1
|
González-Vila A, Ibrahim-Alasoufi AM, Luengo-Mateos M, Pardo-García V, Diaz-López A, Fernández-Rodríguez B, Poutanen M, Ohlsson C, Tena-Sempere M, Diéguez-González C, García-García MDC, Barca-Mayo O. IL-6 decodes sex and diet-dependent circadian and metabolic rhythms. Mol Metab 2025; 97:102171. [PMID: 40412648 PMCID: PMC12159943 DOI: 10.1016/j.molmet.2025.102171] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 05/10/2025] [Accepted: 05/19/2025] [Indexed: 05/27/2025] Open
Abstract
OBJECTIVE Interleukin-6 (IL-6) is a pleiotropic cytokine involved in immune regulation and energy metabolism. Its diurnal secretion influences core circadian components, emphasizing its critical role in circadian biology. Despite known sex differences in immune, circadian, and metabolic processes, how IL-6 integrates these processes remains poorly understood. METHODS IL6 knockout (KO) and control mice of both sexes were phenotyped for circadian and metabolic traits under standard (STD) and high-fat diet (HFD), fasting, and time-restricted feeding. Molecular analyses in muscle, liver, and hypothalamus assessed clock gene expression and IL-6 signaling pathway. Circulating sex steroid hormones were quantified to examine their contribution to the observed sex-specific phenotypes. RESULTS IL-6 deficiency disrupts circadian locomotor and metabolic rhythms in a sex- and diet-dependent manner. Males exhibit impaired light-driven circadian rhythms under STD conditions and metabolic misalignment under HFD, whereas females display greater circadian resilience under STD conditions but increased vulnerability to circadian disruption during HFD. Additionally, IL-6 emerges as a novel regulator of the food-entrainable oscillator (FEO), linking food anticipatory activity and metabolic cycles under both STD and HFD in a sex-dependent manner. CONCLUSIONS These findings identify IL-6 as a critical mediator of circadian-metabolic plasticity, shaping sex- and diet-specific trade-offs between circadian stability and metabolic homeostasis. Our study highlights IL-6 as a potential therapeutic target for mitigating circadian misalignment-associated metabolic disorders, with implications for the timed modulation of IL-6 signaling.
Collapse
Affiliation(s)
- Antía González-Vila
- Physiology Department, Molecular Medicine and Chronic Diseases Research Centre (CiMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Ali Mohammad Ibrahim-Alasoufi
- Physiology Department, Molecular Medicine and Chronic Diseases Research Centre (CiMUS), University of Santiago de Compostela, Santiago de Compostela, Spain; Biology Department, Al-Hussein Bin Talal University, Ma'an, Jordan
| | - María Luengo-Mateos
- Physiology Department, Molecular Medicine and Chronic Diseases Research Centre (CiMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Víctor Pardo-García
- Physiology Department, Molecular Medicine and Chronic Diseases Research Centre (CiMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Alejandro Diaz-López
- Physiology Department, Molecular Medicine and Chronic Diseases Research Centre (CiMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Belén Fernández-Rodríguez
- Physiology Department, Molecular Medicine and Chronic Diseases Research Centre (CiMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Matti Poutanen
- Research Center for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland; Turku Center for Disease Modeling, Turku, Finland
| | - Claes Ohlsson
- Sahlgrenska Osteoporosis Centre, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Manuel Tena-Sempere
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Instituto Maimónides de Investigación Biomédica (IMIBIC)/Hospital Universitario Reina Sofía, Córdoba, Spain; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Carlos Diéguez-González
- Physiology Department, Molecular Medicine and Chronic Diseases Research Centre (CiMUS), University of Santiago de Compostela, Santiago de Compostela, Spain; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - María Del Carmen García-García
- Physiology Department, Molecular Medicine and Chronic Diseases Research Centre (CiMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Olga Barca-Mayo
- Physiology Department, Molecular Medicine and Chronic Diseases Research Centre (CiMUS), University of Santiago de Compostela, Santiago de Compostela, Spain.
| |
Collapse
|
2
|
Peng G, Mosleh E, Yuhas A, Katada K, Kasinathan D, Cherry C, Golson ML. FOXM1 cooperates with ERα to regulate functional β-cell mass. Am J Physiol Endocrinol Metab 2025; 328:E804-E821. [PMID: 40261794 PMCID: PMC12145799 DOI: 10.1152/ajpendo.00438.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/02/2024] [Accepted: 04/11/2025] [Indexed: 04/24/2025]
Abstract
The transcription factor forkhead box (FOX)M1 regulates β-cell proliferation and insulin secretion. Our previous work demonstrates that expressing a constitutively active form of FOXM1 (FOXM1*) in β-cells increases β-cell function, proliferation, and mass in male mice. However, in contrast to what is observed in males, we demonstrate here that in female mice expression of FOXM1* in β-cells does not affect β-cell proliferation or glucose tolerance. Similarly, FOXM1* transduction of male but not female human islets enhances insulin secretion in response to elevated glucose. We therefore examined the mechanism behind this sexual dimorphism. Estrogen contributes to diabetes susceptibility differences between males and females, and estrogen receptor (ER)α is the primary mediator of β-cell estrogen signaling. Moreover, in breast cancer cells, ERα and FOXM1 work together to drive gene expression. We therefore examined whether FOXM1 and ERα functionally interact in β-cells. FOXM1* rescued elevated fasting glucose, glucose intolerance, and homeostatic model assessment of β-cell function (HOMA-B) in female mice with a β-cell-specific ERα deletion. Furthermore, in the presence of estrogen, the FOXM1 and ERα cistromes exhibit significant overlap in βTC6 β-cells. In addition, FOXM1 and ERα binding sites frequently occur in complex enhancers co-occupied by other islet transcription factors. These data indicate that FOXM1 and nuclear ERα cooperate to regulate β-cell function and suggest a general mechanism contributing to the lower incidence of diabetes observed in women.NEW & NOTEWORTHY Here we investigate why the effects of increasing FOXM1 activity in β-cells observed in male mice are not seen in female mice. ERα likely collaborates with FOXM1 and other transcription factors to enhance gene expression related to β-cell function. Higher estrogen levels in females may contribute to their increased insulin secretion and the more severe consequences of losing transcription factors like FOXM1 in males. Overall, these findings shed light on sex differences in diabetes susceptibility.
Collapse
Affiliation(s)
- Guihong Peng
- Department of Medicine, Johns Hopkins University, Baltimore, MD
| | - Elham Mosleh
- Department of Genetics, University of Pennsylvania, Philadelphia, PA
| | - Andrew Yuhas
- Department of Genetics, University of Pennsylvania, Philadelphia, PA
| | - Kay Katada
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Devi Kasinathan
- Department of Physiology, Johns Hopkins University, Baltimore, MD
| | | | - Maria L. Golson
- Department of Medicine, Johns Hopkins University, Baltimore, MD
- Department of Genetics, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
3
|
Choi MJ, Yu J. Menopause and Diabetes Risk Along with Trajectory of β-Cell Function and Insulin Sensitivity: A Community-Based Cohort Study. Healthcare (Basel) 2025; 13:1062. [PMID: 40361840 PMCID: PMC12071248 DOI: 10.3390/healthcare13091062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Revised: 04/30/2025] [Accepted: 05/02/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND The relationship between menopause and diabetes risk is unclear, with some studies indicating a weak association. This study examined changes in diabetes risk, β-cell function, and insulin sensitivity in relation to menopause. METHODS In this community-based cohort study, data from 6684 visits to 1224 women over a 16-year follow-up were analyzed. Diabetes risk changes were assessed in relation to the different menopausal phases: premenopausal (≥3 years before menopause), perimenopausal (2 years before to 1 year after menopause), and postmenopausal (≥2 years after menopause). Changes in β-cell function and insulin sensitivity indices were tracked, and their relationship with diabetes risk was assessed. Generalized estimating equations and linear mixed models were used, adjusting for covariates including age at menopause and obesity. RESULTS Diabetes incidence was 18.6% among participants. The odds ratio (OR) of diabetes increased by 1.03 times annually during the premenopausal period (OR 1.03; 95% CI 1.02-1.04) and decreased during the postmenopausal period (OR 0.96; 95% CI 0.95-0.97). The incident diabetes groups showed a decline in insulin sensitivity and β-cell function, resulting in a decrease in the disposition indices over time. A large change in insulin sensitivity, especially during the period immediately before the onset of diabetes, increased the risk of diabetes (OR 1.88; 95% CI 1.33-2.67). CONCLUSIONS This study indicates an increased diabetes risk during the premenopausal periods, compared with that in the postmenopausal period, independent of age at menopause and obesity. Additionally, a decrease in insulin sensitivity followed by a subsequent decrease in β-cell function depending on the time of onset was related to the risk of diabetes. These findings enhance the understanding of diabetes risk and associated changes in insulin indices in relation to menopause, emphasizing the importance of health management and diabetes prevention for women in menopausal transition.
Collapse
Affiliation(s)
- Mi Jin Choi
- College of Nursing, Gyeongsang National University, Jinju-si 52727, Republic of Korea;
| | - Juyoun Yu
- Department of Nursing, Changwon National University, Changwon-si 51140, Republic of Korea
| |
Collapse
|
4
|
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
|
5
|
Roberts TD, Hutchinson DS, Wootten D, De Blasio MJ, Ritchie RH. Advances in incretin therapies for targeting cardiovascular disease in diabetes. J Mol Cell Cardiol 2025; 202:102-115. [PMID: 40086589 DOI: 10.1016/j.yjmcc.2025.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 02/12/2025] [Accepted: 03/11/2025] [Indexed: 03/16/2025]
Abstract
The global prevalence of obesity is skyrocketing at an alarming rate, with recent data estimating that one-in-eight people are now living with the disease. Obesity is a chronic metabolic disorder that shares underlying pathophysiology with other metabolically-linked diseases such as type 2 diabetes mellitus, cardiovascular disease and diabetic cardiomyopathy. There is a distinct correlation between type 2 diabetes status and the likelihood of heart failure. Of note, there is an apparent sexual dimorphism, with women disproportionately affected with respect to the degree of severity of the cardiac phenotype of diabetic cardiomyopathy that results from diabetes. The current pharmacotherapies available for the attenuation of hyperglycaemia in type 2 diabetes are not always effective, and have varying degrees of efficacy in the setting of heart failure. Insulin can worsen heart failure prognosis whereas metformin, sodium-glucose cotransporter 2 inhibitors (SGLT2i) and more recently, glucagon-like peptide-1 receptor agonists (GLP-1RAs), have demonstrated cardioprotection with their administration. This review will highlight the advancement of incretin therapies for individuals with diabetes and heart failure and explore newly-reported evidence of the clinical usefulness of GLP-1R agonists in this distinct phenotype of heart failure.
Collapse
Affiliation(s)
- Timothy D Roberts
- Heart Failure Pharmacology Laboratory, Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, VIC, Australia
| | - Dana S Hutchinson
- Metabolic G Protein-Coupled Receptor Laboratory, Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, VIC, Australia
| | - Denise Wootten
- Metabolic G Protein-Coupled Receptor Laboratory, Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, VIC, Australia; ARC Centre for Cryo-Electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, Victoria, Australia
| | - Miles J De Blasio
- Heart Failure Pharmacology Laboratory, Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, VIC, Australia.
| | - Rebecca H Ritchie
- Heart Failure Pharmacology Laboratory, Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, VIC, Australia.
| |
Collapse
|
6
|
Miao MY, Han WW, Lyu JQ, Liu ZY, Jiang W, Zhang Z, Zhu Z, Qin LQ, Chen GC. Female reproductive factors and metabolic dysfunction-associated steatotic liver disease: an integrated analysis of population cohort, liver imaging, and genetic data. Am J Obstet Gynecol 2025:S0002-9378(25)00212-1. [PMID: 40204176 DOI: 10.1016/j.ajog.2025.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 03/30/2025] [Accepted: 04/01/2025] [Indexed: 04/11/2025]
Abstract
BACKGROUND Although premenopausal women are at lower risk for metabolic dysfunction-associated steatotic liver disease than men within the same age group, the sex advantage becomes minimal after menopause, suggesting a role for female hormones. OBJECTIVE This study aimed to elucidate the role of reproductive factors in the development of female metabolic dysfunction-associated steatotic liver disease, using an integrated analysis. STUDY DESIGN Up to 269,607 women without metabolic dysfunction-associated steatotic liver disease during baseline recruitment (2006-2010) of the UK Biobank were included. Among these, 21,017 women did not develop metabolic dysfunction-associated steatotic liver disease across the follow-up period (through December 2021) and had the measure of liver proton density fat fraction quantified by magnetic resonance imaging since 2014. Multivariable Cox regression models were applied to assess the prospective relationships of various reproductive factors with incident metabolic dysfunction-associated steatotic liver disease. Multiple linear regression models were used to explore the relationships of reproductive factors with liver proton density fat fraction. A 2-sample Mendelian randomization analysis was conducted to investigate causality in the relationships of certain reproductive factors with metabolic dysfunction-associated steatotic liver disease. RESULTS During a median of 12.65 years of follow-up, 3077 incident cases of metabolic dysfunction-associated steatotic liver disease were identified. Early menarche, a greater number of live births, younger age at first live birth, and oral contraceptives or hormone replacement therapy initiated at a young age were associated with an elevated risk of metabolic dysfunction-associated steatotic liver disease and higher levels of liver proton density fat fraction. Several other reproductive factors (ie, a greater number of miscarriages, surgical menopause at a young age, and prolonged use of hormone replacement therapy) were associated with incident metabolic dysfunction-associated steatotic liver disease but not with liver proton density fat fraction. In the Mendelian randomization analysis, genetically determined earlier age at menarche and younger age at first live birth were substantially associated with increased risk of metabolic dysfunction-associated steatotic liver disease. CONCLUSION Several reproductive factors were associated with the risk of and histologic features of metabolic dysfunction-associated steatotic liver disease, supporting the role of female hormones in the pathogenesis of metabolic dysfunction-associated steatotic liver disease.
Collapse
Affiliation(s)
- Meng-Yuan Miao
- Fourth Affiliated Hospital, School of Public Health, MOE Key Laboratory of Geriatric Diseases and Immunology, Medical College of Soochow University, Suzhou, China
| | - Wen-Wen Han
- Fourth Affiliated Hospital, School of Public Health, MOE Key Laboratory of Geriatric Diseases and Immunology, Medical College of Soochow University, Suzhou, China
| | - Jie-Qiong Lyu
- Fourth Affiliated Hospital, School of Public Health, MOE Key Laboratory of Geriatric Diseases and Immunology, Medical College of Soochow University, Suzhou, China
| | - Zhong-Yue Liu
- Fourth Affiliated Hospital, School of Public Health, MOE Key Laboratory of Geriatric Diseases and Immunology, Medical College of Soochow University, Suzhou, China
| | - Wei Jiang
- Department of Hepatopancreatobiliary Surgery, Ningbo Medical Centre Lihuili Hospital, Ningbo University, Ningbo, China
| | - Zheng Zhang
- Centre of Child Health Management, Children's Hospital of Soochow University, Suzhou, China
| | - Zhengbao Zhu
- Department of Epidemiology, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
| | - Li-Qiang Qin
- Fourth Affiliated Hospital, School of Public Health, MOE Key Laboratory of Geriatric Diseases and Immunology, Medical College of Soochow University, Suzhou, China
| | - Guo-Chong Chen
- Fourth Affiliated Hospital, School of Public Health, MOE Key Laboratory of Geriatric Diseases and Immunology, Medical College of Soochow University, Suzhou, China.
| |
Collapse
|
7
|
Park JH, Cho MH, Lee HS, Shim YS. No, either or both parents with metabolic syndrome: comparative study of its impact on sons and daughters. Front Endocrinol (Lausanne) 2025; 16:1518212. [PMID: 40313487 PMCID: PMC12043446 DOI: 10.3389/fendo.2025.1518212] [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: 10/28/2024] [Accepted: 03/27/2025] [Indexed: 05/03/2025] Open
Abstract
Background Metabolic syndrome (MetS) is known to have parental influence on children's metabolic health, increasing the risk for the cardiometabolic diseases. However, the extent of the association and its sex-specific differences remain unclear. Objective This retrospective, comparative study aimed to investigate the influence of parental MetS on their offspring, utilizing data from the Korea National Health and Nutritional Examination Survey. Methods The study population was narrowed to 5,245 participants. Each sex was further divided into three groups: children with neither parent having MetS were labeled as "No," those with only one affected parent was labeled as "Either," and those with both parents affected were labeled as "Both." Analysis of covariance and multiple regression analysis were used to compare the cardiometabolic risk factors among the three groups. Results Children with one or both parents affected by MetS had significantly higher waist circumference, blood pressure, triglycerides, and fasting glucose levels. These associations were more profound in boys than in girls. Conclusion Metabolic risk factors were more strongly associated with parental MetS in boys than in girls. The number of parental MetS cases appeared to have a proportional impact on metabolic components in boys, unlike the variable results observed in girls. These results emphasize the need for targeted interventions in families with a history of MetS.
