. [PMID: 38680085 PMCID: PMC11166464 DOI: 10.1242/jeb.247537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 04/12/2024] [Indexed: 05/01/2024]
Abstract
Naked mole-rats (NMRs) are among the most hypoxia-tolerant mammals and metabolize only carbohydrates in hypoxia. Glucose is the primary building block of dietary carbohydrates, but how blood glucose is regulated during hypoxia has not been explored in NMRs. We hypothesized that NMRs mobilize glucose stores to support anaerobic energy metabolism in hypoxia. To test this, we treated newborn, juvenile and adult (subordinate and queen) NMRs in normoxia (21% O2) or hypoxia (7, 5 or 3% O2), while measuring metabolic rate, body temperature and blood [glucose]. We also challenged animals with glucose, insulin or insulin-like growth factor-1 (IGF-1) injections and measured the rate of glucose clearance in normoxia and hypoxia. We found that: (1) blood [glucose] increases in moderate hypoxia in queens and pups, but only in severe hypoxia in adult subordinates and juveniles; (2) glucose tolerance is similar between developmental stages in normoxia, but glucose clearance times are 2- to 3-fold longer in juveniles and subordinates than in queens or pups in hypoxia; and (3) reoxygenation accelerates glucose clearance in hypoxic subordinate adults. Mechanistically, (4) insulin and IGF-1 reduce blood [glucose] in subordinates in both normoxia but only IGF-1 impacts blood [glucose] in hypoxic queens. Our results indicate that insulin signaling is impaired by hypoxia in NMRs, but that queens utilize IGF-1 to overcome this limitation and effectively regulate blood glucose in hypoxia. This suggests that sexual maturation impacts blood glucose handling in hypoxic NMR queens, which may allow queens to spend longer periods of time in hypoxic nest chambers.
Collapse
Affiliation(s)
- Mohammad Ojaghi
- Department of Biology, University of Ottawa, Ottawa, ON, Canada, K1N 9A7
| | - Matthew E. Pamenter
- Department of Biology, University of Ottawa, Ottawa, ON, Canada, K1N 9A7
- University of Ottawa Brain and Mind Research Institute, Ottawa, ON, Canada, K1H 8M5
| |
Collapse
|
38
|
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
|
39
|
Liu B, Wei Y, He J, Feng B, Chen Y, Guo R, Griffin MD, Hynes SO, Shen S, Liu Y, Cui H, Ma J, O'Brien T. Human umbilical cord-derived mesenchymal stromal cells improve myocardial fibrosis and restore miRNA-133a expression in diabetic cardiomyopathy. Stem Cell Res Ther 2024; 15:120. [PMID: 38659015 PMCID: PMC11040946 DOI: 10.1186/s13287-024-03715-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 04/02/2024] [Indexed: 04/26/2024] Open
Abstract
BACKGROUND Diabetic cardiomyopathy (DCM) is a serious health-threatening complication of diabetes mellitus characterized by myocardial fibrosis and abnormal cardiac function. Human umbilical cord mesenchymal stromal cells (hUC-MSCs) are a potential therapeutic tool for DCM and myocardial fibrosis via mechanisms such as the regulation of microRNA (miRNA) expression and inflammation. It remains unclear, however, whether hUC-MSC therapy has beneficial effects on cardiac function following different durations of diabetes and which mechanistic aspects of DCM are modulated by hUC-MSC administration at different stages of its development. This study aimed to investigate the therapeutic effects of intravenous administration of hUC-MSCs on DCM following different durations of hyperglycemia in an experimental male model of diabetes and to determine the effects on expression of candidate miRNAs, target mRNA and inflammatory mediators. METHODS A male mouse model of diabetes was induced by multiple low-dose streptozotocin injections. The effects on severity of DCM of intravenous injections of hUC-MSCs and saline two weeks previously were compared at 10 and 18 weeks after diabetes induction. At both time-points, biochemical assays, echocardiography, histopathology, polymerase chain reaction (PCR), immunohistochemistry and enzyme-linked immunosorbent assays (ELISA) were used to analyze blood glucose, body weight, cardiac structure and function, degree of myocardial fibrosis and expression of fibrosis-related mRNA, miRNA and inflammatory mediators. RESULTS Saline-treated diabetic male mice had impaired cardiac function and increased cardiac fibrosis after 10 and 18 weeks of diabetes. At both time-points, cardiac dysfunction and fibrosis were improved in hUC-MSC-treated mice. Pro-fibrotic indicators (α-SMA, collagen I, collagen III, Smad3, Smad4) were reduced and anti-fibrotic mediators (FGF-1, miRNA-133a) were increased in hearts of diabetic animals receiving hUC-MSCs compared to saline. Increased blood levels of pro-inflammatory cytokines (IL-6, TNF, IL-1β) and increased cardiac expression of IL-6 were also observed in saline-treated mice and were reduced by hUC-MSCs at both time-points, but to a lesser degree at 18 weeks. CONCLUSION Intravenous injection of hUC-MSCs ameliorated key functional and structural features of DCM in male mice with diabetes of shorter and longer duration. Mechanistically, these effects were associated with restoration of intra-myocardial expression of miRNA-133a and its target mRNA COL1AI as well as suppression of systemic and localized inflammatory mediators.
Collapse
Affiliation(s)
- Boxin Liu
- Stem Cell Research Center, Hebei Medical University-University of Galway, Hebei Medical University, Hebei Province, 050017, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Hebei Province, 050017, China
- Hebei Technology Innovation Center for Stem Cell and Regenerative Medicine, Hebei Province, China
- Hebei International Joint Research Center for Stem Cell and Regenerative Medicine, Hebei Province, China
| | - Yan Wei
- Stem Cell Research Center, Hebei Medical University-University of Galway, Hebei Medical University, Hebei Province, 050017, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Hebei Province, 050017, China
- Hebei Technology Innovation Center for Stem Cell and Regenerative Medicine, Hebei Province, China
- Hebei International Joint Research Center for Stem Cell and Regenerative Medicine, Hebei Province, China
| | - Jingjing He
- Stem Cell Research Center, Hebei Medical University-University of Galway, Hebei Medical University, Hebei Province, 050017, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Hebei Province, 050017, China
- Hebei Technology Innovation Center for Stem Cell and Regenerative Medicine, Hebei Province, China
- Hebei International Joint Research Center for Stem Cell and Regenerative Medicine, Hebei Province, China
| | - Baofeng Feng
- Stem Cell Research Center, Hebei Medical University-University of Galway, Hebei Medical University, Hebei Province, 050017, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Hebei Province, 050017, China
- Human Anatomy Department, Hebei Medical University, Hebei Province, 050017, China
- Hebei Technology Innovation Center for Stem Cell and Regenerative Medicine, Hebei Province, China
- Hebei International Joint Research Center for Stem Cell and Regenerative Medicine, Hebei Province, China
| | - Yimeng Chen
- Stem Cell Research Center, Hebei Medical University-University of Galway, Hebei Medical University, Hebei Province, 050017, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Hebei Province, 050017, China
- Hebei Technology Innovation Center for Stem Cell and Regenerative Medicine, Hebei Province, China
- Hebei International Joint Research Center for Stem Cell and Regenerative Medicine, Hebei Province, China
| | - Ruiyun Guo
- Stem Cell Research Center, Hebei Medical University-University of Galway, Hebei Medical University, Hebei Province, 050017, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Hebei Province, 050017, China
- Hebei Technology Innovation Center for Stem Cell and Regenerative Medicine, Hebei Province, China
- Hebei International Joint Research Center for Stem Cell and Regenerative Medicine, Hebei Province, China
| | - Matthew D Griffin
- Stem Cell Research Center, Hebei Medical University-University of Galway, Hebei Medical University, Hebei Province, 050017, China
- Regenerative Medicine Institute (REMEDI) at CÚRAM SFI Research Centre for Medical Devices, School of Medicine, University of Galway, Galway, Ireland
| | - Seán O Hynes
- Discipline of Pathology, School of Medicine, University of Galway, Galway, Ireland
| | - Sanbing Shen
- Stem Cell Research Center, Hebei Medical University-University of Galway, Hebei Medical University, Hebei Province, 050017, China
- Regenerative Medicine Institute (REMEDI) at CÚRAM SFI Research Centre for Medical Devices, School of Medicine, University of Galway, Galway, Ireland
| | - Yan Liu
- Department of Endocrinology, Hebei Medical University Third Affiliated Hospital, Shijiazhuang, Hebei, 050051, China
| | - Huixian Cui
- Stem Cell Research Center, Hebei Medical University-University of Galway, Hebei Medical University, Hebei Province, 050017, China.
- Hebei Research Center for Stem Cell Medical Translational Engineering, Hebei Province, 050017, China.
- Human Anatomy Department, Hebei Medical University, Hebei Province, 050017, China.
- Hebei Technology Innovation Center for Stem Cell and Regenerative Medicine, Hebei Province, China.
- Hebei International Joint Research Center for Stem Cell and Regenerative Medicine, Hebei Province, China.
| | - Jun Ma
- Stem Cell Research Center, Hebei Medical University-University of Galway, Hebei Medical University, Hebei Province, 050017, China.
- Hebei Research Center for Stem Cell Medical Translational Engineering, Hebei Province, 050017, China.
- Human Anatomy Department, Hebei Medical University, Hebei Province, 050017, China.
- Hebei Technology Innovation Center for Stem Cell and Regenerative Medicine, Hebei Province, China.
- Hebei International Joint Research Center for Stem Cell and Regenerative Medicine, Hebei Province, China.
| | - Timothy O'Brien
- Stem Cell Research Center, Hebei Medical University-University of Galway, Hebei Medical University, Hebei Province, 050017, China.
- Regenerative Medicine Institute (REMEDI) at CÚRAM SFI Research Centre for Medical Devices, School of Medicine, University of Galway, Galway, Ireland.