Collapse
Affiliation(s)
| | | | | | - Young Suk Shim
- Department of Pediatrics, Ajou University Hospital, Ajou University School of
Medicine, Suwon, Republic of Korea
| |
Collapse
|
8
|
Cruz AGD, Santos JDMD, Alves EDS, Santos ARMD, Trinca BF, Camargo FND, Bovolin GF, Camporez JP. Metabolic effects of late-onset estradiol replacement in high-fat-fed ovariectomized mice. Curr Res Physiol 2025; 8:100144. [PMID: 40331103 PMCID: PMC12051062 DOI: 10.1016/j.crphys.2025.100144] [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: 11/06/2024] [Revised: 03/27/2025] [Accepted: 04/07/2025] [Indexed: 05/08/2025] Open
Abstract
Background Decreased estrogen levels in postmenopausal women negatively impact metabolic health. It is known that estradiol (E2) replacement can reverse this condition. However, there is no consensus on whether the effects mediated by E2 depend on the starting time of E2 replacement after menopause. We aimed to investigate the effects of different onset E2 treatments on glucose tolerance and metabolic parameters in high-fat-fed ovariectomized mice. Material and methods Eight-week-old female C57BL/6J mice were divided into three groups: SHAM, OVX, and E2, to evaluate three different time points of E2 replacement after ovariectomy: early (after 4 weeks), intermediate (after 12 weeks), and late replacement (after 20 weeks). E2 groups received treatment through subcutaneous pellets. Results E2 replacement improved the parameters analyzed independently of the time since ovariectomy, reducing body weight gain and fat mass, as well as increasing the percentage of lean mass. Glucose intolerance, fasting insulin, HOMA-IR, and cholesterol levels were also reduced after treatment with E2. In the liver, there was a decrease in triacylglycerol (TAG) deposition, with no difference in the expression of SREBP1 and ERα proteins. In the muscle, there was a decrease in TAG deposition. In periuterine adipose tissue, there was an increase in the expression of SREBP1, FASN, and SCD, with no difference in the expression of ERα. Conclusions Our findings reinforce the critical role of E2 in regulating both glucose and lipid metabolism and indicate that E2 action on metabolic health was not dependent on time since ovariectomy for the parameters analyzed.
Collapse
Affiliation(s)
| | | | - Ester dos Santos Alves
- Department of Physiology, Ribeirão Preto School of Medicine, University of São Paulo, Brazil
| | | | - Bruna Fantini Trinca
- Department of Physiology, Ribeirão Preto School of Medicine, University of São Paulo, Brazil
| | - Felipe Nunes de Camargo
- Department of Physiology, Ribeirão Preto School of Medicine, University of São Paulo, Brazil
| | | | - João Paulo Camporez
- Department of Physiology, Ribeirão Preto School of Medicine, University of São Paulo, Brazil
| |
Collapse
|
9
|
Guo X, Li H, Zhu B, Wang X, Xu Q, Aquino E, Koo M, Li Q, Cai J, Glaser S, Wu C. HFD feeding for seven months abolishes STING disruption-driven but not female sex-based protection against hepatic steatosis and inflammation in mice. J Nutr Biochem 2025; 135:109770. [PMID: 39284534 PMCID: PMC11620956 DOI: 10.1016/j.jnutbio.2024.109770] [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: 07/02/2024] [Revised: 09/07/2024] [Accepted: 09/11/2024] [Indexed: 10/06/2024]
Abstract
Stimulator of interferon genes (STING) is positively correlated with the degrees of liver inflammation in human metabolic dysfunction-associated steatotic liver disease (MASLD). In addition, STING disruption alleviates MASLD in mice fed a high-fat diet (HFD) for 3 months (3-m-HFD). Here we investigated the role of the duration of dietary feeding in regulating MASLD in mice and explored the involvement of STING in sex differences in MASLD. Both male and female STING-disrupted (STINGgt) and wild-type C57BL/6J mice were fed an HFD for 3 or 7 months (7-m-HFD). Additionally, female STINGgt mice upon ovariectomy (OVX) and 3-m-HFD were analyzed for MASLD. Upon 3-m-HFD, STINGgt mice exhibited decreased severity of MASLD compared to control. However, upon 7-m-HFD, STINGgt mice were comparable with wild-type mice in body weight, fat mass, and MASLD. Regarding regulating the liver RNA transcriptome, 7-m-HFD increased the expression of genes indicating proinflammatory activation of various liver cells. Interestingly, the severity of MASLD in female mice was much lighter than in male mice, regardless of STING disruption. Upon OVX, female STINGgt mice showed significantly increased severity of MASLD relative to sham control but were comparable with male STINGgt mice. Upon treatment with 17-beta estradiol (E2), hepatocytes revealed decreased fat deposition while macrophages displayed decreases in lipopolysaccharide-induced phosphorylation of Nfkb p65 and Jnk p46 independent of STING. These results suggest that 7-m-HFD, without altering female sex-based protection, abolishes STING disruption-driven protection of MASLD, likely through causing proinflammatory activation of multiple types of liver cells to offset the effect of STING disruption.
Collapse
Affiliation(s)
- Xinlei Guo
- Department of Nutrition, Texas A&M University, College Station, Texas, USA
| | - Honggui Li
- Department of Nutrition, Texas A&M University, College Station, Texas, USA
| | - Bilian Zhu
- Department of Nutrition, Texas A&M University, College Station, Texas, USA
| | - Xiaoxiao Wang
- Department of Nutrition, Texas A&M University, College Station, Texas, USA
| | - Qian Xu
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas, USA
| | - Eduardo Aquino
- Department of Nutrition, Texas A&M University, College Station, Texas, USA
| | - Minji Koo
- Department of Nutrition, Texas A&M University, College Station, Texas, USA
| | - Qingsheng Li
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - James Cai
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas, USA
| | - Shannon Glaser
- Department of Medical Physiology, Texas A&M University College of Medicine, Byran, Texas, USA.
| | - Chaodong Wu
- Department of Nutrition, Texas A&M University, College Station, Texas, USA.
| |
Collapse
|
10
|
Shirazpour S, Khaksari M, Gaini AA, Bashiri H, Khoramipour K, Rafie F. Can resisted swimming exercise substitute for the protective effects of estrogen on cardiometabolic risk factors in obese postmenopausal rat model? IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2025; 28:718-727. [PMID: 40343300 PMCID: PMC12057744 DOI: 10.22038/ijbms.2025.82005.17745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 10/30/2024] [Indexed: 05/11/2025]
Abstract
Objectives Following our previous studies on the anti-obesity and cardioprotective effects of 17-beta estradiol (E2), this study was designed to determine the effects of Resisted swimming (RSW) training and E2 (alone and in combination) on cardiometabolic risk factors in an obese postmenopausal rat model. Materials and Methods Female ovariectomized rats (OVX) were given a standard diet (SD) or a 60% high-fat diet (HFD) for 16 weeks and were divided into two groups: SD and HFD. The rats were divided into ten groups to assess the effects of 8 weeks of E2 (1 mg/kg, IP) administration and RSW (5 days a week) on cardiometabolic risk factors. Parameters including body weight, BMI, visceral fat, blood glucose (BG), and cardiac oxidative stress were assessed 72 hr after the last swimming session. Results HFD increased body weight, BMI, visceral fat, and BG levels in OVX rats. Additionally, it negatively affected the lipid profile and cardiac oxidative stress, but both E2 and RSW reduced these parameters in HFD-fed OVX rats. Although RSW and E2 equally prevented these changes, swimming was more effective than estrogen in increasing HDL levels in the SD group. The combination of E2 and RSW had a more significant effect on modulating glucose, TAC, TG, and HDL indices than the individual treatments. Conclusion Overall, RSW ameliorates cardiometabolic risk factors in postmenopausal conditions caused by obesity, probably by modulating cardiac oxidative stress. It is also an effective non-pharmacological treatment for E2 substitution.
Collapse
Affiliation(s)
- Sara Shirazpour
- Department of Physiology, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Khaksari
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Abbas Ali Gaini
- Department of Exercise Physiology, University of Tehran, Tehran, Iran
| | - Hamideh Bashiri
- Institute of Neuropharmacology, Neuroscience Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Kayvan Khoramipour
- Department of Physiology, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), 47012 Valladolid, Spain
| | - Forouzan Rafie
- Institute of Neuropharmacology, Neuroscience Research Center, Kerman University of Medical Sciences, Kerman, Iran
- Emory University School of Medicine Division of Geriatrics and Gerontology, Atlanta, Georgia, USA
| |
Collapse
|
11
|
Sivasinprasasn S, Chattipakorn K, Pratchayasakul W, Chattipakorn SC, Chattipakorn N. N-Acetylcysteine enhances low-dose estrogen efficacy against ischemia-reperfusion injury in estrogen-deprived obese insulin-resistant rats. Menopause 2025; 32:81-90. [PMID: 39626181 DOI: 10.1097/gme.0000000000002452] [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: 12/28/2024]
Abstract
OBJECTIVES Postmenopausal women are at higher risk of metabolic syndrome and cardiovascular disease, which are aggravated by obesity. Although estrogen provides cardiometabolic protection, chronic high-dose treatment could be harmful. This study investigated the efficacy of combined N-acetylcysteine (NAC) and low-dose estrogen treatment against cardiometabolic dysfunction in female estrogen-deprived obese rats with cardiac ischemia-reperfusion (I/R) injury. METHODS Bilateral ovariectomized (O) female Wistar rats were fed a high-fat diet (H) for 12 weeks. Then, rats were treated for 4 weeks with one of the following: vehicle (OH; sesame oil), regular-dose estrogen (E; 50 μg/kg/d), low-dose estrogen (e; 25 μg/kg/d), NAC (N; 100 mg/kg/d), or combined low-dose estradiol with NAC (eN). All rats then underwent cardiac I/R injury, and the left ventricle (LV) function and mitochondrial function were investigated (n = 6/group). Statistical analysis was performed by one-way ANOVA followed by Fisher's least significant difference post hoc test. RESULTS Body weight, visceral fat, plasma glucose, and plasma cholesterol were significantly increased with impaired LV function and heart rate variability in OH rats. OH-E rats had decreased plasma insulin and Homeostatic Model Assessment for Insulin Resistance index. Both OH-E and OH-eN rats had similarly improved heart rate variability and LV function. During cardiac I/R, OH-E and OH-eN rats had preserved left ventricular ejection fraction, stroke volume, and attenuated arrhythmias. Impaired cardiac mitochondrial function and infarct size were similarly reduced in OH-E and OH-eN rats. CONCLUSIONS Combined NAC and low-dose estrogen treatment shares similar efficacy as regular-dose estrogen in attenuating cardiac dysfunction, cardiac mitochondrial dysfunction, and protecting the heart against I/R injury in estrogen-deprived obese insulin-resistant rats.
Collapse
Affiliation(s)
| | - Kenneth Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | | | | | | |
Collapse
|
12
|
Bin Pan, Xie Y, Shao W, Fang X, Han D, Li J, Hong X, Tu W, Shi J, Yang M, Tian F, Xia M, Hu J, Ren J, Kan H, Xu Y, Li W. Prenatal exposure to PM 2.5 disturbs the glucose metabolism of offspring fed with high-fat diet in a gender-dependent manner. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 288:117404. [PMID: 39615301 DOI: 10.1016/j.ecoenv.2024.117404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 11/07/2024] [Accepted: 11/22/2024] [Indexed: 12/09/2024]
Abstract
Studies have shown that maternal exposure to PM2.5 could potentially disrupt glucose and lipid metabolism in offspring supplied with high-fat diet, yet whether this effect is gender-dependent or not and the underlying biological mechanisms are not well understood. In our current research, female ICR mice were exposed to filtered air (FA) or concentrated ambient PM2.5 (CAP) before and during pregnancy. The offspring mice were fed with control diet (CD) or high-fat diet (HFD) for 9 weeks, and their metabolic conditions were analyzed. Our findings reveal that maternal exposure to PM2.5 induced glucose intolerance and insulin resistance in female offspring fed with HFD but not in males. Specifically, hepatic insulin resistance as indicated by significantly decreased AKT phosphorylation (p-AKT) level, changed liver structure as indicated by increased ballooning and steatosis based on H&E staining images, and impaired liver function as indicated by up-regulated ALT activity were observed in HFD-fed female offspring from CAP-exposed mothers in comparison to those from FA-exposed ones. Further analysis indicated that these impacts of prenatal PM2.5 exposure on glucose metabolism in offspring may result from disturbed gluconeogenesis and induced inflammatory response in liver. Our research underscores that prenatal PM2.5 exposure induces glucose metabolism abnormalities in offspring fed with HFD in a gender-dependent manner, and the liver potentially serves as a key player in mediating these effects of maternal PM2.5 exposure.
Collapse
Affiliation(s)
- Bin Pan
- NHC Key Lab of Reproduction Regulation, Shanghai Engineering Research Center of Reproductive Health Drugs and Devices, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of pharmacy, Fudan University, Shanghai 200237, China; Department of Environmental Health, School of Public Health, Fudan University, Shanghai 200032, China
| | - Yuanting Xie
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai 200032, China
| | - Wenpu Shao
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai 200032, China
| | - Xinyi Fang
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai 200032, China
| | - Dongyang Han
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai 200032, China
| | - Jingyu Li
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai 200032, China
| | - Xiaoqing Hong
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai 200032, China
| | - Wenyue Tu
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai 200032, China
| | - Jiayi Shi
- NHC Key Lab of Reproduction Regulation, Shanghai Engineering Research Center of Reproductive Health Drugs and Devices, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of pharmacy, Fudan University, Shanghai 200237, China
| | - Mingjun Yang
- NHC Key Lab of Reproduction Regulation, Shanghai Engineering Research Center of Reproductive Health Drugs and Devices, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of pharmacy, Fudan University, Shanghai 200237, China
| | - Fang Tian
- NHC Key Lab of Reproduction Regulation, Shanghai Engineering Research Center of Reproductive Health Drugs and Devices, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of pharmacy, Fudan University, Shanghai 200237, China
| | - Minjie Xia
- NHC Key Lab of Reproduction Regulation, Shanghai Engineering Research Center of Reproductive Health Drugs and Devices, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of pharmacy, Fudan University, Shanghai 200237, China
| | - Jingying Hu
- NHC Key Lab of Reproduction Regulation, Shanghai Engineering Research Center of Reproductive Health Drugs and Devices, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of pharmacy, Fudan University, Shanghai 200237, China
| | - Jianke Ren
- NHC Key Lab of Reproduction Regulation, Shanghai Engineering Research Center of Reproductive Health Drugs and Devices, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of pharmacy, Fudan University, Shanghai 200237, China
| | - Haidong Kan
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai 200032, China
| | - Yanyi Xu
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai 200032, China.
| | - Weihua Li
- NHC Key Lab of Reproduction Regulation, Shanghai Engineering Research Center of Reproductive Health Drugs and Devices, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of pharmacy, Fudan University, Shanghai 200237, China.
| |
Collapse
|
13
|
Moraes FCAD, Moro LD, Souza MEC, Rodrigues ALSDO, Sano VKT, Barbosa BF, Pacheco LG, Cunha DF, Queiroz OLD, Souza DDSMD, Feio D, Stecca C, Burbano RMR. Prevalence of cardiometabolic outcomes in women who underwent salpingo-oophorectomy to prevent hereditary breast and ovarian cancer: a meta-analysis. Fam Cancer 2024; 24:5. [PMID: 39546060 DOI: 10.1007/s10689-024-00431-x] [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: 01/25/2024] [Accepted: 09/20/2024] [Indexed: 11/17/2024]
Abstract
Risk reduction salpingo-oophorectomy (RRSO) is usually performed in women with hereditary breast and ovarian cancer (HBOC) syndrome for BRCA1 and BRCA2 gene mutation carriers, resulting in surgical menopause, which is more associated with a high risk of cardiovascular and metabolic disease than in premenopausal and natural menopausal women. This study assessed the prevalence of cardiovascular and metabolic outcomes in women who underwent salpingo-oophorectomy as a preventive measure against HBOC. This meta-analysis assessed prevalence rates for four metabolic/cardiovascular conditions: myocardial infarction, hypertension, hypercholesterolemia, and type 2 diabetes mellitus. DerSimonian and Laird random-effects models were applied to all analyses, with 95% confidence interval (CI). Heterogeneity was assessed with I². We used OpenMeta Analyst software for statistical analysis. A total of five retrospective studies and one observational study involving 1,320 patients were included. The average body mass index (BMI) was 25.97 kg/m2 and the average waist circumference was 87.94 cm. The analysis across a mean 4.94-year follow-up revealed prevalence rates for acute myocardial infarction of 1.5% (95% CI 0.3-2.7; P = 0.077; I²=56.25%), hypertension of 28% (95% CI 6.9-49.1; P < 0.001; I2 = 98.42%), hypercholesterolemia of 27.2% (95% CI 6.8-47.6; P < 0.001; I²=98.67%), and type 2 diabetes mellitus of 3.3% (95% CI 1.1-5.5; P < 0.001; I²=82.44%). Our findings suggest that there is no marked increase in cardiovascular risk among women with HBOC undergoing RRSO.