| |
Collapse
|
40
|
Xu Y, Zhou C, Zong M, Zhu J, Guo X, Sun Z. High-protein high-konjac glucomannan diets changed glucose and lipid metabolism by modulating colonic microflora and bile acid profiles in healthy mouse models. Food Funct 2024; 15:4446-4461. [PMID: 38563504 DOI: 10.1039/d4fo00159a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
High protein and fiber diets are becoming increasingly popular for weight loss; however, the benefits or risks of high protein and fiber diets with a normal calorie level for healthy individuals still need to be elucidated. In this study, we explored the role and mechanisms of long-term high protein and/or konjac glucomannan diets on the metabolic health of healthy mouse models. We found that high konjac glucomannan contents improved the glucose tolerance of mice and both high protein and high konjac glucomannan contents improved the serum lipid profile but increased the TNF-α levels. In the liver, high dietary protein contents reduced the expression of the FASN gene related to fatty acid synthesis. Interactions of dietary protein and fiber were shown in the signaling pathways related to lipid and glucose metabolism of the liver and the inflammatory status of the colon, wherein the high protein and high konjac glucomannan diet downregulated the expression of the SREBF1 and FXR genes in the liver and downregulated the expression of TNF-α genes in the colon compared to the high protein diet. High konjac glucomannan contents reduced the colonic secondary bile acid levels including DCA and LCA; this was largely associated with the changed microbiota profile and also contributed to improved lipid and glucose homeostasis. In conclusion, high protein diets improved lipid homeostasis and were not a risk to metabolic health, while high fiber diets improved glucose and lipid homeostasis by modulating colonic microbiota and bile acid profiles, and a high protein diet supplemented with konjac glucomannan might improve hepatic lipid homeostasis and colonic inflammation in healthy mouse models through long-term intervention.
Collapse
Affiliation(s)
- Yetong Xu
- Laboratory for Bio-Feed and Molecular Nutrition, Department of Animal Science and Technology, Southwest University, Chongqing 400715, P. R. China.
| | - Chengyu Zhou
- Laboratory for Bio-Feed and Molecular Nutrition, Department of Animal Science and Technology, Southwest University, Chongqing 400715, P. R. China.
| | - Minyue Zong
- Laboratory for Bio-Feed and Molecular Nutrition, Department of Animal Science and Technology, Southwest University, Chongqing 400715, P. R. China.
| | - Junwei Zhu
- Laboratory for Bio-Feed and Molecular Nutrition, Department of Animal Science and Technology, Southwest University, Chongqing 400715, P. R. China.
| | - Xutong Guo
- Laboratory for Bio-Feed and Molecular Nutrition, Department of Animal Science and Technology, Southwest University, Chongqing 400715, P. R. China.
| | - Zhihong Sun
- Laboratory for Bio-Feed and Molecular Nutrition, Department of Animal Science and Technology, Southwest University, Chongqing 400715, P. R. China.
| |
Collapse
|
41
|
Yang W, Jiang W, Liao W, Yan H, Ai W, Pan Q, Brashear WA, Xu Y, He L, Guo S. An estrogen receptor α-derived peptide improves glucose homeostasis during obesity. Nat Commun 2024; 15:3410. [PMID: 38649684 PMCID: PMC11035554 DOI: 10.1038/s41467-024-47687-6] [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: 09/25/2023] [Accepted: 04/10/2024] [Indexed: 04/25/2024] Open
Abstract
Estrogen receptor α (ERα) plays a crucial role in regulating glucose and energy homeostasis during type 2 diabetes mellitus (T2DM). However, the underlying mechanisms remain incompletely understood. Here we find a ligand-independent effect of ERα on the regulation of glucose homeostasis. Deficiency of ERα in the liver impairs glucose homeostasis in male, female, and ovariectomized (OVX) female mice. Mechanistic studies reveal that ERα promotes hepatic insulin sensitivity by suppressing ubiquitination-induced IRS1 degradation. The ERα 1-280 domain mediates the ligand-independent effect of ERα on insulin sensitivity. Furthermore, we identify a peptide based on ERα 1-280 domain and find that ERα-derived peptide increases IRS1 stability and enhances insulin sensitivity. Importantly, administration of ERα-derived peptide into obese mice significantly improves glucose homeostasis and serum lipid profiles. These findings pave the way for the therapeutic intervention of T2DM by targeting the ligand-independent effect of ERα and indicate that ERα-derived peptide is a potential insulin sensitizer for the treatment of T2DM.
Collapse
Affiliation(s)
- Wanbao Yang
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX, 77843, USA
| | - Wen Jiang
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX, 77843, USA
| | - Wang Liao
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX, 77843, USA
| | - Hui Yan
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX, 77843, USA
| | - Weiqi Ai
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX, 77843, USA
| | - Quan Pan
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX, 77843, USA
| | - Wesley A Brashear
- High Performance Research Computing, Texas A&M University, College Station, TX, USA
| | - Yong Xu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Ling He
- Departments of Pediatrics and Pharmacology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Shaodong Guo
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX, 77843, USA.
| |
Collapse
|
42
|
Brian MS, Chaudhry BA, D’Amelio M, Waite EE, Dennett JG, O’Neill DF, Feairheller DL. Post-meal exercise under ecological conditions improves post-prandial glucose levels but not 24-hour glucose control. J Sports Sci 2024; 42:728-736. [PMID: 38858835 PMCID: PMC11240283 DOI: 10.1080/02640414.2024.2363688] [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: 11/13/2023] [Accepted: 05/28/2024] [Indexed: 06/12/2024]
Abstract
We investigated whether post-meal walking (PMW) improved post-prandial glucose and 24h glucose control under free-living conditions among physically inactive young women. METHODS Young women (Age: 20±1years; percent body fat: 28.2 ± 12%; BMI: 23.8 ± 4.2kg·m-1) completed a randomised crossover study to assess if PMW confers benefit. On the PMW day, women completed three bouts of brisk walks, and on the Control day they were instructed to follow normal habitual activities. Continuous glucose monitors captured post-prandial and 24h glucose, and physical activity monitors tracked physical activity throughout the study. RESULTS PMW walking increased total daily step count (Control = 9,159 ± 2,962 steps vs. PMW = 14,611±3,891 steps, p<0.001) and activity scores (Control=33.87±1.16 METs·h vs. PMW = 36.11±1.58 METs·h, p < 0.001). PMW led to lower 3h average post-prandial glucose (main effect of condition, p=0.011) and 3h post-prandial area under curve glucose responses (main effect of condition, p = 0.027) compared to the control condition. Post hoc analysis revealed the largest decline occurred after dinner (3h average glucose Control = 7.55±1.21 mmol/L vs. PMW = 6.71 ± 0.80mmol/L, p = 0.039), when insulin sensitivity is typically diminished. Despite improvements in post-prandial glucose control, this did not translate to improvements in 24h glucose control (p > 0.05). CONCLUSION Physically inactive and metabolically healthy young women, PMW improves post-prandial glucose but not 24h glucose control.