Collapse
Affiliation(s)
| | - Lucca Dal Moro
- School of Medicine, Federal University of Pará, Belém, PA, 66073-005, Brazil
| | | | | | | | - Bárbara Ferraz Barbosa
- Department of Medicine, University of Aquino Bolivia, Santa Cruz de la Sierra, SC, 0701, Bolivia
| | - Lucas Gama Pacheco
- School of Medicine, Federal University of Pará, Belém, PA, 66073-005, Brazil
| | | | | | | | - Danielle Feio
- School of Medicine, Federal University of Pará, Belém, PA, 66073-005, Brazil
| | - Carlos Stecca
- Mackenzie Evangelical University Hospital, Curitiba, Paraná, 80730-150, Brazil
| | | |
Collapse
|
14
|
Zhu BT, Liao QQ, Tian HY, Yu DJ, Xie T, Sun XL, Zhou XM, Han YX, Zhao YJ, El-Kassas M, Liu XX, Sun XD, Zhang YY. Estrogen: the forgotten player in metaflammation. Front Pharmacol 2024; 15:1478819. [PMID: 39575382 PMCID: PMC11578702 DOI: 10.3389/fphar.2024.1478819] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 10/21/2024] [Indexed: 11/24/2024] Open
Abstract
Metaflammation is low-grade inflammation triggered by chronic metabolic imbalance and caused by dysregulated metabolites in metabolic inflammatory syndrome (MIS), which includes four diseases: obesity, type 2 diabetes mellitus (T2DM), atherosclerosis (AS), and nonalcoholic fatty liver diseases (NAFLD, recently proposed to be replaced by metabolic dysfunction-associated steatotic liver disease, MASLD). These diseases exhibit apparent sex dimorphism as regards MIS. Estrogen not only plays a crucial role in gender differences in adults but also possesses an anti-inflammatory effect on many metabolic diseases. In this study, we present a prediction of the differential proteins and signal transduction of estrogen in MIS through network pharmacology and review the validated studies on obesity, T2DM, AS, and NAFLD. Subsequently, we compared them to obtain valuable targets, identify current gaps, and provide perspectives for future research on the mechanisms of estrogen in metaflammation.
Collapse
Affiliation(s)
- Bao-Ting Zhu
- West China School of Pharmacy, West China School of Basic Medical Sciences and Forensic Medicine, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Qing-Qing Liao
- West China School of Pharmacy, West China School of Basic Medical Sciences and Forensic Medicine, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Hai-Ying Tian
- West China School of Pharmacy, West China School of Basic Medical Sciences and Forensic Medicine, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Dao-Jiang Yu
- West China School of Pharmacy, West China School of Basic Medical Sciences and Forensic Medicine, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, China
| | - Teng Xie
- West China School of Pharmacy, West China School of Basic Medical Sciences and Forensic Medicine, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Xi-Lu Sun
- West China School of Pharmacy, West China School of Basic Medical Sciences and Forensic Medicine, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Xin-Meng Zhou
- West China School of Pharmacy, West China School of Basic Medical Sciences and Forensic Medicine, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Ying-Xuan Han
- West China School of Pharmacy, West China School of Basic Medical Sciences and Forensic Medicine, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yu-Jie Zhao
- Medical College, Tibet University, Lasa, China
| | - Mohamed El-Kassas
- Endemic Medicine Department, Faculty of Medicine, Helwan University, Cairo, Egypt
- Liver Disease Research Center, College of Medicine, King Saud University, Riyadh, Saudi Arabia
- Steatotic Liver Disease Study Foundation in Middle East and North Africa (SLMENA), Cairo, Egypt
| | - Xiu-Xiu Liu
- West China School of Pharmacy, West China School of Basic Medical Sciences and Forensic Medicine, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Xiao-Dong Sun
- West China School of Pharmacy, West China School of Basic Medical Sciences and Forensic Medicine, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
- Medical College, Tibet University, Lasa, China
| | - Yuan-Yuan Zhang
- West China School of Pharmacy, West China School of Basic Medical Sciences and Forensic Medicine, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, China
| |
Collapse
|
15
|
Kusumo LE, Gilley-Connor KR, Johnson MG, Hall GM, Gillett AE, McCready RG, Vichaya EG. Hyperglycemia sensitizes female mice to stress-induced depressive-like behavior in an inflammation-independent manner. Psychoneuroendocrinology 2024; 169:107151. [PMID: 39098101 DOI: 10.1016/j.psyneuen.2024.107151] [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: 04/30/2024] [Revised: 07/23/2024] [Accepted: 07/27/2024] [Indexed: 08/06/2024]
Abstract
BACKGROUND Depression is a multifaceted disorder that represents one of the most common causes of disability. The risk for developing depression is increased in women and among individuals with chronic diseases. For example, individuals in the United States with diabetes mellitus (DM) are at a twofold increased risk of developing depression compared to the general population and approximately one-quarter of women with diabetes have comorbid depression. The neurobiological mechanisms underlying this association between diabetes and depression is not fully understood and is particularly under-investigated in female models. We sought to explore the role of neuroinflammation in diabetes-induced depression in a female mouse model of hyperglycemia. METHODS To this end, we utilized female C57BL/6 J mice to (1) characterize the depressive-like symptoms in response to 75 mg/kg/day dose of streptozotocin (STZ) over 5 days, a dose reported to induce hyperglycemia in female mice (n=20), (2) determine if female hyperglycemic mice are sensitized to unpredictable chronic mild stress (UCMS)-induced depressive-like behavior and neuroinflammation (n=28), and (3) investigate if female hyperglycemic mice are primed to respond to a subthreshold dose of lipopolysaccharide (LPS), an acute inflammatory challenge (n=21). RESULTS Our results demonstrate that female mice exhibit robust hyperglycemia but limited evidence of depressive-like behavior in response to 75 mg/kg STZ. Additionally, we observe that healthy female mice have limited response to our stress protocol; however, hyperglycemic mice display increased stress-sensitivity as indicated by increased immobility in the forced swim test. While STZ mice show evidence of mild neuroinflammation, this effect was blunted by stress. Further, STZ mice failed to display a sensitization to inflammation-induced depressive-like behavior. CONCLUSION We interpret this data to indicate that while STZ-induced hyperglycemia does increase vulnerability to stress-induced depressive-like behavior, this effect is not a consequence of neuroinflammatory priming. Future studies will seek to better understand the mechanisms underlying this sensitization.
Collapse
Affiliation(s)
- Laura E Kusumo
- Department of Psychology & Neuroscience, Baylor University, Waco, TX 76798, United States
| | - Kayla R Gilley-Connor
- Department of Psychology & Neuroscience, Baylor University, Waco, TX 76798, United States
| | - Madilyn G Johnson
- Department of Psychology & Neuroscience, Baylor University, Waco, TX 76798, United States
| | - Grace M Hall
- Department of Psychology & Neuroscience, Baylor University, Waco, TX 76798, United States
| | - Avery E Gillett
- Department of Psychology & Neuroscience, Baylor University, Waco, TX 76798, United States
| | - Riley G McCready
- Department of Psychology & Neuroscience, Baylor University, Waco, TX 76798, United States
| | - Elisabeth G Vichaya
- Department of Psychology & Neuroscience, Baylor University, Waco, TX 76798, United States.
| |
Collapse
|
16
|
Feng B, Tang P, He S, Peng Z, Mo Y, Zhu L, Wei Q. Associations between antimony exposure and glycated hemoglobin levels in adolescents aged 12-19 years: results from the NHANES 2013-2016. Front Public Health 2024; 12:1439034. [PMID: 39484344 PMCID: PMC11524935 DOI: 10.3389/fpubh.2024.1439034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 09/30/2024] [Indexed: 11/03/2024] Open
Abstract
Objective This study aimed to investigate the association between antimony (Sb) exposure and glycated hemoglobin (HbA1c) levels in adolescents. Methods A cross-sectional study of 751 adolescents aged 12-19 years was conducted via the National Health and Nutrition Examination Survey (NHANES, 2013-2016). Survey-weighted linear regression and restricted cubic spline (RCS) analyses were applied to evaluate the relationship of urinary Sb exposure with HbA1c. Results A significant relationship was observed between urinary Sb concentrations and HbA1c levels (percent change: 0.93; 95% CI: 0.42, 1.45) after full adjustment. After converting urinary Sb levels to a categorical variable by tertiles (T1-T3), the highest quantile was associated with a significant increase in HbA1c (percent change: 1.45; 95% CI: 0.38, 2.53) compared to T1. The RCS models showed a monotonically increasing relationship of urinary Sb with HbA1c. Subgroup analyses revealed a sex-specific relationship between urinary Sb exposure and HbA1c with a significant positive association in males and a non-significant positive association in females. Sensitivity analyses further confirmed the relationship between urinary Sb and HbA1c, even after excluding participants who were overweight or obese (percent change: 1.58%, 95% CI: 0.88, 2.28) and those with serum cotinine levels ≥ 1 ng/mL (percent change: 1.14%, 95% CI: 0.49, 1.80). Conclusion Our findings indicated that increased Sb exposure may correlate with higher HbA1c levels, especially in male adolescents. More studies are needed to further explore and validate the potential mechanisms.
Collapse
Affiliation(s)
- Baoying Feng
- Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
- Birth Defects Research Laboratory, Birth Defects Prevention and Control Institute of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
- Guangxi Clinical Research Center for Pediatric Disease, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Peng Tang
- Department of Epidemiology, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Sheng He
- Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
- Birth Defects Research Laboratory, Birth Defects Prevention and Control Institute of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
- Guangxi Clinical Research Center for Pediatric Disease, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
- Birth Defects Research Laboratory, Guangxi Key Laboratory of Reproductive Health and Birth Defect Prevention, Nanning, Guangxi, China
| | - Zhenren Peng
- Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
- Birth Defects Research Laboratory, Birth Defects Prevention and Control Institute of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
- Guangxi Clinical Research Center for Pediatric Disease, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
- Birth Defects Research Laboratory, Guangxi Key Laboratory of Reproductive Health and Birth Defect Prevention, Nanning, Guangxi, China
| | - Yan Mo
- Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
- Birth Defects Research Laboratory, Birth Defects Prevention and Control Institute of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
- Guangxi Clinical Research Center for Pediatric Disease, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Liqiong Zhu
- Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Qiufen Wei
- Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
- Birth Defects Research Laboratory, Birth Defects Prevention and Control Institute of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
- Guangxi Clinical Research Center for Pediatric Disease, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| |
Collapse
|
17
|
De Vos K, Mavrogiannis A, Wolters JC, Schlenner S, Wierda K, Cortés Calabuig Á, Chinnaraj R, Dermesrobian V, Armoudjian Y, Jacquemyn M, Corthout N, Daelemans D, Annaert P. Tankyrase1/2 inhibitor XAV-939 reverts EMT and suggests that PARylation partially regulates aerobic activities in human hepatocytes and HepG2 cells. Biochem Pharmacol 2024; 227:116445. [PMID: 39053638 DOI: 10.1016/j.bcp.2024.116445] [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: 12/06/2023] [Revised: 07/16/2024] [Accepted: 07/18/2024] [Indexed: 07/27/2024]
Abstract
The maintenance of a highly functional metabolic epithelium in vitro is challenging. Metabolic impairments in primary human hepatocytes (PHHs) over time is primarily due to epithelial-to-mesenchymal transitioning (EMT). The immature hepatoma cell line HepG2 was used as an in vitro model to explore strategies for enhancing the hepatic phenotype. The phenotypic characterization includes measuring the urea cycle, lipid storage, tricarboxylic acid-related metabolites, reactive oxygen species, endoplasmic reticulum calcium efflux, mitochondrial membrane potentials, oxygen consumptions rate, and CYP450 biotransformation capacity. Expression studies were performed with transcriptomics, co-immunoprecipitation and proteomics. CRISPR/Cas9 was also employed to genetically engineer HepG2 cells. After confirming that PHHs develop an EMT phenotype, expression of tankyrase1/2 was found to increase over time. EMT was reverted when blocking tankyrases1/2-dependent poly-ADP-ribosylation (PARylation) activity, by biochemical and genetic perturbation. Wnt/β-catenin inhibitor XAV-939 blocks tankyrase1/2 and treatment elevated several oxygen-consuming reactions (electron-transport chain, OXHPOS, CYP450 mono-oxidase activity, phase I/II xenobiotic biotransformation, and prandial turnover), suggesting that cell metabolism was enhanced. Glutathione-dependent redox homeostasis was also significantly improved in the XAV-939 condition. Oxygen consumption rate and proteomics experiments in tankyrase1/2 double knockout HepG2 cells then uncovered PARylation as master regulator of aerobic-dependent cell respiration. Furthermore, novel tankyrase1/2-dependent PARylation targets, including mitochondrial DLST, and OGDH, were revealed. This work exposed a new mechanistic framework by linking PARylation to respiration and metabolism, thereby broadening the current understanding that underlies these vital processes. XAV-939 poses an immediate and straightforward strategy to improve aerobic activities, and metabolism, in (immature) cell cultures.
Collapse
Affiliation(s)
- Kristof De Vos
- Laboratory of Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Adamantios Mavrogiannis
- Adaptive Immunology, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
| | - Justina Clarinda Wolters
- Section Systems Medicine of Metabolism and Signaling, Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen 9713 AV, the Netherlands
| | - Susan Schlenner
- Adaptive Immunology, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
| | - Keimpe Wierda
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Electrophysiology Unit, VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium
| | | | - Reena Chinnaraj
- KU Leuven Flow and Mass Cytometry Facility, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
| | - Vera Dermesrobian
- KU Leuven Flow and Mass Cytometry Facility, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
| | | | - Maarten Jacquemyn
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, KU Leuven, Rega Institute, 3000 Leuven, Belgium
| | - Nikky Corthout
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; VIB Bio Imaging Core, 3000 Leuven, Belgium
| | - Dirk Daelemans
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, KU Leuven, Rega Institute, 3000 Leuven, Belgium
| | - Pieter Annaert
- Laboratory of Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium; BioNotus GCV, 2845 Niel, Belgium.
| |
Collapse
|
18
|
Presta M, Zoratto F, Mulder D, Ottomana AM, Pisa E, Arias Vásquez A, Slattery DA, Glennon JC, Macrì S. Hyperglycemia and cognitive impairments anticipate the onset of an overt type 2 diabetes-like phenotype in TALLYHO/JngJ mice. Psychoneuroendocrinology 2024; 167:107102. [PMID: 38896988 DOI: 10.1016/j.psyneuen.2024.107102] [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: 02/20/2024] [Revised: 04/30/2024] [Accepted: 06/08/2024] [Indexed: 06/21/2024]
Abstract
Type 2 Diabetes mellitus (T2DM) is a metabolic disorder characterized by chronic hyperglycemia, resulting from deficits in insulin secretion, insulin action, or both. Whilst the role of insulin in the peripheral nervous system has been ascertained in countless studies, its role in the central nervous system (CNS) is emerging only recently. Brain insulin has been lately associated with brain disorders like Alzheimer's disease, obsessive compulsive disorder, and attention deficit hyperactivity disorder. Thus, understanding the role of insulin as a common risk factor for mental and somatic comorbidities may disclose novel preventative and therapeutic approaches. We evaluated general metabolism (glucose tolerance, insulin sensitivity, energy expenditure, lipid metabolism, and polydipsia) and cognitive capabilities (attention, cognitive flexibility, and memory), in adolescent, young adult, and adult male and female TALLYHO/JngJ mice (TH, previously reported to constitute a valid experimental model of T2DM due to impaired insulin signaling). Adult TH mice have also been studied for alterations in gut microbiota diversity and composition. While TH mice exhibited profound deficits in cognitive flexibility and altered glucose metabolism, we observed that these alterations emerged either much earlier (males) or independent of (females) a comprehensive constellation of symptoms, isomorphic to an overt T2DM-like phenotype (insulin resistance, polydipsia, higher energy expenditure, and altered lipid metabolism). We also observed significant sex-dependent alterations in gut microbiota alpha diversity and taxonomy in adult TH mice. Deficits in insulin signaling may represent a common risk factor for both T2DM and CNS-related deficits, which may stem from (partly) independent mechanisms.