Collapse
Affiliation(s)
- Michael S. Brian
- Department of Kinesiology, University of New Hampshire, Durham, NH, USA
| | - Bilal A. Chaudhry
- Department of Kinesiology, University of New Hampshire, Durham, NH, USA
| | - Maison D’Amelio
- Department of Kinesiology, University of New Hampshire, Durham, NH, USA
| | - Emily E. Waite
- Department of Kinesiology, University of New Hampshire, Durham, NH, USA
| | - John G. Dennett
- Department of Kinesiology, University of New Hampshire, Durham, NH, USA
| | | | - Deborah L. Feairheller
- Department of Kinesiology, University of New Hampshire, Durham, NH, USA
- Department of Kinesiology, California State University San Marcos, San Marcos, CA, USA
| |
Collapse
|
43
|
Karhiaho IP, Kurki SH, Parviainen HI, Kullamaa L, Färkkilä MA, Matikainen N, Tuomi T. The hidden epidemic: Uncovering incidental fatty liver disease and its metabolic comorbidities by datamining in a hospital data lake - A real-world cohort study. Diabetes Res Clin Pract 2024; 210:111609. [PMID: 38479446 DOI: 10.1016/j.diabres.2024.111609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 02/20/2024] [Accepted: 03/10/2024] [Indexed: 03/18/2024]
Abstract
AIMS To identify individuals with incidental fatty liver disease (FLD), and to evaluate its prevalence, metabolic co-morbidities and impact on follow-up. METHODS We leveraged the data-lake of Helsinki Uusimaa Hospital district (Finland) with a population of 1.7 million (specialist and primary care). A phrase recognition script on abdominal imaging reports (2008-2020) identified/excluded FLD or cirrhosis; we extracted ICD-codes, laboratory and BMI data. RESULTS Excluding those with other liver diseases, the prevalence of FLD was 29% (steatosis yes/no, N=61,271/155,521; cirrhosis, N=3502). The false positive and negative rates were 5-6%. Only 1.6% of the FLD cases had the ICD code recorded and 32% had undergone full clinical evaluation for associated co-morbidities. Of the 35-65-year-old individuals with FLD, 20% had diabetes, 42% prediabetes and 28% a high liver fibrosis index. FLD was independently predicted by diabetes (OR 1.56, CI 1.46-1.66, p = 2.3 * 10^-41), BMI (1.46, 1.42-1.50, p = 1.7 * 10^-154) and plasma triglyceride level (1.5, 1.43-1.57, p = 3.5 * 10^-68). Alanine aminotransferase level mildly increased (1.12, 1.08-1.16, p = 2.2 * 10^-9) and high age decreased the risk (0.92, 0.89-0.94, p = 4.65*10^-09). Half of the cases had normal ALT. CONCLUSIONS The incidental radiological finding of FLD is reliable and associated with metabolic risks but largely ignored, although it should lead to metabolic and hepatic follow-up.
Collapse
Affiliation(s)
- Iiro P Karhiaho
- Endocrinology, Abdominal Center, Helsinki University Hospital, Helsinki, Finland
| | - Samu H Kurki
- Endocrinology, Abdominal Center, Helsinki University Hospital, Helsinki, Finland; Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Helka I Parviainen
- Medical Imaging Center, Department of Radiology, Helsinki University Hospital and University of Helsinki and Department of Radiology, Vaasa Central Hospital, Wellbeing Services County of Ostrobothnia, Finland
| | - Liisa Kullamaa
- Endocrinology, Abdominal Center, Helsinki University Hospital, Helsinki, Finland
| | - Martti A Färkkilä
- Gastroenterology, Abdominal Center, Helsinki University Hospital, Helsinki, Finland
| | - Niina Matikainen
- Endocrinology, Abdominal Center, Helsinki University Hospital, Helsinki, Finland; Research Program of Clinical and Molecular Metabolism, University of Helsinki, Helsinki, Finland
| | - Tiinamaija Tuomi
- Endocrinology, Abdominal Center, Helsinki University Hospital, Helsinki, Finland; Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland; Research Program of Clinical and Molecular Metabolism, University of Helsinki, Helsinki, Finland; Folkhälsan Research Centre, Helsinki, Finland; Lund University, Diabetes Centre, Malmo, Sweden.
| |
Collapse
|
44
|
Lu Y, Jia Y, Lu J, Liu J, Xu Y, Liu Y, Chen K. Progenies of gestational diabetes mellitus exhibit sex disparity in metabolism after respective therapies of insulin, glibenclamide, and metformin in dams during pregnancy. Arch Physiol Biochem 2024; 130:183-195. [PMID: 34689672 DOI: 10.1080/13813455.2021.1991957] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 09/28/2021] [Accepted: 10/06/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND The aim of this study was to compare the sex-dependent intergenerational effects of insulin, glibenclamide, and metformin on glucose and lipid metabolism in the offspring born to GDM mice. METHODS The murine GDM was induced by high fat diet. The offspring were grouped based on the treatments in maternal mice. ITT and GTT were performed at 4th and 8th weeks of age, respectively. Serum levels of TC, TG, HDL-C, and LDL-C plus hepatic levels of TG and TC, were respectively determined by enzymatic kits. Western blotting was conducted to detect related proteins in the livers from offspring. RESULTS The dyslipidaemia, hepatic lipid abnormality, and insulin insensitivity caused by GDM were persistently normalised in male adult offspring by the respective therapies of insulin, glibenclamide, and metformin during maternal pregnancy. Specifically, the decreases in plasma TC, TG, and LDL-C levels (29%, 37.8%, and 57.7%, respectively, p ˂ .05) and in hepatic lipid contents (TC 31.3% and TG 39.2%, p ˂ .05), the increases in hepatic phosphorylation levels of AKT, CPT1A, PPAR-α, and PPAR-γ (57.1%, 91.7%, 68%, and 173.3%, respectively, p ˂ .05) and the inhibition of G6Pase, PEPCK, and HMGCS1 (35.7%, 68.8%, and 77.3% respectively, p ˂ .05) were still observed in the male offspring born to treated GDM mice from 4th to 8th week of age. Unexpectedly, the aforementioned parameters in female progenies in different groups were not significantly changed compared with controls. CONCLUSIONS Respective treatments in GDM mice during pregnancy with insulin, glibenclamide, and metformin have the long-term persistent effects in male offspring, while female progenies born to untreated dams showed an autonomous inhibition of intergenerational relay of glucose and lipid dysregulation. Our current findings may imply a sex-dependent strategy of medical care for GDM mothers and their offspring.NoveltiesRespective interventions of insulin, glibenclamide, and metformin on dams exerted the persisted effects on male progenies.Therapies of three drugs on dams had the similarly improved effects in offspring.Female offspring autonomously corrected their dysregulated glucose-lipid metabolism caused by gestational diabetes mellitus (GDM) in dams.
Collapse
Affiliation(s)
- Yao Lu
- Department of Anesthesiology, the First Affiliated Hospital, Anhui Medical University, Hefei, PR China
| | - Yajing Jia
- Department of Health Inspection and Quarantine, School of Public Health, Anhui Medical University, Hefei, PR China
| | - Jing Lu
- Department of Nutrition and Food Science, School of Public Health, Anhui Medical University, Hefei, PR China
| | - Juan Liu
- Department of Health Inspection and Quarantine, School of Public Health, Anhui Medical University, Hefei, PR China
| | - Yuxin Xu
- Department of Ophthalmology of Second Affiliated Hospital, Anhui Medical University, Hefei, PR China
| | - Yong Liu
- AIER Hefei Eye Hospital Affiliated to Anhui Medical University, Hefei, PR China
| | - Keyang Chen
- Department of Health Inspection and Quarantine, School of Public Health, Anhui Medical University, Hefei, PR China
| |
Collapse
|
45
|
Lee SJL, Van Rens FECA, Peiffer JJ. Effects of In-Exercise Carbohydrate Supplementation on Prolonged High-Intensity Exercise Performance in Oral Contraceptive Users. Int J Sports Physiol Perform 2024; 19:356-364. [PMID: 38237580 DOI: 10.1123/ijspp.2023-0143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 11/28/2023] [Accepted: 12/07/2023] [Indexed: 03/23/2024]
Abstract
PURPOSE To examine the impact of oral contraceptive (OC) phases on performance, physiological, and subjective responses to prolonged, intensive exercise when carbohydrate (CHO) stores are reduced. METHODS Ten well-trained female cyclists using monophasic OC completed 4 identical trials (>150 min) under conditions of in-trial 60-g·h-1 CHO supplementation (CHO+) or placebo (CHO-) during the sugar- (SUG) and active-pill (ACT) phases of their OC cycle. Each trial comprised two 400-kcal time trials (TT) separated by 1 hour of submaximal cycling at first ventilatory threshold. RESULTS Change in completion time from TT1 to TT2 was minimized in CHO+ compared with CHO- (4.06 [2.55] vs 6.08 [5.33] min; P = .019, effect size = -0.36). An interaction effect of OC and CHO was observed for time to complete TT (P = .006), mean TT power (P = .002), mean TT heart rate (P = .002), and posttrial emotional balance (P = .020) and negative emotional state (P = .033). In ACT, mean TT power and heart rate were higher in CHO+ when compared with CHO-, resulting in faster TTs in CHO+ and improved posttrial emotional well-being. When CHO was not supplemented, TT power and heart rate were higher in SUG when compared with ACT, resulting in faster TTs in SUG and improved posttrial emotional balance. CONCLUSION CHO depletion during ACT negatively influenced TT performance and emotional well-being when compared with SUG. Irrespective of OC pill phase, CHO supplementation should be prioritized to sustain performance and improve postexercise recovery-stress balance.