Collapse
Affiliation(s)
- Martina Presta
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, Rome 00161, Italy; Department of Physiology and Pharmacology, Sapienza University of Rome, Rome 00185, Italy
| | - Francesca Zoratto
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, Rome 00161, Italy
| | - Danique Mulder
- Donders Institute for Brain, Cognition and Behaviour, Departments of Psychiatry and Human Genetics, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Angela Maria Ottomana
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, Rome 00161, Italy; Neuroscience Unit, Department of Medicine, University of Parma, Parma 43100, Italy
| | - Edoardo Pisa
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, Rome 00161, Italy
| | - Alejandro Arias Vásquez
- Donders Institute for Brain, Cognition and Behaviour, Departments of Psychiatry and Human Genetics, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - David A Slattery
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt, Germany
| | - Jeffrey C Glennon
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Ireland
| | - Simone Macrì
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, Rome 00161, Italy.
| |
Collapse
|
19
|
Visa M, Berggren PO. Sex-dependent intra-islet structural rearrangements affecting alpha-to-beta cell interactions lead to adaptive enhancements of Ca 2+ dynamics in prediabetic beta cells. Diabetologia 2024; 67:1663-1682. [PMID: 38814444 PMCID: PMC11343800 DOI: 10.1007/s00125-024-06173-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 04/09/2024] [Indexed: 05/31/2024]
Abstract
AIMS/HYPOTHESIS Prediabetic pancreatic beta cells can adapt their function to maintain normoglycaemia for a limited period of time, after which diabetes mellitus will manifest upon beta cell exhaustion. Understanding sex-specific beta cell compensatory mechanisms and their failure in prediabetes (impaired glucose tolerance) is crucial for early disease diagnosis and individualised treatment. Our aims were as follows: (1) to determine the key time points of the progression from beta cells' functional adaptations to their failure in vivo; and (2) to mechanistically explain in vivo sex-specific beta cell compensatory mechanisms and their failure in prediabetes. METHODS Islets from male and female transgenic Ins1CreERT2-GCaMP3 mice were transplanted into the anterior chamber of the eye of 10- to 12-week-old sex-matched C57BL/6J mice. Recipient mice were fed either a control diet (CD) or western diet (WD) for a maximum of 4 months. Metabolic variables were evaluated monthly. Beta cell cytoplasmic free calcium concentration ([Ca2+]i) dynamics were monitored in vivo longitudinally by image fluorescence of the GCaMP3 reporter islets. Global islet beta cell [Ca2+]i dynamics in line with single beta cell [Ca2+]i analysis were used for beta cell coordination studies. The glucagon receptor antagonist L-168,049 (4 mmol/l) was applied topically to the transplanted eyes to evaluate in vivo the effect of glucagon on beta cell [Ca2+]idynamics. Human islets from non-diabetic women and men were cultured for 24 h in either a control medium or high-fat/high-glucose medium in the presence or absence of the glucagon receptor antagonist L-168,049. [Ca2+]i dynamics of human islets were evaluated in vitro after 1 h exposure to Fura-10. RESULTS Mice fed a WD for 1 month displayed increased beta cell [Ca2+]i dynamics linked to enhanced insulin secretion as a functional compensatory mechanism in prediabetes. Recruitment of inactive beta cells in WD-fed mice explained the improved beta cell function adaptation observed in vivo; this occurred in a sex-specific manner. Mechanistically, this was attributable to an intra-islet structural rearrangement involving alpha cells. These sex-dependent cytoarchitecture reorganisations, observed in both mice and humans, induced enhanced paracrine input from adjacent alpha cells, adjusting the glucose setpoint and amplifying the insulin secretion pathway. When WD feeding was prolonged, female mice maintained the adaptive mechanism due to their intrinsically high proportion of alpha cells. In males, [Ca2+]i dynamics progressively declined subsequent to glucose stimulation while insulin secretion continue to increase, suggesting uncoordinated beta cell function as an early sign of diabetes. CONCLUSIONS/INTERPRETATION We identified increased coordination of [Ca2+]i dynamics as a beta cell functional adaptation mechanisms in prediabetes. Importantly, we uncovered the mechanisms by which sex-dependent beta cell [Ca2+]i dynamics coordination is orchestrated by an intra-islet structure reorganisation increasing the paracrine input from alpha cells on beta cell function. Moreover, we identified reduced [Ca2+]i dynamics coordination in response to glucose as an early sign of diabetes preceding beta cell secretory dysfunction, with males being more vulnerable. Alterations in coordination capacity of [Ca2+]i dynamics may thus serve as an early marker for beta cell failure in prediabetes.
Collapse
Affiliation(s)
- Montse Visa
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden.
| | - Per-Olof Berggren
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden.
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA.
- Tecnológico de Monterrey, Real San Agustín, Mexico.
- West China Hospital, Sichuan University, Chengdu, China.
- School of Biomedical Sciences, Ulster University, Coleraine, UK.
| |
Collapse
|
20
|
Ghoshal K, Luther JM, Pakala SB, Chetyrkin S, Falck JR, Zent R, Wasserman DH, Pozzi A. Epoxygenase Cyp2c44 Regulates Hepatic Lipid Metabolism and Insulin Signaling by Controlling FATP2 Localization and Activation of the DAG/PKCδ Axis. Diabetes 2024; 73:1229-1243. [PMID: 38743615 PMCID: PMC11262046 DOI: 10.2337/db23-0493] [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: 06/21/2023] [Accepted: 05/05/2024] [Indexed: 05/16/2024]
Abstract
Cytochrome P450 epoxygenase Cyp2c44, a murine epoxyeicosatrienoic acid (EET)-producing enzyme, promotes insulin sensitivity, and Cyp2c44-/- mice show hepatic insulin resistance. Because insulin resistance leads to hepatic lipid accumulation and hyperlipidemia, we hypothesized that Cyp2c44 regulates hepatic lipid metabolism. Standard chow diet (SCD)-fed male Cyp2c44-/- mice had significantly decreased EET levels and increased hepatic and plasma lipid levels compared with wild-type mice. We showed increased hepatic plasma membrane localization of the FA transporter 2 (FATP2) and total unsaturated fatty acids and diacylglycerol (DAG) levels. Cyp2c44-/- mice had impaired glucose tolerance and increased hepatic plasma membrane-associated PKCδ and phosphorylated IRS-1, two negative regulators of insulin signaling. Surprisingly, SCD and high-fat diet (HFD)-fed Cyp2c44-/- mice had similar glucose tolerance and hepatic plasma membrane PKCδ levels, suggesting that SCD-fed Cyp2c44-/- mice have reached their maximal glucose intolerance. Inhibition of PKCδ resulted in decreased IRS-1 serine phosphorylation and improved insulin-mediated signaling in Cyp2c44-/- hepatocytes. Finally, Cyp2c44-/- HFD-fed mice treated with the analog EET-A showed decreased hepatic plasma membrane FATP2 and PCKδ levels with improved glucose tolerance and insulin signaling. In conclusion, loss of Cyp2c44 with concomitant decreased EET levels leads to increased hepatic FATP2 plasma membrane localization, DAG accumulation, and PKCδ-mediated attenuation of insulin signaling. Thus, Cyp2c44 acts as a regulator of lipid metabolism by linking it to insulin signaling. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Kakali Ghoshal
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN
| | - James M Luther
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN
| | - Suman B Pakala
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN
| | - Sergei Chetyrkin
- Mass Spectrometry Research Center, Vanderbilt University School of Medicine, Nashville, TN
| | | | - Roy Zent
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN
- Department of Veterans Affairs, Nashville, Nashville, TN
| | - David H Wasserman
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN
| | - Ambra Pozzi
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN
- Department of Veterans Affairs, Nashville, Nashville, TN
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN
| |
Collapse
|
21
|
Tonet NS, da Silva Marçal DF, da Silva FN, Brunetta HS, Mori MADS, Dos Santos GJ, Moreira ELG, Rafacho A. Moderate chronic sleep perturbation impairs glucose and lipid homeostasis in rats. Sleep 2024; 47:zsae118. [PMID: 38788154 DOI: 10.1093/sleep/zsae118] [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: 01/27/2024] [Revised: 04/23/2024] [Indexed: 05/26/2024] Open
Abstract
STUDY OBJECTIVES Sleep deprivation is a potential risk factor for metabolic diseases, including obesity and type 2 diabetes. We evaluated the impacts of moderate chronic sleep deprivation on glucose and lipid homeostasis in adult rats. METHODS Wistar rats (both sexes) were sleep-perturbed daily for 2 hours at the early (06:00-08:00) and the late light cycle (16:00-18:00) five days a week (except weekends) for 4 weeks. RESULTS Sleep perturbation (SP) resulted in reduced body weight gain in both sexes, associated with altered food intake and reduced adiposity. SP did not alter the short- or long-term memories or cause anxiogenic behavior. No major changes were observed in the plasma insulin, leptin, triacylglycerol, non-esterified fatty acids, and blood glucose upon SP. After SP, females exhibited a transitory glucose intolerance, while males became glucose intolerant at the end of the experimental period. Male rats also developed higher insulin sensitivity at the end of the SP protocol. Morphometric analyses revealed no changes in hepatic glycogen deposition, pancreatic islet mass, islet-cell distribution, or adrenal cortex thickness in SP rats from both sexes, except for lower adipocyte size compared with controls. We did not find homogeneous changes in the relative expression of circadian and metabolic genes in muscle or hepatic tissues from the SP rats. CONCLUSIONS Moderate chronic SP reduces visceral adiposity and causes glucose intolerance with a more pronounced impact on male rats, reinforcing the metabolic risks of exposure to sleep disturbances.
Collapse
Affiliation(s)
- Natália Stinghen Tonet
- Laboratory of Investigation in Chronic Diseases (LIDoC), Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Brazil
- Graduate Program in Biochemistry, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Danilo Francisco da Silva Marçal
- Laboratory of Investigation in Chronic Diseases (LIDoC), Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Flavia Natividade da Silva
- Laboratory of Investigation in Chronic Diseases (LIDoC), Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Henver Simionato Brunetta
- Department of Biochemistry and Tissue Biology, Institute of Biology, Universidade Estadual de Campinas, Campinas, Brazil
| | - Marcelo Alves da Silva Mori
- Department of Biochemistry and Tissue Biology, Institute of Biology, Universidade Estadual de Campinas, Campinas, Brazil
| | - Gustavo Jorge Dos Santos
- Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Eduardo Luiz Gasnhar Moreira
- Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Alex Rafacho
- Laboratory of Investigation in Chronic Diseases (LIDoC), Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Brazil
- Graduate Program in Biochemistry, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Brazil
- Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Brazil
| |
Collapse
|
22
|
Anunciado-Koza RVP, Yin H, Bilodeau CL, Cooke D, Ables GP, Ryzhov S, Koza RA. Interindividual differences of dietary fat-inducible Mest in white adipose tissue of C57BL/6J mice are not heritable. Obesity (Silver Spring) 2024; 32:1144-1155. [PMID: 38616328 PMCID: PMC11132930 DOI: 10.1002/oby.24020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 02/01/2024] [Accepted: 02/20/2024] [Indexed: 04/16/2024]
Abstract
OBJECTIVE Differences in white adipose tissue (WAT) expression of mesoderm-specific transcript (Mest) in C57BL6/J mice fed a high-fat diet (HFD) are concomitant with and predictive for the development of obesity. However, the basis for differences in WAT Mest among mice is unknown. This study investigated whether HFD-inducible WAT Mest, as well as susceptibility to obesity, is transmissible from parents to offspring. METHODS WAT biopsies of mice fed an HFD for 2 weeks identified parents with low and high WAT Mest for breeding. Obesity phenotypes, WAT Mest, hepatic gene expression, and serum metabolites were determined in offspring fed an HFD for 2 weeks. RESULTS Offspring showed no heritability of obesity or WAT Mest phenotypes from parents but did show hepatic and serum metabolite changes consistent with their WAT Mest. Importantly, retired male breeders showed WAT Mest expression congruent with initial WAT biopsies even though HFD exposure occurred early in life. CONCLUSIONS Disparity of HFD-induced Mest in mice is not heritable but, rather, is reestablished during each generation and remains fixed from an early age to adulthood. Short-term HFD feeding reveals variation of WAT Mest expression within isogenic mice that is positively associated with the development of obesity.
Collapse
Affiliation(s)
| | - Haifeng Yin
- MaineHealth Institute for Research, Scarborough, Maine, USA
| | | | - Diana Cooke
- Orentreich Foundation for the Advancement of Science, Inc., Cold Spring, New York, USA
| | - Gene P. Ables
- Orentreich Foundation for the Advancement of Science, Inc., Cold Spring, New York, USA
| | - Sergey Ryzhov
- MaineHealth Institute for Research, Scarborough, Maine, USA
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, Maine, USA
- Department of Medicine, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Robert A. Koza
- MaineHealth Institute for Research, Scarborough, Maine, USA
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, Maine, USA
- Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| |
Collapse
|
23
|
Croft AJ, Kelly C, Chen D, Haw TJ, Balachandran L, Murtha LA, Boyle AJ, Sverdlov AL, Ngo DTM. Sex-based differences in short- and longer-term diet-induced metabolic heart disease. Am J Physiol Heart Circ Physiol 2024; 326:H1219-H1251. [PMID: 38363215 PMCID: PMC11381029 DOI: 10.1152/ajpheart.00467.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 01/30/2024] [Accepted: 02/12/2024] [Indexed: 02/17/2024]
Abstract
Sex-based differences in the development of obesity-induced cardiometabolic dysfunction are well documented, however, the specific mechanisms are not completely understood. Obesity has been linked to dysregulation of the epitranscriptome, but the role of N6-methyladenosine (m6A) RNA methylation has not been investigated in relation to the sex differences during obesity-induced cardiac dysfunction. In the current study, male and female C57BL/6J mice were subjected to short- and long-term high-fat/high-sucrose (HFHS) diet to induce obesogenic stress. Cardiac echocardiography showed males developed systolic and diastolic dysfunction after 4 mo of diet, but females maintained normal cardiac function despite both sexes being metabolically dysfunctional. Cardiac m6A machinery gene expression was differentially regulated by duration of HFHS diet in male, but not female mice, and left ventricular ejection fraction correlated with RNA machinery gene levels in a sex- and age-dependent manner. RNA-sequencing of cardiac transcriptome revealed that females, but not males may undergo protective cardiac remodeling early in the course of obesogenic stress. Taken together, our study demonstrates for the first time that cardiac RNA methylation machinery genes are regulated early during obesogenic stress in a sex-dependent manner and may play a role in the sex differences observed in cardiometabolic dysfunction.NEW & NOTEWORTHY Sex differences in obesity-associated cardiomyopathy are well documented but incompletely understood. We show for the first time that RNA methylation machinery genes may be regulated in response to obesogenic diet in a sex- and age-dependent manner and levels may correspond to cardiac systolic function. Our cardiac RNA-seq analysis suggests female, but not male mice may be protected from cardiac dysfunction by a protective cardiac remodeling response early during obesogenic stress.
Collapse
Affiliation(s)
- Amanda J Croft
- School of Medicine and Public Health, College of Health Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Conagh Kelly
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- School of Biomedical Sciences and Pharmacy, College of Health Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Australia
| | - Dongqing Chen
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- School of Biomedical Sciences and Pharmacy, College of Health Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Australia
| | - Tatt Jhong Haw
- School of Medicine and Public Health, College of Health Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Lohis Balachandran
- School of Medicine and Public Health, College of Health Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Lucy A Murtha
- School of Medicine and Public Health, College of Health Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Andrew J Boyle
- School of Medicine and Public Health, College of Health Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- Hunter New England Local Health District, Newcastle, New South Wales, Australia
| | - Aaron L Sverdlov
- School of Medicine and Public Health, College of Health Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- Hunter New England Local Health District, Newcastle, New South Wales, Australia
| | - Doan T M Ngo
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- School of Biomedical Sciences and Pharmacy, College of Health Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Australia
| |
Collapse
|
24
|
Lee Y, Sarkar A, Tassey J, Levi JN, Lee S, Liu NQ, Drake AC, Magallanes J, Stevic U, Lu J, Ge D, Tang H, Mkaratigwa T, Bian F, Shkhyan R, Bonaguidi M, Evseenko D. Inactivation of a non-canonical gp130 signaling arm attenuates chronic systemic inflammation and multimorbidity induced by a high-fat diet. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.08.588362. [PMID: 38645030 PMCID: PMC11030339 DOI: 10.1101/2024.04.08.588362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Interleukin-6 (IL-6) is a major pro-inflammatory cytokine for which the levels in plasma demonstrate a robust correlation with age and body mass index (BMI) as part of the senescence-associated secretory phenotype. IL-6 cytokines also play a crucial role in metabolic homeostasis and regenerative processes, primarily via the canonical STAT3 pathway. Thus, selective modulation of IL-6 signaling may offer a unique opportunity for therapeutic interventions. Recently, we discovered that a non-canonical signaling pathway downstream of tyrosine (Y) 814 within the intracellular domain of gp130, the IL-6 co-receptor, is responsible for the recruitment and activation of SRC family of kinases (SFK). Mice with constitutive genetic inactivation of gp130 Y814 (F814 mice) show accelerated resolution of inflammatory response and superior regenerative outcomes in skin wound healing and posttraumatic models of osteoarthritis. The current study was designed to explore if selective genetic or pharmacological inhibition of the non-canonical gp130-Y814/SFK signaling reduces systemic chronic inflammation and multimorbidity in a high-fat diet (HFD)-induced model of accelerated aging. F814 mice showed significantly reduced inflammatory response to HFD in adipose and liver tissue, with significantly reduced levels of systemic inflammation compared to wild type mice. F814 mice were also protected from HFD-induced bone loss and cartilage degeneration. Pharmacological inhibition of gp130-Y814/SFK in mice on HFD mirrored the effects observed in F814 mice on HFD; furthermore, this pharmacological treatment also demonstrated a marked increase in physical activity levels and protective effects against inflammation-associated suppression of neurogenesis in the brain tissue compared to the control group. These findings suggest that selective inhibition of SFK signaling downstream of gp130 receptor represents a promising strategy to alleviate systemic chronic inflammation. Increased degenerative changes and tissue senescence are inevitable in obese and aged organisms, but we demonstrated that the systemic response and inflammation-associated multi-morbidity can be therapeutically mitigated.