Collapse
Affiliation(s)
- Serene J L Lee
- Center for Healthy Aging, Murdoch University, Perth, WA, Australia
- Murdoch Applied Sports Science Laboratory, Murdoch University, Perth, WA, Australia
| | | | - Jeremiah J Peiffer
- Center for Healthy Aging, Murdoch University, Perth, WA, Australia
- Murdoch Applied Sports Science Laboratory, Murdoch University, Perth, WA, Australia
| |
Collapse
|
46
|
Gebreyesus LH, Choi S, Neequaye P, Mahmoud M, Mahmoud M, Ofosu-Boateng M, Twum E, Nnamani DO, Wang L, Yadak N, Ghosh S, Gonzalez FJ, Gyamfi MA. Pregnane X receptor knockout mitigates weight gain and hepatic metabolic dysregulation in female C57BL/6 J mice on a long-term high-fat diet. Biomed Pharmacother 2024; 173:116341. [PMID: 38428309 PMCID: PMC10983615 DOI: 10.1016/j.biopha.2024.116341] [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: 12/16/2023] [Revised: 02/09/2024] [Accepted: 02/23/2024] [Indexed: 03/03/2024] Open
Abstract
Obesity is a significant risk factor for several chronic diseases. However, pre-menopausal females are protected against high-fat diet (HFD)-induced obesity and its adverse effects. The pregnane X receptor (PXR, NR1I2), a xenobiotic-sensing nuclear receptor, promotes short-term obesity-associated liver disease only in male mice but not in females. Therefore, the current study investigated the metabolic and pathophysiological effects of a long-term 52-week HFD in female wild-type (WT) and PXR-KO mice and characterized the PXR-dependent molecular pathways involved. After 52 weeks of HFD ingestion, the body and liver weights and several markers of hepatotoxicity were significantly higher in WT mice than in their PXR-KO counterparts. The HFD-induced liver injury in WT female mice was also associated with upregulation of the hepatic mRNA levels of peroxisome proliferator-activated receptor gamma (Pparg), its target genes, fat-specific protein 27 (Fsp27), and the liver-specific Fsp27b involved in lipid accumulation, apoptosis, and inflammation. Notably, PXR-KO mice displayed elevated hepatic Cyp2a5 (anti-obesity gene), aldo-keto reductase 1b7 (Akr1b7), glutathione-S-transferase M3 (Gstm3) (antioxidant gene), and AMP-activated protein kinase (AMPK) levels, contributing to protection against long-term HFD-induced obesity and inflammation. RNA sequencing analysis revealed a general blunting of the transcriptomic response to HFD in PXR-KO compared to WT mice. Pathway enrichment analysis demonstrated enrichment by HFD for several pathways, including oxidative stress and redox pathway, cholesterol biosynthesis, and glycolysis/gluconeogenesis in WT but not PXR-KO mice. In conclusion, this study provides new insights into the molecular mechanisms by which PXR deficiency protects against long-term HFD-induced severe obesity and its adverse effects in female mice.
Collapse
Affiliation(s)
- Lidya H Gebreyesus
- Department of Pharmaceutical Sciences, The University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN 38163, USA
| | - Sora Choi
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, NC 27707, USA
| | - Prince Neequaye
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, NC 27707, USA
| | - Mattia Mahmoud
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, NC 27707, USA
| | - Mia Mahmoud
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, NC 27707, USA
| | - Malvin Ofosu-Boateng
- Department of Pharmaceutical Sciences, The University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN 38163, USA
| | - Elizabeth Twum
- Department of Pharmaceutical Sciences, The University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN 38163, USA
| | - Daniel O Nnamani
- Department of Pharmaceutical Sciences, The University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN 38163, USA
| | - Lijin Wang
- Center for Computational Biology, Duke-NUS Medical School, 8 College Road, Singapore
| | - Nour Yadak
- Department of Pathology and Laboratory Medicine, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Sujoy Ghosh
- Center for Computational Biology, Duke-NUS Medical School, 8 College Road, Singapore; Bioinformatics and Computational Biology Core, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| | - Frank J Gonzalez
- Center for Cancer Research, National Cancer Institute, Building 37, Room 3106, Bethesda, MD 20892, USA
| | - Maxwell A Gyamfi
- Department of Pharmaceutical Sciences, The University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN 38163, USA; Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, NC 27707, USA.
| |
Collapse
|
47
|
Liu H, Wu Y, Zhu H, Wang P, Chen T, Xia A, Zhao Z, He D, Chen X, Xu J, Ji L. Association between napping and type 2 diabetes mellitus. Front Endocrinol (Lausanne) 2024; 15:1294638. [PMID: 38590820 PMCID: PMC10999583 DOI: 10.3389/fendo.2024.1294638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 03/12/2024] [Indexed: 04/10/2024] Open
Abstract
As the incidence of type 2 diabetes mellitus (T2DM) is increasing rapidly and its consequences are severe, effective intervention and prevention, including sleep-related interventions, are urgently needed. As a component of sleep architecture, naps, alone or in combination with nocturnal sleep, may influence the onset and progression of T2DM. Overall, napping is associated with an increased risk of T2DM in women, especially in postmenopausal White women. Our study showed that napping >30 minutes (min) increased the risk of T2DM by 8-21%. In addition, non-optimal nighttime sleep increases T2DM risk, and this effect combines with the effect of napping. For nondiabetic patients, napping >30 min could increase the risks of high HbA1c levels and impaired fasting glucose (IFG), which would increase the risk of developing T2DM later on. For diabetic patients, prolonged napping may further impair glycemic control and increase the risk of developing diabetic complications (e.g., diabetic nephropathy) in the distant future. The following three mechanisms are suggested as interpretations for the association between napping and T2DM. First, napping >30 min increases the levels of important inflammatory factors, including interleukin 6 and C-reactive protein, elevating the risks of inflammation, associated adiposity and T2DM. Second, the interaction between postmenopausal hormonal changes and napping further increases insulin resistance. Third, prolonged napping may also affect melatonin secretion by interfering with nighttime sleep, leading to circadian rhythm disruption and further increasing the risk of T2DM. This review summarizes the existing evidence on the effect of napping on T2DM and provides detailed information for future T2DM intervention and prevention strategies that address napping.