Collapse
|
25
|
Kumari R, Ponte ME, Franczak E, Prom JC, O'Neil MF, Sardiu ME, Lutkewitte AJ, Christenson LK, Shankar K, Morris EM, Thyfault JP. VCD-induced menopause mouse model reveals reprogramming of hepatic metabolism. Mol Metab 2024; 82:101908. [PMID: 38432400 PMCID: PMC10944007 DOI: 10.1016/j.molmet.2024.101908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 02/23/2024] [Accepted: 02/26/2024] [Indexed: 03/05/2024] Open
Abstract
OBJECTIVE Menopause adversely impacts systemic energy metabolism and increases the risk of metabolic disease(s) including hepatic steatosis, but the mechanisms are largely unknown. Dosing female mice with vinyl cyclohexene dioxide (VCD) selectively causes follicular atresia in ovaries, leading to a murine menopause-like phenotype. METHODS In this study, we treated female C57BL6/J mice with VCD (160 mg/kg i.p. for 20 consecutive days followed by verification of the lack of estrous cycling) to investigate changes in body composition, energy expenditure (EE), hepatic mitochondrial function, and hepatic steatosis across different dietary conditions. RESULTS VCD treatment induced ovarian follicular loss and increased follicle-stimulating hormone (FSH) levels in female mice, mimicking a menopause-like phenotype. VCD treatment did not affect body composition, or EE in mice on a low-fat diet (LFD) or in response to a short-term (1-week) high-fat, high sucrose diet (HFHS). However, the transition to a HFHS lowered cage activity in VCD mice. A chronic HFHS diet (16 weeks) significantly increased weight gain, fat mass, and hepatic steatosis in VCD-treated mice compared to HFHS-fed controls. In the liver, VCD mice showed suppressed hepatic mitochondrial respiration on LFD, while chronic HFHS resulted in compensatory increases in hepatic mitochondrial respiration. Also, liver RNA sequencing revealed that VCD promoted global upregulation of hepatic lipid/cholesterol synthesis pathways. CONCLUSION Our findings suggest that the VCD-induced menopause model compromises hepatic mitochondrial function and lipid/cholesterol homeostasis that sets the stage for HFHS diet-induced steatosis while also increasing susceptibility to obesity.
Collapse
Affiliation(s)
- Roshan Kumari
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA; Research Service, Kansas City Veterans Affairs Medical Center, Kansas City, MO, USA; KU Diabetes Institute and Kansas Center for Metabolism and Obesity, University of Kansas Medical Center, Kansas City, KS, USA; Center for Children's Healthy Lifestyles and Nutrition, Kansas City, MO, USA
| | - Michael E Ponte
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA; KU Diabetes Institute and Kansas Center for Metabolism and Obesity, University of Kansas Medical Center, Kansas City, KS, USA
| | - Edziu Franczak
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA; Research Service, Kansas City Veterans Affairs Medical Center, Kansas City, MO, USA; KU Diabetes Institute and Kansas Center for Metabolism and Obesity, University of Kansas Medical Center, Kansas City, KS, USA
| | - John C Prom
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Maura F O'Neil
- Department of Pathology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Mihaela E Sardiu
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, KS, USA; KU Diabetes Institute and Kansas Center for Metabolism and Obesity, University of Kansas Medical Center, Kansas City, KS, USA
| | - Andrew J Lutkewitte
- KU Diabetes Institute and Kansas Center for Metabolism and Obesity, University of Kansas Medical Center, Kansas City, KS, USA; Department of Internal Medicine, Division of Endocrinology, Diabetes, and Clinical Pharmacology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Lane K Christenson
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Kartik Shankar
- Department of Pediatrics, Anschutz Medical Campus, University of Colorado, Aurora, CO, USA
| | - E Matthew Morris
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA; KU Diabetes Institute and Kansas Center for Metabolism and Obesity, University of Kansas Medical Center, Kansas City, KS, USA; Center for Children's Healthy Lifestyles and Nutrition, Kansas City, MO, USA.
| | - John P Thyfault
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA; Research Service, Kansas City Veterans Affairs Medical Center, Kansas City, MO, USA; KU Diabetes Institute and Kansas Center for Metabolism and Obesity, University of Kansas Medical Center, Kansas City, KS, USA; Department of Internal Medicine, Division of Endocrinology, Diabetes, and Clinical Pharmacology, University of Kansas Medical Center, Kansas City, KS, USA; Center for Children's Healthy Lifestyles and Nutrition, Kansas City, MO, USA.
| |
Collapse
|
26
|
Chen A, Handzel A, Sau L, Cui L, Kelley ST, Thackray VG. Metabolic dysregulation and gut dysbiosis linked to hyperandrogenism in female mice. Endocrinol Diabetes Metab 2024; 7:e443. [PMID: 37872876 PMCID: PMC10782063 DOI: 10.1002/edm2.443] [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: 02/06/2023] [Revised: 07/03/2023] [Accepted: 07/05/2023] [Indexed: 10/25/2023] Open
Abstract
INTRODUCTION Polycystic ovary syndrome (PCOS) is a common endocrine pathology in women. In addition to infertility, women with PCOS have metabolic dysregulation which predisposes them to Type 2 diabetes, cardiovascular disease and non-alcoholic fatty liver disease. Moreover, women with PCOS have changes in their gut microbial community that may be indicative of dysbiosis. While hyperandrogenism is associated with both the development of metabolic dysfunction and gut dysbiosis in females, the mechanisms involved are not well understood. METHODS We used dihydrotestosterone (DHT) and ovariectomy (OVX) mouse models coupled with metabolic assessments and 16S rRNA gene sequencing to explore the contributions of hyperandrogenism and oestrogen deficiency to the development of insulin resistance and gut microbial dysbiosis in pubertal female mice. RESULTS We demonstrated that, while DHT treatment or OVX alone were insufficient to induce insulin resistance during the pubertal-to-adult transition, combining OVX with DHT resulted in insulin resistance similar to that observed in letrozole-treated mice with elevated testosterone and decreased oestrogen levels. In addition, our results showed that OVX and DHT in combination resulted in a distinct shift in the gut microbiome compared to DHT or OVX alone, suggesting that the substantial metabolic dysregulation occurring in the OVX + DHT model was accompanied by unique changes in the abundances of gut bacteria including S24-7, Rikenellaceae and Mucispirillum schaedleri. CONCLUSIONS While hyperandrogenism plays an important role in the development of metabolic dysregulation in female mice, our results indicate that investigation into additional factors influencing insulin resistance and the gut microbiome during the pubertal-to-adult transition could provide additional insight into the pathophysiology of PCOS.
Collapse
Affiliation(s)
- Annie Chen
- Department of Obstetrics, Gynecology and Reproductive SciencesUniversity of CaliforniaSan Diego, La JollaCaliforniaUSA
| | - Alex Handzel
- Bioinformatics and Medical Informatics ProgramSan Diego State UniversitySan DiegoCaliforniaUSA
| | - Lillian Sau
- Department of Obstetrics, Gynecology and Reproductive SciencesUniversity of CaliforniaSan Diego, La JollaCaliforniaUSA
| | - Laura Cui
- Department of Obstetrics, Gynecology and Reproductive SciencesUniversity of CaliforniaSan Diego, La JollaCaliforniaUSA
| | - Scott T. Kelley
- Bioinformatics and Medical Informatics ProgramSan Diego State UniversitySan DiegoCaliforniaUSA
- Department of BiologySan Diego State UniversitySan DiegoCaliforniaUSA
| | - Varykina G. Thackray
- Department of Obstetrics, Gynecology and Reproductive SciencesUniversity of CaliforniaSan Diego, La JollaCaliforniaUSA
| |
Collapse
|
27
|
Rishabh, Bansal S, Goel A, Gupta S, Malik D, Bansal N. Unravelling the Crosstalk between Estrogen Deficiency and Gut-biotaDysbiosis in the Development of Diabetes Mellitus. Curr Diabetes Rev 2024; 20:e240124226067. [PMID: 38275037 DOI: 10.2174/0115733998275953231129094057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 10/17/2023] [Accepted: 10/23/2023] [Indexed: 01/27/2024]
Abstract
Estrogens are classically considered essential hormonal signals, but they exert profound effects in a number of physiological and pathological states, including glucose homeostasis and insulin resistance. Estrogen deficiency after menopause in most women leads to increased androgenicity and changes in body composition, and it is recommended to manipulate the β-cell function of the pancreas, insulin-induced glucose transport, and hepatic glucose output, hence, the increasing incidence of type 2 diabetes mellitus. Recently, studies have reported that gut biota alteration due to estrogen deficiency contributes to altered energy metabolism and, hence, accentuates the pathology of diabetes mellitus. Emerging research suggests estrogen deficiency via genetic disposition or failure of ovaries to function in old age modulates the insulin resistance and glucose secretion workload on pancreatic beta cells by decreasing the levels of good bacteria such as Akkermansia muciniphila, Bifidobacterium spp., Lactobacillus spp., Faecalibacterium prausnitzii, Roseburia spp., and Prevotella spp., and increasing the levels of bad bacteria's such as Bacteroides spp., Clostridium difficile, Escherichia coli, and Enterococcus spp. Alteration in these bacteria's concentrations in the gut further leads to the development of impaired glucose uptake by the muscles, increased gluconeogenesis in the liver, and increased lipolysis and inflammation in the adipose tissues. Thus, the present review paper aims to clarify the intricate interactions between estrogen deficiency, gut microbiota regulation, and the development of diabetes mellitus.
Collapse
Affiliation(s)
- Rishabh
- M.M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana, India
| | - Seema Bansal
- M.M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana, India
| | - Akriti Goel
- M.M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana, India
| | - Sumeet Gupta
- M.M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana, India
| | - Deepti Malik
- Department of Biochemistry, All India Institute of Medical Sciences Bilaspur, HP, India
| | - Nitin Bansal
- Department of Pharmacy, Chaudhary Bansilal University, Bhiwani, India
| |
Collapse
|
28
|
Kumari R, Ponte ME, Franczak E, Prom JC, O'Neil MF, Sardiu ME, Lutkewitte AJ, Shankar K, Morris EM, Thyfault JP. VCD-induced menopause mouse model reveals reprogramming of hepatic metabolism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.14.571644. [PMID: 38168213 PMCID: PMC10760158 DOI: 10.1101/2023.12.14.571644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Menopause adversely impacts systemic energy metabolism and increases the risk of metabolic disease(s) including hepatic steatosis, but the mechanisms are largely unknown. Dosing female mice with vinyl cyclohexene dioxide (VCD) selectively causes follicular atresia in ovaries, leading to a murine menopause-like phenotype. In this study, we treated female C57BL6/J mice with VCD (160mg/kg i.p. for 20 consecutive days followed by verification of the lack of estrous cycling) to investigate changes in body composition, energy expenditure (EE), hepatic mitochondrial function, and hepatic steatosis across different dietary conditions. VCD treatment induced ovarian follicular loss and increased follicle-stimulating hormone (FSH) levels in female mice, mimicking a menopause-like phenotype. VCD treatment did not affect body composition, or EE in mice on a low-fat diet or in response to a short-term (1-week) high-fat, high sucrose diet (HFHS). However, the transition to a HFHS lowered cage activity in VCD mice. A chronic HFHS diet (16 weeks) significantly increased weight gain, fat mass, and hepatic steatosis in VCD-treated mice compared to HFHS-fed controls. In the liver, VCD mice showed suppressed hepatic mitochondrial respiration on LFD, while chronic HFHS diet resulted in compensatory increases in hepatic mitochondrial respiration. Also, liver RNA sequencing revealed that VCD promoted global upregulation of hepatic lipid/cholesterol synthesis pathways. Our findings suggest that the VCD- induced menopause model compromises hepatic mitochondrial function and lipid/cholesterol homeostasis that sets the stage for HFHS diet-induced steatosis while also increasing susceptibility to obesity.
Collapse
|
29
|
Todero J, Douillet C, Shumway AJ, Koller BH, Kanke M, Phuong DJ, Stýblo M, Sethupathy P. Molecular and Metabolic Analysis of Arsenic-Exposed Humanized AS3MT Mice. ENVIRONMENTAL HEALTH PERSPECTIVES 2023; 131:127021. [PMID: 38150313 PMCID: PMC10752418 DOI: 10.1289/ehp12785] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 10/30/2023] [Accepted: 12/04/2023] [Indexed: 12/29/2023]
Abstract
BACKGROUND Chronic exposure to inorganic arsenic (iAs) has been associated with type 2 diabetes (T2D). However, potential sex divergence and the underlying mechanisms remain understudied. iAs is not metabolized uniformly across species, which is a limitation of typical exposure studies in rodent models. The development of a new "humanized" mouse model overcomes this limitation. In this study, we leveraged this model to study sex differences in the context of iAs exposure. OBJECTIVES The aim of this study was to determine if males and females exhibit different liver and adipose molecular profiles and metabolic phenotypes in the context of iAs exposure. METHODS Our study was performed on wild-type (WT) 129S6/SvEvTac and humanized arsenic + 3 methyl transferase (human AS3MT) 129S6/SvEvTac mice treated with 400 ppb of iAs via drinking water ad libitum. After 1 month, mice were sacrificed and the liver and gonadal adipose depots were harvested for iAs quantification and sequencing-based microRNA and gene expression analysis. Serum blood was collected for fasting blood glucose, fasting plasma insulin, and homeostatic model assessment for insulin resistance (HOMA-IR). RESULTS We detected sex divergence in liver and adipose markers of diabetes (e.g., miR-34a, insulin signaling pathways, fasting blood glucose, fasting plasma insulin, and HOMA-IR) only in humanized (not WT) mice. In humanized female mice, numerous genes that promote insulin sensitivity and glucose tolerance in both the liver and adipose are elevated compared to humanized male mice. We also identified Klf11 as a putative master regulator of the sex divergence in gene expression in humanized mice. DISCUSSION Our study underscored the importance of future studies leveraging the humanized mouse model to study iAs-associated metabolic disease. The findings suggested that humanized males are at increased risk for metabolic dysfunction relative to humanized females in the context of iAs exposure. Future investigations should focus on the detailed mechanisms that underlie the sex divergence. https://doi.org/10.1289/EHP12785.
Collapse
Affiliation(s)
- Jenna Todero
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Christelle Douillet
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Alexandria J. Shumway
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Beverly H. Koller
- Department of Genetics, School of Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Matt Kanke
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Daryl J. Phuong
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Miroslav Stýblo
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Praveen Sethupathy
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| |
Collapse
|
30
|
Zhu X, Cai J, Wang Y, Liu X, Chen X, Wang H, Wu Z, Bao W, Fan H, Wu S. A High-Fat Diet Increases the Characteristics of Gut Microbial Composition and the Intestinal Damage Associated with Non-Alcoholic Fatty Liver Disease. Int J Mol Sci 2023; 24:16733. [PMID: 38069055 PMCID: PMC10706137 DOI: 10.3390/ijms242316733] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 11/21/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
The prevalence of non-alcoholic fatty liver disease (NAFLD) is increasing annually, and emerging evidence suggests that the gut microbiota plays a causative role in the development of NAFLD. However, the role of gut microbiota in the development of NAFLD remains unclear and warrants further investigation. Thus, C57BL/6J mice were fed a high-fat diet (HFD), and we found that the HFD significantly induced obesity and increased the accumulation of intrahepatic lipids, along with alterations in serum biochemical parameters. Moreover, it was observed that the HFD also impaired gut barrier integrity. It was revealed via 16S rRNA gene sequencing that the HFD increased gut microbial diversity, which enriched Colidextribacter, Lachnospiraceae-NK4A136-group, Acetatifactor, and Erysipelatoclostridium. Meanwhile, it reduced the abundance of Faecalibaculum, Muribaculaceae, and Coriobacteriaceae-UCG-002. The predicted metabolic pathways suggest that HFD enhances the chemotaxis and functional activity of gut microbiota pathways associated with flagellar assembly, while also increasing the risk of intestinal pathogen colonization and inflammation. And the phosphotransferase system, streptomycin biosynthesis, and starch/sucrose metabolism exhibited decreases. These findings reveal the composition and predictive functions of the intestinal microbiome in NAFLD, further corroborating the association between gut microbiota and NAFLD while providing novel insights into its potential application in gut microbiome research for NAFLD patients.
Collapse
Affiliation(s)
- Xiaoyang Zhu
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (X.Z.); (Y.W.); (X.L.); (X.C.); (H.W.); (Z.W.); (W.B.)
| | - Jiajia Cai
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China;
| | - Yifu Wang
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (X.Z.); (Y.W.); (X.L.); (X.C.); (H.W.); (Z.W.); (W.B.)
| | - Xinyu Liu
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (X.Z.); (Y.W.); (X.L.); (X.C.); (H.W.); (Z.W.); (W.B.)
| | - Xiaolei Chen
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (X.Z.); (Y.W.); (X.L.); (X.C.); (H.W.); (Z.W.); (W.B.)
| | - Haifei Wang
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (X.Z.); (Y.W.); (X.L.); (X.C.); (H.W.); (Z.W.); (W.B.)
| | - Zhengchang Wu
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (X.Z.); (Y.W.); (X.L.); (X.C.); (H.W.); (Z.W.); (W.B.)
| | - Wenbin Bao
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (X.Z.); (Y.W.); (X.L.); (X.C.); (H.W.); (Z.W.); (W.B.)
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China;
| | - Hairui Fan
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (X.Z.); (Y.W.); (X.L.); (X.C.); (H.W.); (Z.W.); (W.B.)
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Shenglong Wu
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (X.Z.); (Y.W.); (X.L.); (X.C.); (H.W.); (Z.W.); (W.B.)
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China;
| |
Collapse
|
31
|
Yang W, Jiang W, Guo S. Regulation of Macronutrients in Insulin Resistance and Glucose Homeostasis during Type 2 Diabetes Mellitus. Nutrients 2023; 15:4671. [PMID: 37960324 PMCID: PMC10647592 DOI: 10.3390/nu15214671] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 10/30/2023] [Accepted: 11/02/2023] [Indexed: 11/15/2023] Open
Abstract
Insulin resistance is an important feature of metabolic syndrome and a precursor of type 2 diabetes mellitus (T2DM). Overnutrition-induced obesity is a major risk factor for the development of insulin resistance and T2DM. The intake of macronutrients plays a key role in maintaining energy balance. The components of macronutrients distinctly regulate insulin sensitivity and glucose homeostasis. Precisely adjusting the beneficial food compound intake is important for the prevention of insulin resistance and T2DM. Here, we reviewed the effects of different components of macronutrients on insulin sensitivity and their underlying mechanisms, including fructose, dietary fiber, saturated and unsaturated fatty acids, and amino acids. Understanding the diet-gene interaction will help us to better uncover the molecular mechanisms of T2DM and promote the application of precision nutrition in practice by integrating multi-omics analysis.
Collapse
Affiliation(s)
| | | | - Shaodong Guo
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX 77843, USA; (W.Y.); (W.J.)
| |
Collapse
|
32
|
Wang CY, Wu CZ, Chen FY, Pei D, Huang LY. Changes in insulin resistance, glucose effectiveness, and first and second phases of insulin secretion in women aged 45-60 years old in Taiwan. J Chin Med Assoc 2023; 86:897-901. [PMID: 37559215 DOI: 10.1097/jcma.0000000000000976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/11/2023] Open
Abstract
BACKGROUND In women after menopause, the incidence of diabetes mellitus increases. Increased insulin resistance (IR), decreased glucose effectiveness (GE), and the first and second phases of insulin secretion (FPIS and SPIS), are the four most important factors that trigger glucose intolerance and diabetes (diabetogenic factor [DF]). In the cross-sectional study, we enrolled nondiabetic women between the ages of 45 and 60 years to observe the changes in DFs during the perimenopausal period and to elucidate the underlying mechanisms of diabetes in menopausal women. METHODS We randomly enrolled 4194 women who underwent health checkups. Using demographic and biochemical data, IR, FPIS, SPIS, and GE were calculated using previously published equations. The relationship between the DFs and age was evaluated using a simple correlation. RESULTS Body mass index, blood pressure, fasting plasma glucose, low-density lipoprotein cholesterol, triglyceride, and SPIS were higher, and GE was lower in older women (≥52 years old). A significant decrease in GE and increased SPIS were observed with age. However, no changes were observed in IR or FPIS. CONCLUSION The IR and FPIS did not change during perimenopause. Increased SPIS may compensate for the decrease in GE, which is probably one of the reasons for the higher incidence of diabetes in menopausal women.
Collapse
Affiliation(s)
- Chen-Yu Wang
- Department of Obstetrics and Gynecology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Chung-Ze Wu
- Division of Endocrinology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan, ROC
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC
| | - Fang-Yu Chen
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Fu Jen Catholic University Hospital, School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan, ROC
| | - Dee Pei
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Fu Jen Catholic University Hospital, School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan, ROC
| | - Li-Ying Huang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Fu Jen Catholic University Hospital, School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan, ROC
| |
Collapse
|
33
|
Eng PC, Forlano R, Tan T, Manousou P, Dhillo WS, Izzi-Engbeaya C. Non-alcoholic fatty liver disease in women - Current knowledge and emerging concepts. JHEP Rep 2023; 5:100835. [PMID: 37771547 PMCID: PMC10522907 DOI: 10.1016/j.jhepr.2023.100835] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 05/26/2023] [Accepted: 06/05/2023] [Indexed: 09/30/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a major cause of liver disease worldwide, affecting up to 30% of adults. Progression to non-alcoholic steatohepatitis (NASH) is a key risk factor for cirrhosis, hepatocellular carcinoma and cardiovascular events. Alterations in reproductive hormones are linked to the development and/or progression of NAFLD/NASH in women. Women with polycystic ovary syndrome and those with oestrogen deficiency are at increased risk of NAFLD/NASH, with higher mortality rates in older women compared to men of similar ages. NAFLD/NASH is currently the leading indication for liver transplantation in women without hepatocellular carcinoma. Therefore, a better understanding of NAFLD in women is needed to improve outcomes. In this review, we discuss the hormonal and non-hormonal factors that contribute to NAFLD development and progression in women. Furthermore, we highlight areas of focus for clinical practice and for future research.
Collapse
Affiliation(s)
- Pei Chia Eng
- Department of Metabolism, Digestion and Reproduction, Imperial College London, UK
- Department of Diabetes and Endocrinology, Imperial College Healthcare NHS Trust, London, UK
| | - Roberta Forlano
- Department of Metabolism, Digestion and Reproduction, Imperial College London, UK
- Department of Hepatology, Imperial College Healthcare NHS Trust, London, UK
| | - Tricia Tan
- Department of Metabolism, Digestion and Reproduction, Imperial College London, UK
- Department of Diabetes and Endocrinology, Imperial College Healthcare NHS Trust, London, UK
| | - Pinelopi Manousou
- Department of Metabolism, Digestion and Reproduction, Imperial College London, UK
- Department of Hepatology, Imperial College Healthcare NHS Trust, London, UK
| | - Waljit S. Dhillo
- Department of Metabolism, Digestion and Reproduction, Imperial College London, UK
- Department of Diabetes and Endocrinology, Imperial College Healthcare NHS Trust, London, UK
| | - Chioma Izzi-Engbeaya
- Department of Metabolism, Digestion and Reproduction, Imperial College London, UK
- Department of Diabetes and Endocrinology, Imperial College Healthcare NHS Trust, London, UK
| |
Collapse
|
34
|
Rousseau-Ralliard D, Chavatte-Palmer P, Couturier-Tarrade A. The Effect of Maternal Exposure to a Diet High in Fats and Cholesterol on the Placental Function and Phenotype of the Offspring in a Rabbit Model: A Summary Review of About 15 Years of Research. Int J Mol Sci 2023; 24:14547. [PMID: 37834002 PMCID: PMC10572169 DOI: 10.3390/ijms241914547] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/21/2023] [Accepted: 09/23/2023] [Indexed: 10/15/2023] Open
Abstract
The rates of obesity and being overweight are increasing all around the world, especially among women of childbearing age, in part due to overconsumption of lipids. The aim of this summary review was to present the cellular and molecular effects of a hyperlipidic high-cholesterol (H) diet on the maternal and offspring phenotype at the early embryonic, neonatal, weaning and adult stages while considering the effects of sex and to identify the window(s) of vulnerability linked to this exposure in a rabbit model. Before breeding, the H diet induced dyslipidemia and aortic atherosclerosis lesions and increased the number of atretic follicles. In the offspring, the H diet disrupted the embryonic phenotype and induced fetal hypotrophy associated with sex-specific disturbances of the feto-placental unit. In adulthood, the offspring of the H dams were heavier and hyperphagic and had increased blood pressure associated with disturbed gonadal development in both sexes. Vulnerability windows were explored via embryo transfers. The maternal gestational diet was shown to play a key role in the feto-placental phenotype, and preconception programming was unquestionably also observed. These two periods could represent windows of intervention in the context of obesity or being overweight to limit fetal and placental consequences.
Collapse
Affiliation(s)
- Delphine Rousseau-Ralliard
- BREED, INRAE, UVSQ, Université Paris-Saclay, 78350 Jouy-en-Josas, France; (D.R.-R.); (P.C.-P.)
- BREED, Ecole Nationale Vétérinaire d’Alfort, 94700 Maisons-Alfort, France
| | - Pascale Chavatte-Palmer
- BREED, INRAE, UVSQ, Université Paris-Saclay, 78350 Jouy-en-Josas, France; (D.R.-R.); (P.C.-P.)
- BREED, Ecole Nationale Vétérinaire d’Alfort, 94700 Maisons-Alfort, France
| | - Anne Couturier-Tarrade
- BREED, INRAE, UVSQ, Université Paris-Saclay, 78350 Jouy-en-Josas, France; (D.R.-R.); (P.C.-P.)
- BREED, Ecole Nationale Vétérinaire d’Alfort, 94700 Maisons-Alfort, France
| |
Collapse
|
35
|
Chang M, Shao Z, Wei W, Shen P, Shen G. Sex-specific prevalence and risk factors of metabolic-associated fatty liver disease among 75,570 individuals in eastern China. Front Endocrinol (Lausanne) 2023; 14:1241169. [PMID: 37822594 PMCID: PMC10563804 DOI: 10.3389/fendo.2023.1241169] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 09/08/2023] [Indexed: 10/13/2023] Open
Abstract
Background Metabolic-associated fatty liver disease (MAFLD) is a newly proposed definition and there is limited data on MAFLD prevalence. We aimed to investigate the prevalence of MAFLD in an eastern Chinese population. Methods This cross-sectional study included participants from an eastern Chinese population who underwent regular health checkups. Based on current diagnostic criteria, MAFLD was diagnosed in individuals with both hepatic steatosis and metabolic disorders. The overall and stratified prevalence derived based on sex, age, body mass index (BMI), and various metabolic disorders were estimated. Multivariate logistic regression analysis was used to determine the risk factors for MAFLD. Results Among the 75,570 participants, the overall prevalence of MAFLD was 37.32%, with higher rates in men (45.66%) than in women (23.91%). MAFLD prevalence was highest in men aged 40-49 years (52.21%) and women aged 70-79 years (44.77%). In all the BMI subgroups, the prevalence was higher in men than in women. In both sexes, the prevalence of MAFLD increased as BMI levels increased. Furthermore, MAFLD was associated with metabolic disorders, especially in the female participants with severe obesity (odds ratio 58.318; 95% confidence interval: 46.978-72.397). Conclusion MAFLD is prevalent in the general adult population in eastern China. Sex-specific differences in MAFLD prevalence were identified based on age, BMI, and metabolic disorders. MAFLD is associated with metabolic disorders, particularly obesity.
Collapse
Affiliation(s)
| | | | | | - Peipu Shen
- Department of Health Management Center, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Guifang Shen
- Department of Health Management Center, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
36
|
Afonso-Alí A, Porrini E, Teixido-Trujillo S, Pérez-Pérez JA, Luis-Lima S, Acosta-González NG, Sosa-Paz I, Díaz-Martín L, Rodríguez-González C, Rodríguez-Rodríguez AE. The Role of Gender Differences and Menopause in Obesity-Related Renal Disease, Renal Inflammation and Lipotoxicity. Int J Mol Sci 2023; 24:12984. [PMID: 37629165 PMCID: PMC10455320 DOI: 10.3390/ijms241612984] [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: 07/06/2023] [Revised: 08/08/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
The pathogenesis of obesity-related-renal disease is unknown. Menopause can promote renal disease in obese women, but this interaction is unclear. In a previous study, we observed that obese male and female mice developed albuminuria, hyperfiltration, and glomerulomegaly, and these changes were more severe in those obese ovariectomized females. In this study, we also evaluated renal inflammation and lipotoxicity in that animal model. For six months, 43 males and 36 females C57BL6/J mice were randomized to standard diet (SD) or high fat diet (HFD). A group of female animals on SD or HFD was ovariectomized to simulate menopause. We evaluated cytokines: NF-κβ p65, IL-1β, MCP-1, TNF-α, total lipid content, lipid classes, and fatty acid profile in total lipid and individual lipid classes in renal tissue and urine. We found that obese males and females showed higher NF-kβ p-65, TNF-α and MCP-1 in renal tissue, and obese females ovariectomized had higher IL-1β and TNF-α compared with not-ovariectomized. Also, obese animals showed lower proinflammatory and higher anti-inflammatory fatty acids in kidney total lipids, while obese females ovariectomized had a more exacerbated pattern. In brief, obesity induces inflammation and an unbalanced lipidic profile in renal tissue. This pattern seems to be enhanced in obesity after menopause.
Collapse
Affiliation(s)
- Aaron Afonso-Alí
- ITB (Instituto Tecnologías Biomédicas), University of La Laguna, 38200 Tenerife, Spain; (A.A.-A.); (S.T.-T.); (L.D.-M.); (C.R.-G.); (A.E.R.-R.)
| | - Esteban Porrini
- ITB (Instituto Tecnologías Biomédicas), University of La Laguna, 38200 Tenerife, Spain; (A.A.-A.); (S.T.-T.); (L.D.-M.); (C.R.-G.); (A.E.R.-R.)
- Research Unit, Hospital Universitario de Canarias, 38200 Tenerife, Spain
| | - Silvia Teixido-Trujillo
- ITB (Instituto Tecnologías Biomédicas), University of La Laguna, 38200 Tenerife, Spain; (A.A.-A.); (S.T.-T.); (L.D.-M.); (C.R.-G.); (A.E.R.-R.)
- Research Unit, Hospital Universitario de Canarias, 38200 Tenerife, Spain
| | - José Antonio Pérez-Pérez
- Department of Animal Biology, Edaphology and Geology, Faculty of Biology, University of La Laguna, 38204 Tenerife, Spain; (J.A.P.-P.); (N.G.A.-G.); (I.S.-P.)
| | - Sergio Luis-Lima
- Department of Laboratory Medicine, Complejo Hospitalario Universitario de Canarias, 38200 San Cristóbal de La Laguna, Spain;
| | - Nieves Guadalupe Acosta-González
- Department of Animal Biology, Edaphology and Geology, Faculty of Biology, University of La Laguna, 38204 Tenerife, Spain; (J.A.P.-P.); (N.G.A.-G.); (I.S.-P.)
| | - Irene Sosa-Paz
- Department of Animal Biology, Edaphology and Geology, Faculty of Biology, University of La Laguna, 38204 Tenerife, Spain; (J.A.P.-P.); (N.G.A.-G.); (I.S.-P.)
| | - Laura Díaz-Martín
- ITB (Instituto Tecnologías Biomédicas), University of La Laguna, 38200 Tenerife, Spain; (A.A.-A.); (S.T.-T.); (L.D.-M.); (C.R.-G.); (A.E.R.-R.)
- Research Unit, Hospital Universitario de Canarias, 38200 Tenerife, Spain
| | - Covadonga Rodríguez-González
- ITB (Instituto Tecnologías Biomédicas), University of La Laguna, 38200 Tenerife, Spain; (A.A.-A.); (S.T.-T.); (L.D.-M.); (C.R.-G.); (A.E.R.-R.)
- Department of Animal Biology, Edaphology and Geology, Faculty of Biology, University of La Laguna, 38204 Tenerife, Spain; (J.A.P.-P.); (N.G.A.-G.); (I.S.-P.)
| | - Ana Elena Rodríguez-Rodríguez
- ITB (Instituto Tecnologías Biomédicas), University of La Laguna, 38200 Tenerife, Spain; (A.A.-A.); (S.T.-T.); (L.D.-M.); (C.R.-G.); (A.E.R.-R.)