Collapse
Affiliation(s)
- Hongyi Liu
- School of Public Health, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Yingxin Wu
- School of Public Health, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Hui Zhu
- Department of Internal Medicine, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Penghao Wang
- School of Public Health, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Tao Chen
- School of Public Health, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Anyu Xia
- Department of Clinical Medicine, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Zhijia Zhao
- School of Public Health, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Da He
- Department of Obstetrics and Gynecology, Yinzhou District Maternal and Child Health Care Institute, Ningbo, Zhejiang, China
| | - Xiang Chen
- Department of Obstetrics and Gynecology, Yinzhou District Maternal and Child Health Care Institute, Ningbo, Zhejiang, China
| | - Jin Xu
- School of Public Health, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
- Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, China
| | - Lindan Ji
- Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
48
|
Muhammad A, Hixon JC, Pharmacy Yusuf A, Rivas Zarete JI, Johnson I, Miller J, Adu-Addai B, Yates C, Mahavadi S. Sex-specific epigenetics drive low GPER expression in gastrointestinal smooth muscles in type 2 diabetic mice. Sci Rep 2024; 14:5633. [PMID: 38453938 PMCID: PMC10920797 DOI: 10.1038/s41598-024-54213-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 02/09/2024] [Indexed: 03/09/2024] Open
Abstract
Type 2 diabetes mellitus (T2D) causes gastroparesis, delayed intestinal transit, and constipation, for unknown reasons. Complications are predominant in women than men (particularly pregnant and postmenopausal women), suggesting a female hormone-mediated mechanism. Low G-protein coupled estrogen receptor (GPER) expression from epigenetic modifications may explain it. We explored sexually differentiated GPER expression and gastrointestinal symptoms related to GPER alterations in wild-type (WT) and T2D mice (db/db). We also created smooth muscle-specific GPER knockout (GPER KO) mice to phenotypically explore the effect of GPER deficiency on gastrointestinal motility. GPER mRNA and protein expression, DNA methylation and histone modifications were measured from stomach and colon samples of db/db and WT mice. Changes in gut motility were also evaluated as daily fecal pellet production patterns. We found that WT female tissues have the highest GPER mRNA and protein expressions. The expression is lowest in all db/db. GPER downregulation is associated with promoter hypermethylation and reduced enrichment of H3K4me3 and H3K27ac marks around the GPER promoter. We also observed sex-specific disparities in fecal pellet production patterns of the GPER KO mice compared to WT. We thus, conclude that T2D impairs gut GPER expression, and epigenetic sex-specific mechanisms matter in the downregulation.
Collapse
Affiliation(s)
- Aliyu Muhammad
- Department of Biology, Center for Cancer Research, Tuskegee University, Tuskegee, AL, 36088, USA
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, P.M.B. 1044, Zaria, Kaduna State, Nigeria
| | - Juanita C Hixon
- Department of Biology, Center for Cancer Research, Tuskegee University, Tuskegee, AL, 36088, USA
| | | | - Jatna I Rivas Zarete
- Department of Biomedical Sciences, College of Veterinary Medicine, Tuskegee University, Tuskegee, AL, 36088, USA
| | - India Johnson
- Department of Biology, Center for Cancer Research, Tuskegee University, Tuskegee, AL, 36088, USA
| | - Jamial Miller
- Department of Biology, Center for Cancer Research, Tuskegee University, Tuskegee, AL, 36088, USA
| | - Benjamin Adu-Addai
- Department of Biomedical Sciences, College of Veterinary Medicine, Tuskegee University, Tuskegee, AL, 36088, USA
| | - Clayton Yates
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sunila Mahavadi
- Department of Biology, Center for Cancer Research, Tuskegee University, Tuskegee, AL, 36088, USA.
| |
Collapse
|
49
|
Hahn MK, Giacca A, Pereira S. In vivo techniques for assessment of insulin sensitivity and glucose metabolism. J Endocrinol 2024; 260:e230308. [PMID: 38198372 PMCID: PMC10895285 DOI: 10.1530/joe-23-0308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 01/10/2024] [Indexed: 01/12/2024]
Abstract
Metabolic tests are vital to determine in vivo insulin sensitivity and glucose metabolism in preclinical models, usually rodents. Such tests include glucose tolerance tests, insulin tolerance tests, and glucose clamps. Although these tests are not standardized, there are general guidelines for their completion and analysis that are constantly being refined. In this review, we describe metabolic tests in rodents as well as factors to consider when designing and performing these tests.