- Research Unit, Hospital Universitario de Canarias, 38200 Tenerife, Spain
| |
Collapse
|
37
|
Sacharidou A, Chambliss K, Peng J, Barrera J, Tanigaki K, Luby-Phelps K, Özdemir İ, Khan S, Sirsi SR, Kim SH, Katzenellenbogen BS, Katzenellenbogen JA, Kanchwala M, Sathe AA, Lemoff A, Xing C, Hoyt K, Mineo C, Shaul PW. Endothelial ERα promotes glucose tolerance by enhancing endothelial insulin transport to skeletal muscle. Nat Commun 2023; 14:4989. [PMID: 37591837 PMCID: PMC10435471 DOI: 10.1038/s41467-023-40562-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 08/01/2023] [Indexed: 08/19/2023] Open
Abstract
The estrogen receptor (ER) designated ERα has actions in many cell and tissue types that impact glucose homeostasis. It is unknown if these include mechanisms in endothelial cells, which have the potential to influence relative obesity, and processes in adipose tissue and skeletal muscle that impact glucose control. Here we show that independent of impact on events in adipose tissue, endothelial ERα promotes glucose tolerance by enhancing endothelial insulin transport to skeletal muscle. Endothelial ERα-deficient male mice are glucose intolerant and insulin resistant, and in females the antidiabetogenic actions of estradiol (E2) are absent. The glucose dysregulation is due to impaired skeletal muscle glucose disposal that results from attenuated muscle insulin delivery. Endothelial ERα activation stimulates insulin transcytosis by skeletal muscle microvascular endothelial cells. Mechanistically this involves nuclear ERα-dependent upregulation of vesicular trafficking regulator sorting nexin 5 (SNX5) expression, and PI3 kinase activation that drives plasma membrane recruitment of SNX5. Thus, coupled nuclear and non-nuclear actions of ERα promote endothelial insulin transport to skeletal muscle to foster normal glucose homeostasis.
Collapse
Affiliation(s)
- Anastasia Sacharidou
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Ken Chambliss
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Jun Peng
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Jose Barrera
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Keiji Tanigaki
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Katherine Luby-Phelps
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - İpek Özdemir
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Sohaib Khan
- University of Cincinnati Cancer Institute, Department of Cancer and Cell Biology, University of Cincinnati College of Medicine, Cincinnati, OH, 45219, USA
| | - Shashank R Sirsi
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Sung Hoon Kim
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Benita S Katzenellenbogen
- Departments of Physiology and Cell Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | | | - Mohammed Kanchwala
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Adwait A Sathe
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Andrew Lemoff
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Chao Xing
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Kenneth Hoyt
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Chieko Mineo
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| | - Philip W Shaul
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
38
|
Fabre A, Tramunt B, Montagner A, Mouly C, Riant E, Calmy ML, Adlanmerini M, Fontaine C, Burcelin R, Lenfant F, Arnal JF, Gourdy P. Membrane estrogen receptor-α contributes to female protection against high-fat diet-induced metabolic disorders. Front Endocrinol (Lausanne) 2023; 14:1215947. [PMID: 37529599 PMCID: PMC10390233 DOI: 10.3389/fendo.2023.1215947] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 06/23/2023] [Indexed: 08/03/2023] Open
Abstract
Background Estrogen Receptor α (ERα) is a significant modulator of energy balance and lipid/glucose metabolisms. Beyond the classical nuclear actions of the receptor, rapid activation of intracellular signaling pathways is mediated by a sub-fraction of ERα localized to the plasma membrane, known as Membrane Initiated Steroid Signaling (MISS). However, whether membrane ERα is involved in the protective metabolic actions of endogenous estrogens in conditions of nutritional challenge, and thus contributes to sex differences in the susceptibility to metabolic diseases, remains to be clarified. Methods Male and female C451A-ERα mice, harboring a point mutation which results in the abolition of membrane localization and MISS-related effects of the receptor, and their wild-type littermates (WT-ERα) were maintained on a normal chow diet (NCD) or fed a high-fat diet (HFD). Body weight gain, body composition and glucose tolerance were monitored. Insulin sensitivity and energy balance regulation were further investigated in HFD-fed female mice. Results C451A-ERα genotype had no influence on body weight gain, adipose tissue accumulation and glucose tolerance in NCD-fed mice of both sexes followed up to 7 months of age, nor male mice fed a HFD for 12 weeks. In contrast, compared to WT-ERα littermates, HFD-fed C451A-ERα female mice exhibited: 1) accelerated fat mass accumulation, liver steatosis and impaired glucose tolerance; 2) whole-body insulin resistance, assessed by hyperinsulinemic-euglycemic clamps, and altered insulin-induced signaling in skeletal muscle and liver; 3) significant decrease in energy expenditure associated with histological and functional abnormalities of brown adipose tissue and a defect in thermogenesis regulation in response to cold exposure. Conclusion Besides the well-characterized role of ERα nuclear actions, membrane-initiated ERα extra-nuclear signaling contributes to female, but not to male, protection against HFD-induced obesity and associated metabolic disorders in mouse.
Collapse
Affiliation(s)
- Aurélie Fabre
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Institut National de la Santé et de la Recherche Médicale (INSERM)/Université Paul Sabatier (UPS), Université Toulouse 3, Toulouse, France
| | - Blandine Tramunt
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Institut National de la Santé et de la Recherche Médicale (INSERM)/Université Paul Sabatier (UPS), Université Toulouse 3, Toulouse, France
- Service de Diabétologie, Maladies Métaboliques et Nutrition, Centre Hospitalier Universitaire (CHU) de Toulouse, Toulouse, France
| | - Alexandra Montagner
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Institut National de la Santé et de la Recherche Médicale (INSERM)/Université Paul Sabatier (UPS), Université Toulouse 3, Toulouse, France
| | - Céline Mouly
- Service d’Endocrinologie et Nutrition, Centre Hospitalier Universitaire (CHU) de Toulouse, Toulouse, France
| | - Elodie Riant
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Institut National de la Santé et de la Recherche Médicale (INSERM)/Université Paul Sabatier (UPS), Université Toulouse 3, Toulouse, France
| | - Marie-Lou Calmy
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Institut National de la Santé et de la Recherche Médicale (INSERM)/Université Paul Sabatier (UPS), Université Toulouse 3, Toulouse, France
| | - Marine Adlanmerini
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Institut National de la Santé et de la Recherche Médicale (INSERM)/Université Paul Sabatier (UPS), Université Toulouse 3, Toulouse, France
| | - Coralie Fontaine
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Institut National de la Santé et de la Recherche Médicale (INSERM)/Université Paul Sabatier (UPS), Université Toulouse 3, Toulouse, France
| | - Rémy Burcelin
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Institut National de la Santé et de la Recherche Médicale (INSERM)/Université Paul Sabatier (UPS), Université Toulouse 3, Toulouse, France
| | - Françoise Lenfant
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Institut National de la Santé et de la Recherche Médicale (INSERM)/Université Paul Sabatier (UPS), Université Toulouse 3, Toulouse, France
| | - Jean-François Arnal
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Institut National de la Santé et de la Recherche Médicale (INSERM)/Université Paul Sabatier (UPS), Université Toulouse 3, Toulouse, France
| | - Pierre Gourdy
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Institut National de la Santé et de la Recherche Médicale (INSERM)/Université Paul Sabatier (UPS), Université Toulouse 3, Toulouse, France
- Service de Diabétologie, Maladies Métaboliques et Nutrition, Centre Hospitalier Universitaire (CHU) de Toulouse, Toulouse, France
| |
Collapse
|
39
|
Daniel JM, Lindsey SH, Mostany R, Schrader LA, Zsombok A. Cardiometabolic health, menopausal estrogen therapy and the brain: How effects of estrogens diverge in healthy and unhealthy preclinical models of aging. Front Neuroendocrinol 2023; 70:101068. [PMID: 37061205 PMCID: PMC10725785 DOI: 10.1016/j.yfrne.2023.101068] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/23/2023] [Accepted: 04/10/2023] [Indexed: 04/17/2023]
Abstract
Research in preclinical models indicates that estrogens are neuroprotective and positively impact cognitive aging. However, clinical data are equivocal as to the benefits of menopausal estrogen therapy to the brain and cognition. Pre-existing cardiometabolic disease may modulate mechanisms by which estrogens act, potentially reducing or reversing protections they provide against cognitive decline. In the current review we propose mechanisms by which cardiometabolic disease may alter estrogen effects, including both alterations in actions directly on brain memory systems and actions on cardiometabolic systems, which in turn impact brain memory systems. Consideration of mechanisms by which estrogen administration can exert differential effects dependent upon health phenotype is consistent with the move towards precision or personalized medicine, which aims to determine which treatment interventions will work for which individuals. Understanding effects of estrogens in both healthy and unhealthy models of aging is critical to optimizing the translational link between preclinical and clinical research.
Collapse
Affiliation(s)
- Jill M Daniel
- Department of Psychology and Brain Institute, Tulane University, New Orleans, LA, United States.
| | - Sarah H Lindsey
- Department of Pharmacology and Brain Institute, Tulane University, New Orleans, LA, United States
| | - Ricardo Mostany
- Department of Pharmacology and Brain Institute, Tulane University, New Orleans, LA, United States
| | - Laura A Schrader
- Department of Cell & Molecular Biology and Brain Institute, Tulane University, New Orleans, LA, United States
| | - Andrea Zsombok
- Department of Physiology and Brain Institute, Tulane University, New Orleans, LA, United States
| |
Collapse
|
40
|
Pepe GJ, Albrecht ED. Microvascular Skeletal-Muscle Crosstalk in Health and Disease. Int J Mol Sci 2023; 24:10425. [PMID: 37445602 DOI: 10.3390/ijms241310425] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/13/2023] [Accepted: 06/14/2023] [Indexed: 07/15/2023] Open
Abstract
As an organ system, skeletal muscle is essential for the generation of energy that underpins muscle contraction, plays a critical role in controlling energy balance and insulin-dependent glucose homeostasis, as well as vascular well-being, and regenerates following injury. To achieve homeostasis, there is requirement for "cross-talk" between the myogenic and vascular components and their regulatory factors that comprise skeletal muscle. Accordingly, this review will describe the following: [a] the embryonic cell-signaling events important in establishing vascular and myogenic cell-lineage, the cross-talk between endothelial cells (EC) and myogenic precursors underpinning the development of muscle, its vasculature and the satellite-stem-cell (SC) pool, and the EC-SC cross-talk that maintains SC quiescence and localizes ECs to SCs and angio-myogenesis postnatally; [b] the vascular-myocyte cross-talk and the actions of insulin on vasodilation and capillary surface area important for the uptake of glucose/insulin by myofibers and vascular homeostasis, the microvascular-myocyte dysfunction that characterizes the development of insulin resistance, diabetes and hypertension, and the actions of estrogen on muscle vasodilation and growth in adults; [c] the role of estrogen in utero on the development of fetal skeletal-muscle microvascularization and myofiber hypertrophy required for metabolic/vascular homeostasis after birth; [d] the EC-SC interactions that underpin myofiber vascular regeneration post-injury; and [e] the role of the skeletal-muscle vasculature in Duchenne muscular dystrophy.
Collapse
Affiliation(s)
- Gerald J Pepe
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA 23501, USA
| | - Eugene D Albrecht
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
41
|
Visniauskas B, Reverte V, Abshire CM, Ogola BO, Rosales CB, Galeas-Pena M, Sure VN, Sakamuri SSVP, Harris NR, Kilanowski-Doroh I, Mcnally AB, Horton AC, Zimmerman M, Katakam PVG, Lindsey SH, Prieto MC. High-plasma soluble prorenin receptor is associated with vascular damage in male, but not female, mice fed a high-fat diet. Am J Physiol Heart Circ Physiol 2023; 324:H762-H775. [PMID: 36930656 PMCID: PMC10151046 DOI: 10.1152/ajpheart.00638.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 03/02/2023] [Accepted: 03/13/2023] [Indexed: 03/18/2023]
Abstract
Plasma soluble prorenin receptor (sPRR) displays sexual dimorphism and is higher in women with type 2 diabetes mellitus (T2DM). However, the contribution of plasma sPRR to the development of vascular complications in T2DM remains unclear. We investigated if plasma sPRR contributes to sex differences in the activation of the systemic renin-angiotensin-aldosterone system (RAAS) and vascular damage in a model of high-fat diet (HFD)-induced T2DM. Male and female C57BL/6J mice were fed either a normal fat diet (NFD) or an HFD for 28 wk to assess changes in blood pressure, cardiometabolic phenotype, plasma prorenin/renin, sPRR, and ANG II. After completing dietary protocols, tissues were collected from males to assess vascular reactivity and aortic reactive oxygen species (ROS). A cohort of male mice was used to determine the direct contribution of increased systemic sPRR by infusion. To investigate the role of ovarian hormones, ovariectomy (OVX) was performed at 32 wk in females fed either an NFD or HFD. Significant sex differences were found after 28 wk of HFD, where only males developed T2DM and increased plasma prorenin/renin, sPRR, and ANG II. T2DM in males was accompanied by nondipping hypertension, carotid artery stiffening, and aortic ROS. sPRR infusion in males induced vascular thickening instead of material stiffening caused by HFD-induced T2DM. While intact females were less prone to T2DM, OVX increased plasma prorenin/renin, sPRR, and systolic blood pressure. These data suggest that sPRR is a novel indicator of systemic RAAS activation and reflects the onset of vascular complications during T2DM regulated by sex.NEW & NOTEWORTHY High-fat diet (HFD) for 28 wk leads to type 2 diabetes mellitus (T2DM) phenotype, concomitant with increased plasma soluble prorenin receptor (sPRR), nondipping blood pressure, and vascular stiffness in male mice. HFD-fed female mice exhibiting a preserved cardiometabolic phenotype until ovariectomy revealed increased plasma sPRR and blood pressure. Plasma sPRR may indicate the status of systemic renin-angiotensin-aldosterone system (RAAS) activation and the onset of vascular complications during T2DM in a sex-dependent manner.
Collapse
Affiliation(s)
- Bruna Visniauskas
- Department of Physiology, Tulane University School of Medicine, New Orleans, Louisiana, United States
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States
- Tulane Center for Sex-Based Biology and Medicine, New Orleans, Louisiana, United States
| | - Virginia Reverte
- Department of Physiology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Caleb M Abshire
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Benard O Ogola
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
| | - Carla B Rosales
- Department of Physiology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Michelle Galeas-Pena
- Department of Physiology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Venkata N Sure
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Siva S V P Sakamuri
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Nicholas R Harris
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Isabella Kilanowski-Doroh
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Alexandra B Mcnally
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Alec C Horton
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Margaret Zimmerman
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Prasad V G Katakam
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Sarah H Lindsey
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States
- Tulane Center for Sex-Based Biology and Medicine, New Orleans, Louisiana, United States
- Tulane Hypertension and Renal Center of Excellence, New Orleans, Louisiana, United States
| | - Minolfa C Prieto
- Department of Physiology, Tulane University School of Medicine, New Orleans, Louisiana, United States
- Tulane Center for Sex-Based Biology and Medicine, New Orleans, Louisiana, United States
- Tulane Hypertension and Renal Center of Excellence, New Orleans, Louisiana, United States
| |
Collapse
|
42
|
Talarico CHZ, Alves ES, Dos Santos JDM, Sucupira FGS, Araujo LCC, Camporez JP. Progesterone Has No Impact on the Beneficial Effects of Estradiol Treatment in High-Fat-Fed Ovariectomized Mice. Curr Issues Mol Biol 2023; 45:3965-3976. [PMID: 37232722 DOI: 10.3390/cimb45050253] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 04/20/2023] [Accepted: 04/25/2023] [Indexed: 05/27/2023] Open
Abstract
In recent decades, clinical and experimental studies have revealed that estradiol contributes enormously to glycemic homeostasis. However, the same consensus does not exist in women during menopause who undergo replacement with progesterone or conjugated estradiol and progesterone. Since most hormone replacement treatments in menopausal women are performed with estradiol (E2) and progesterone (P4) combined, this work aimed to investigate the effects of progesterone on energy metabolism and insulin resistance in an experimental model of menopause (ovariectomized female mice-OVX mice) fed a high-fat diet (HFD). OVX mice were treated with E2 or P4 (or both combined). OVX mice treated with E2 alone or combined with P4 displayed reduced body weight after six weeks of HFD feeding compared to OVX mice and OVX mice treated with P4 alone. These data were associated with improved glucose tolerance and insulin sensitivity in OVX mice treated with E2 (alone or combined with P4) compared to OVX and P4-treated mice. Additionally, E2 treatment (alone or combined with P4) reduced both hepatic and muscle triglyceride content compared with OVX control mice and OVX + P4 mice. There were no differences between groups regarding hepatic enzymes in plasma and inflammatory markers. Therefore, our results revealed that progesterone replacement alone does not seem to influence glucose homeostasis and ectopic lipid accumulation in OVX mice. These results will help expand knowledge about hormone replacement in postmenopausal women associated with metabolic syndrome and non-alcoholic fatty liver disease.