Collapse
Affiliation(s)
- Margaret K Hahn
- Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
- Department of Pharmacology, University of Toronto, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
- Banting & Best Diabetes Centre, Toronto, Ontario, Canada
| | - Adria Giacca
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
- Banting & Best Diabetes Centre, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Sandra Pereira
- Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
50
|
Sugiyama N, Uehara O, Kawano Y, Paudel D, Morikawa T, Nakamoto N, Kato S, Takayama T, Nagasawa T, Miura H, Abiko Y, Furuichi Y. Ingenuity pathway analysis of gingival epithelial cells stimulated with estradiol and progesterone. J Oral Biosci 2024; 66:26-34. [PMID: 37949170 DOI: 10.1016/j.job.2023.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 11/03/2023] [Accepted: 11/06/2023] [Indexed: 11/12/2023]
Abstract
OBJECTIVE Periodontal disease is a risk factor for preterm delivery, and elevated female hormone levels during pregnancy promote hormone-dependent periodontopathogenic bacterial growth and gingivitis. Although the saliva of pregnant women contains female hormones at elevated levels, their effects on the gingiva are poorly understood. Therefore, in this study, we investigated the effects of estradiol and progesterone stimulation on gingival epithelial cells via ingenuity pathway analysis. METHODS Human gingival epithelial progenitors were cultured in a CnT-Prime medium; 17β-estradiol (E2) and progesterone (P4) were used as the reagents. Cells treated with dimethyl sulfoxide alone were used as the control group. Cells in the control and experimental groups were incubated for 12 h. RNA was extracted from the cultured cells, RNA-Seq was performed, and pathway analysis was conducted. RESULTS Differentially expressed genes were detected for 699 (over 2-fold increase) and 348 (decrease) genes in group E2 and for 1448 (increase) and 924 (decrease) genes in group P4 compared with those in the control group (FDR <0.05, n = 4). The z-scores of the pathways suggest that E2 and P4 increased the activity of the wound healing signaling pathway. The activation of this pathway was higher in the E2 and P4 groups than that in the control group. CONCLUSIONS The results of this study suggest that estradiol and progesterone may affect gingival homeostasis and wound healing.
Collapse
Affiliation(s)
- Nodoka Sugiyama
- Division of Periodontology and Endodontology, Department of Oral Rehabilitation, School of Dentistry, Health, Sciences University of Hokkaido, 1757, Ishikari-Tobetsu, Kanazawa, Hokkaido, 061-0293, Japan.
| | - Osamu Uehara
- Division of Disease Controlrol and Molecular Epidemiology, Department of Oral Growth and Development, School of Dentistry, Health Sciences University of Hokkaido, 1757, Ishikari-Tobetsu, Kanazawa, Hokkaido, 061- 0293, Japan
| | - Yutaka Kawano
- Department of Gastroenterology and Oncology Tokushima University Graduate School of Biomedical Sciences, Tokushima, 3-18-15, Kuramoto-cho, Tokushima City, Tokushima, 770-8503, Japan
| | - Durga Paudel
- Advanced Research Promotion Center, Health Sciences University of Hokkaido, 1757, Ishikari- Tobetsu, Kanazawa, Hokkaido, 061-0293, Japan
| | - Tetsuro Morikawa
- Division of Oral Medicine and Pathology, Department of Human Biology and Pathophysiology, School of Dentistry, Health Sciences University of Hokkaido, 1757, Ishikari-Tobetsu, Kanazawa, Hokkaido, 061-0293, Japan
| | - Norihiro Nakamoto
- Division of Periodontology and Endodontology, Department of Oral Rehabilitation, School of Dentistry, Health, Sciences University of Hokkaido, 1757, Ishikari-Tobetsu, Kanazawa, Hokkaido, 061-0293, Japan
| | - Satsuki Kato
- Division of Periodontology and Endodontology, Department of Oral Rehabilitation, School of Dentistry, Health, Sciences University of Hokkaido, 1757, Ishikari-Tobetsu, Kanazawa, Hokkaido, 061-0293, Japan
| | - Tetsuji Takayama
- Department of Gastroenterology and Oncology Tokushima University Graduate School of Biomedical Sciences, Tokushima, 3-18-15, Kuramoto-cho, Tokushima City, Tokushima, 770-8503, Japan
| | - Toshiyuki Nagasawa
- Division of Advanced Clinical Education, Department of Integrated Dental Education, School of Dentistry, Health, Sciences University of Hokkaido, 1757, Ishikari-Tobetsu, Kanazawa, Hokkaido, 061-0293, Japan
| | - Hiroko Miura
- Division of Disease Controlrol and Molecular Epidemiology, Department of Oral Growth and Development, School of Dentistry, Health Sciences University of Hokkaido, 1757, Ishikari-Tobetsu, Kanazawa, Hokkaido, 061- 0293, Japan
| | - Yoshihiro Abiko
- Division of Oral Medicine and Pathology, Department of Human Biology and Pathophysiology, School of Dentistry, Health Sciences University of Hokkaido, 1757, Ishikari-Tobetsu, Kanazawa, Hokkaido, 061-0293, Japan.
| | - Yasushi Furuichi
- Division of Periodontology and Endodontology, Department of Oral Rehabilitation, School of Dentistry, Health, Sciences University of Hokkaido, 1757, Ishikari-Tobetsu, Kanazawa, Hokkaido, 061-0293, Japan
| |
Collapse
|