Collapse
Affiliation(s)
- Carlos H Z Talarico
- Department of Physiology, Ribeirao Preto School of Medicine, University of Sao Paulo, Ribeirão Preto 14049-900, Brazil
| | - Ester S Alves
- Department of Physiology, Ribeirao Preto School of Medicine, University of Sao Paulo, Ribeirão Preto 14049-900, Brazil
| | - Jessica D M Dos Santos
- Department of Physiology, Ribeirao Preto School of Medicine, University of Sao Paulo, Ribeirão Preto 14049-900, Brazil
| | - Felipe G S Sucupira
- Department of Physiology, Ribeirao Preto School of Medicine, University of Sao Paulo, Ribeirão Preto 14049-900, Brazil
| | - Layanne C C Araujo
- Department of Physiology, Ribeirao Preto School of Medicine, University of Sao Paulo, Ribeirão Preto 14049-900, Brazil
| | - João Paulo Camporez
- Department of Physiology, Ribeirao Preto School of Medicine, University of Sao Paulo, Ribeirão Preto 14049-900, Brazil
| |
Collapse
|
43
|
Christensen A, Pike CJ. Effects of APOE Genotype and Western Diet on Metabolic Phenotypes in Female Mice. Metabolites 2023; 13:metabo13020287. [PMID: 36837905 PMCID: PMC9959618 DOI: 10.3390/metabo13020287] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/09/2023] [Accepted: 02/13/2023] [Indexed: 02/18/2023] Open
Abstract
Western diets high in sugars and saturated fats have been reported to induce metabolic and inflammatory impairments that are associated with several age-related disorders, including Alzheimer's disease (AD) and type 2 diabetes (T2D). The apolipoprotein E (APOE) genotype is associated with metabolic and inflammatory outcomes that contribute to risks for AD and T2D, with the APOE4 genotype increasing risks relative to the more common APOE3 allele. In this study, we investigated the impacts of the APOE genotype on systemic and neural effects of the Western diet. Female mice with knock-in of human APOE3 or APOE4 were exposed to control or Western diet for 13 weeks. In the control diet, we observed that APOE4 mice presented with impaired metabolic phenotypes, exhibiting greater adiposity, higher plasma leptin and insulin levels, and poorer glucose clearance than APOE3 mice. Behaviorally, APOE4 mice exhibited worse performance in a hippocampal-dependent learning task. In visceral adipose tissue, APOE4 mice exhibited generally higher expression levels of macrophage- and inflammation-related genes. The cerebral cortex showed a similar pattern, with higher expression of macrophage- and inflammation-related genes in APOE4 than APOE3 mice. Exposure to the Western diet yielded modest, statistically non-significant effects on most metabolic, behavioral, and gene expression measures in both APOE genotypes. Interestingly, the Western diet resulted in reduced gene expression of a few macrophage markers, specifically in APOE4 mice. The observed relative resistance to the Western diet suggests protective roles of both female sex and young adult age. Further, the data demonstrate that APOE4 is associated with deleterious systemic and neural phenotypes and an altered response to a metabolic stressor, findings relevant to the understanding of interactions between the APOE genotype and risks for metabolic disorders.
Collapse
|
44
|
Marsh ML, Oliveira MN, Vieira-Potter VJ. Adipocyte Metabolism and Health after the Menopause: The Role of Exercise. Nutrients 2023; 15:444. [PMID: 36678314 PMCID: PMC9862030 DOI: 10.3390/nu15020444] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/09/2023] [Accepted: 01/10/2023] [Indexed: 01/19/2023] Open
Abstract
Postmenopausal women represent an important target population in need of preventative cardiometabolic approaches. The loss of estrogen following the menopause eliminates protections against metabolic dysfunction, largely due to its role in the health and function of adipose tissue. In addition, some studies associate the menopause with reduced physical activity, which could potentially exacerbate the deleterious cardiometabolic risk profile accompanying the menopause. Meanwhile, exercise has adipocyte-specific effects that may alleviate the adverse impact of estrogen loss through the menopausal transition period and beyond. Exercise thus remains the best therapeutic agent available to mitigate menopause-associated metabolic dysfunction and represents a vital behavioral strategy to prevent and alleviate health decline in this population.
Collapse
|
45
|
Peng G, Mosleh E, Yuhas A, Katada K, Cherry C, Golson ML. FOXM1 acts sexually dimorphically to regulate functional β-cell mass. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.12.523673. [PMID: 36711451 PMCID: PMC9882186 DOI: 10.1101/2023.01.12.523673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The transcription factor FOXM1 regulates β-cell proliferation and insulin secretion. Our previous work demonstrates that expressing an activated form of FOXM1 (FOXM1*) in β cells increases β-cell proliferation and mass in aged male mice. Additionally, FOXM1* enhances β-cell function even in young mice, in which no β-cell mass elevation occurs. Here, we demonstrate that FOXM1 acts in a sexually dimorphic manner in the β cell. Expression of FOXM1* in female mouse β cells does not affect β-cell proliferation or glucose tolerance. Transduction of male but not female human islets with FOXM1* enhances insulin secretion in response to elevated glucose. Estrogen contributes to diabetes susceptibility differences between males and females, and the estrogen receptor (ER)α is the primary mediator of β-cell estrogen signaling. We show that FOXM1* can rescue impaired glucose tolerance in female mice with a pancreas-wide ERα deletion. Further, FOXM1 and ERα binding sites overlap with each other and with other β-cell-enriched transcription factors, including ISL1, PAX6, MAF, and GATA. These data indicate that FOMX1 and ERα cooperate to regulate β-cell function and suggest a general mechanism contributing to the lower incidence of diabetes observed in women.
Collapse
|
46
|
Estrogen as a key regulator of energy homeostasis and metabolic health. Biomed Pharmacother 2022; 156:113808. [DOI: 10.1016/j.biopha.2022.113808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/02/2022] [Accepted: 10/03/2022] [Indexed: 11/23/2022] Open
|
47
|
Aladhami AK, Unger CA, Hope MC, Cotham WE, Velázquez KT, Enos RT. Augmenting Skeletal Muscle Estrogen Does not Prevent or Rescue Obesity-linked Metabolic Impairments in Female Mice. Endocrinology 2022; 163:6678809. [PMID: 36039699 DOI: 10.1210/endocr/bqac146] [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: 06/23/2022] [Indexed: 11/19/2022]
Abstract
AIMS We developed a novel mouse model with increased skeletal muscle estrogen content via inducible, skeletal-muscle-specific aromatase overexpression (SkM-Arom↑). We proposed to examine the effect that increased skeletal muscle estrogen both in gonadally intact and ovariectomized (OVX) female mice has on preventing or rescuing high-fat diet (HFD)-induced obesity. METHODS In the prevention experiment, gonadally intact and OVX SkM-Arom↑ mice and littermate controls were fed a low-fat diet (LFD) or HFD for 13 weeks. SkM-Arom↑ was induced at the initiation of dietary treatment. In the intervention experiment, gonadally intact and OVX SkM-Arom↑ mice and littermate controls were fed an HFD for 14 weeks before induction of SkM-Arom↑ for 6 weeks. Glucose tolerance, insulin action, adipose tissue inflammation, and body composition were assessed. Liquid chromatography-mass spectrometry was used to determine circulating and skeletal muscle steroid content. RESULTS SkM-Arom↑ significantly increased skeletal muscle 17β-estradiol (E2) and estrone (E1) in both experiments. Interestingly, this resulted in leakage of estrogens into circulation, producing a physiologically relevant E2 concentration. Consequently, bone mineral density (BMD) was enhanced and adipose tissue inflammation was reduced in the prevention experiment only. However, no benefits were seen with respect to changes in adiposity or metabolic outcomes. CONCLUSION We show that increasing skeletal muscle estrogen content does not provide a metabolic benefit in gonadally intact and OVX female mice in the setting of obesity. However, a chronic physiological concentration of circulating E2 can improve BMD and reduce adipose tissue inflammation independently of a metabolic benefit or changes in adiposity.
Collapse
Affiliation(s)
- Ahmed K Aladhami
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina 29209, USA
- University of Baghdad, Nursing College, Baghdad, Iraq
| | - Christian A Unger
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina 29209, USA
| | - Marion C Hope
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina 29209, USA
| | - William E Cotham
- Department of Chemistry and Biochemistry, College of Arts and Science, University of South Carolina, Columbia, South Carolina 29208, USA
| | - Kandy T Velázquez
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina 29209, USA
| | - Reilly T Enos
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina 29209, USA
| |
Collapse
|
48
|
Congenital adiponectin deficiency mitigates high-fat-diet-induced obesity in gonadally intact male and female, but not in ovariectomized mice. Sci Rep 2022; 12:16668. [PMID: 36198723 PMCID: PMC9534911 DOI: 10.1038/s41598-022-21228-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 09/23/2022] [Indexed: 11/24/2022] Open
Abstract
Epidemiological literature indicates that women are less susceptible to type II diabetes (T2D) than males. The general consensus is that estrogen is protective, whereas its deficiency in post-menopause is associated with adiposity and impaired insulin sensitivity. However, epidemiological data suggests that males are more prone to developing T2D, and at a lower BMI, compared to females during post-menopausal years; suggesting that another factor, other than estrogen, protects females. We proposed to determine if adiponectin (APN) serves as this protective factor. An initial experiment was performed in which gonadally intact male and female mice were fed either a purified low-fat diet (LFD) or high-fat diet (HFD) (40% kcals from fat) for 16 weeks. An additional group of HFD ovariectomy (OVX) mice were included to assess estrogen deficiency’s impact on obesity. Body composition, adipose tissue inflammation, ectopic lipid accumulation as well as glucose metabolism and insulin resistance were assessed. In corroboration with previous data, estrogen deficiency (OVX) exacerbated HFD-induced obesity in female mice. However, despite a higher body fat percentage and a similar degree of hepatic and skeletal muscle lipid accumulation, female OVX HFD-fed mice exhibited enhanced insulin sensitivity relative to HFD-fed males. Therefore, a subsequent HFD experiment was performed utilizing male and female (both gonadally intact and OVX) APN deficient mice (APN−/−) and wildtype littermates to determine if APN is the factor which protects OVX females from the similar degree of metabolic dysfunction as males in the setting of obesity. Indirect calorimetry was used to determine observed phenotype differences. APN deficiency limited adiposity and mitigated HFD-induced insulin resistance and adipose tissue inflammation in gonadally intact male and female, but not in OVX mice. Using indirect calorimetry, we uncovered that slight, but non-statistically significant differences in food intake and energy expenditure leading to a net difference in energy balance likely explain the reduced body weight exhibited by male APN-deficient mice. In conclusion, congenital APN deficiency is protective against obesity development in gonadally intact mice, however, in the setting of estrogen deficiency (OVX) this is not true. These findings suggest that gonadal status dictates the protective effects of congenital APN deficiency in the setting of HFD-induced obesity.
Collapse
|
49
|
Nicotinamide riboside kinase 1 protects against diet and age-induced pancreatic β-cell failure. Mol Metab 2022; 66:101605. [PMID: 36165811 PMCID: PMC9557729 DOI: 10.1016/j.molmet.2022.101605] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 09/10/2022] [Accepted: 09/16/2022] [Indexed: 12/29/2022] Open
Abstract
OBJECTIVE Disturbances in NAD+ metabolism have been described as a hallmark for multiple metabolic and age-related diseases, including type 2 diabetes. While alterations in pancreatic β-cell function are critical determinants of whole-body glucose homeostasis, the role of NAD+ metabolism in the endocrine pancreas remains poorly explored. Here, we aimed to evaluate the role of nicotinamide riboside (NR) metabolism in maintaining NAD+ levels and pancreatic β-cell function in pathophysiological conditions. METHODS Whole body and pancreatic β-cell-specific NRK1 knockout (KO) mice were metabolically phenotyped in situations of high-fat feeding and aging. We also analyzed pancreatic β-cell function, β-cell mass and gene expression. RESULTS We first demonstrate that NRK1, the essential enzyme for the utilization of NR, is abundantly expressed in pancreatic β-cells. While NR treatment did not alter glucose-stimulated insulin secretion in pancreatic islets from young healthy mice, NRK1 knockout mice displayed glucose intolerance and compromised β-cells response to a glucose challenge upon high-fat feeding or aging. Interestingly, β cell dysfunction stemmed from the functional failure of other organs, such as liver and kidney, and the associated changes in circulating peptides and hormones, as mice lacking NRK1 exclusively in β-cells did not show altered glucose homeostasis. CONCLUSIONS This work unveils a new physiological role for NR metabolism in the maintenance of glucose tolerance and pancreatic β-cell function in high-fat feeding or aging conditions.
Collapse
|
50
|
Costa AJ, Oliveira RB, Wachilewski P, Nishino MS, Bassani TB, Stilhano RS, Cerutti JM, Nozima B, Porto CS, Pereira GJDS, Ramirez AL, Smaili SS, Ureshino RP. Membrane estrogen receptor ERα activation improves tau clearance via autophagy induction in a tauopathy cell model. Brain Res 2022; 1795:148079. [PMID: 36088959 DOI: 10.1016/j.brainres.2022.148079] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 08/26/2022] [Accepted: 09/04/2022] [Indexed: 11/16/2022]
Abstract
Alzheimer's disease (AD) is the most prevalent aging-associated neurodegenerative disease, with a higher incidence in women than men. There is evidence that sex hormone replacement therapy, particularly estrogen, reduces memory loss in menopausal women. Neurofibrillary tangles are associated with tau protein aggregation, a characteristic of AD and other tauopathies. In this sense, autophagy is a promising cellular process to remove these protein aggregates. This study evaluated the autophagy mechanisms involved in neuroprotection induced by 17β-estradiol (E2) in a Tet-On inducible expression tauopathy cell model (EGFP-tau WT or with the P301L mutation, 0N4R isoform). The results indicated that 17β-estradiol induces autophagy by activating AMPK in a concentration-dependent manner, independent of mTOR signals. The estrogen receptor α (ERα) agonist, PPT, also induced autophagy, while the ERα antagonist, MPP, substantially attenuated the 17β-estradiol-mediated autophagy induction. Notably, 17β-estradiol increased LC3-II levels and phosphorylated and total tau protein clearance in the EGFP-tau WT cell line but not in EGPF-tau P301L. Similar results were observed with E2-BSA, a plasma membrane-impermeable estrogen, suggesting membrane ERα involvement in non-genomic estrogenic pathway activation. Furthermore, 17β-estradiol-induced autophagy led to EGFP-tau protein clearance. These results demonstrate that modulating autophagy via the estrogenic pathway may represent a new therapeutic target for treating AD.
Collapse
Affiliation(s)
- Angelica Jardim Costa
- Universidade Federal de São Paulo, Escola Paulista de Medicina, Department of Pharmacology, São Paulo, SP, Brazil
| | - Rafaela Brito Oliveira
- Universidade Federal de São Paulo, Instituto de Ciências Ambientais, Químicas e Farmacêuticas, Department of Biological Sciences, Diadema, SP, Brazil; Universidade Federal de São Paulo, Escola Paulista de Medicina, Laboratory of Molecular and Translational Endocrinology, São Paulo, SP, Brazil
| | - Patrícia Wachilewski
- Universidade Federal de São Paulo, Instituto de Ciências Ambientais, Químicas e Farmacêuticas, Department of Biological Sciences, Diadema, SP, Brazil; Universidade Federal de São Paulo, Escola Paulista de Medicina, Laboratory of Molecular and Translational Endocrinology, São Paulo, SP, Brazil
| | - Michelle Sayuri Nishino
- Universidade Federal de São Paulo, Instituto de Ciências Ambientais, Químicas e Farmacêuticas, Department of Biological Sciences, Diadema, SP, Brazil; Universidade Federal de São Paulo, Escola Paulista de Medicina, Laboratory of Molecular and Translational Endocrinology, São Paulo, SP, Brazil
| | - Taysa Bervian Bassani
- Universidade Federal de São Paulo, Instituto de Ciências Ambientais, Químicas e Farmacêuticas, Department of Biological Sciences, Diadema, SP, Brazil; Universidade Federal de São Paulo, Escola Paulista de Medicina, Laboratory of Molecular and Translational Endocrinology, São Paulo, SP, Brazil
| | - Roberta Sessa Stilhano
- Department of Physiological Sciences, Faculdade de Ciências Médicas da Santa Casa de São Paulo, São Paulo, SP, Brazil
| | - Janete Maria Cerutti
- Universidade Federal de São Paulo, Escola Paulista de Medicina, Department of Morphology and Genetics, São Paulo, SP, Brazil
| | - Bruno Nozima
- Universidade Federal de São Paulo, Escola Paulista de Medicina, Department of Morphology and Genetics, São Paulo, SP, Brazil
| | - Catarina Segreti Porto
- Universidade Federal de São Paulo, Escola Paulista de Medicina, Department of Pharmacology, São Paulo, SP, Brazil
| | | | | | - Soraya Soubhi Smaili
- Universidade Federal de São Paulo, Escola Paulista de Medicina, Department of Pharmacology, São Paulo, SP, Brazil
| | - Rodrigo Portes Ureshino
- Universidade Federal de São Paulo, Instituto de Ciências Ambientais, Químicas e Farmacêuticas, Department of Biological Sciences, Diadema, SP, Brazil; Universidade Federal de São Paulo, Escola Paulista de Medicina, Laboratory of Molecular and Translational Endocrinology, São Paulo, SP, Brazil.
| |
Collapse
|