1
|
Yuan Y, Shin S, Shi Z, Shu G, Jiang Y. Postnatal Tamoxifen Exposure Induces Long-Lasting Changes to Adipose Tissue in Adult Mice. Mol Biotechnol 2024; 66:2322-2331. [PMID: 37642828 DOI: 10.1007/s12033-023-00828-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 07/14/2023] [Indexed: 08/31/2023]
Abstract
Tamoxifen (TAM) is commonly administered to a variety of inducible or conditional transgenic mice that contain Cre recombinase fused with ER. While the impacts of adult TAM treatment are well documented in the field of adipose biology, the long-term effects of postnatal TAM treatment on adult life are still understudied. In this study, we investigated whether postnatal TAM treatment had long-lasting effects on adult body composition and adiposity in male and female mice, fed either with chow or a high-fat diet (HFD). We found that postnatal, but not adult, TAM treatment had long-lasting impacts on female mice, resulting in lower body weight, lower fat mass, and smaller adipocytes. In contrast, postnatal exposure to TAM impaired male but not female cold-induced adipose beiging capacity. Interestingly, upon HFD feeding, the sex-dependent effects of TAM on adult life disappeared, and both female and male mice showed a more obese phenotype with impaired glucose tolerance. These findings suggest that postnatal TAM injection exerts a long-lasting impact on adipose tissue in adult life in a sex- and diet-dependent manner.
Collapse
Affiliation(s)
- Yexian Yuan
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Sunhye Shin
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
- Major of Food and Nutrition, Division of Applied Food System, Seoul Women's University, Seoul, 01797, Korea
| | - Zuoxiao Shi
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Gang Shu
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Yuwei Jiang
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
2
|
Yang W, Jiang W, Liao W, Yan H, Ai W, Pan Q, Brashear WA, Xu Y, He L, Guo S. An estrogen receptor α-derived peptide improves glucose homeostasis during obesity. Nat Commun 2024; 15:3410. [PMID: 38649684 PMCID: PMC11035554 DOI: 10.1038/s41467-024-47687-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 04/10/2024] [Indexed: 04/25/2024] Open
Abstract
Estrogen receptor α (ERα) plays a crucial role in regulating glucose and energy homeostasis during type 2 diabetes mellitus (T2DM). However, the underlying mechanisms remain incompletely understood. Here we find a ligand-independent effect of ERα on the regulation of glucose homeostasis. Deficiency of ERα in the liver impairs glucose homeostasis in male, female, and ovariectomized (OVX) female mice. Mechanistic studies reveal that ERα promotes hepatic insulin sensitivity by suppressing ubiquitination-induced IRS1 degradation. The ERα 1-280 domain mediates the ligand-independent effect of ERα on insulin sensitivity. Furthermore, we identify a peptide based on ERα 1-280 domain and find that ERα-derived peptide increases IRS1 stability and enhances insulin sensitivity. Importantly, administration of ERα-derived peptide into obese mice significantly improves glucose homeostasis and serum lipid profiles. These findings pave the way for the therapeutic intervention of T2DM by targeting the ligand-independent effect of ERα and indicate that ERα-derived peptide is a potential insulin sensitizer for the treatment of T2DM.
Collapse
Affiliation(s)
- Wanbao Yang
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX, 77843, USA
| | - Wen Jiang
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX, 77843, USA
| | - Wang Liao
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX, 77843, USA
| | - Hui Yan
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX, 77843, USA
| | - Weiqi Ai
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX, 77843, USA
| | - Quan Pan
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX, 77843, USA
| | - Wesley A Brashear
- High Performance Research Computing, Texas A&M University, College Station, TX, USA
| | - Yong Xu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Ling He
- Departments of Pediatrics and Pharmacology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Shaodong Guo
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX, 77843, USA.
| |
Collapse
|
3
|
Nedyalkova M, Robeva R, Romanova J, Yovcheva K, Lattuada M, Simeonov V. In silico screening of potential agonists of a glucagon-like peptide-1 receptor among female sex hormone derivatives. J Biomol Struct Dyn 2024:1-12. [PMID: 38587907 DOI: 10.1080/07391102.2024.2330714] [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: 08/16/2023] [Accepted: 03/09/2024] [Indexed: 04/10/2024]
Abstract
Glucagon-like peptide-1 (GLP-1) is an intestinal hormone that exerts its pleiotropic effects through a specific GLP-1 receptor (GLP-1R). The hormone-receptor complex might regulate glucose-dependent insulin secretion, and energy homeostasis; moreover, it could decrease inflammation and provide cardio- and neuroprotection. Additionally, the beneficial influence of GLP-1 on obesity in women might lead to improvement of their ovarian function. The links between metabolism and reproduction are tightly connected, and it is not surprising that different estrogen derivatives, estrogen-receptor modulator (SERM) and progestins used for gonadal and oncological disorders might influence carbohydrate and lipid metabolism. However, their possible influence on the GLP-1R has not been studied. The docking scores and top-ranked poses of raloxifene were much higher than those observed for other investigated SERMs and estradiol per se. Among different studied progestins, drospirenone showed slightly higher affinity to GLP-1R. Herein, the same data set of the drugs is evaluated by molecular dynamics (MD) simulations and compared with the obtained docking result. Notably, it is demonstrated that the used docking protocol and the applied MD calculations ranked the same ligand (raloxifene) as the best one. In the present study, raloxifene might exert an allosteric influence on GLP-1R signaling, which might contribute to potential beneficial effects on metabolism and weight regulation. However, further experimental and clinical studies are needed to reveal if the GLP-1R modulation has a real biological impact.
Collapse
Affiliation(s)
- Miroslava Nedyalkova
- Department of Chemistry, Fribourg University, Fribourg, Switzerland
- Department of Inorganic Chemistry, Faculty of Chemistry and Pharmacy, University of Sofia 'St. Kl. Ohridski', Sofia, Bulgaria
- Swiss National Center for Competence in Research (NCCR) Bio-inspired Materials, University of Fribourg, Fribourg, Switzerland
| | - Ralitsa Robeva
- Department of Endocrinology, Faculty of Medicine, Medical University-Sofia, Sofia, Bulgaria
| | - Julia Romanova
- Department of Inorganic Chemistry, Faculty of Chemistry and Pharmacy, University of Sofia 'St. Kl. Ohridski', Sofia, Bulgaria
| | - Kirila Yovcheva
- Department of Inorganic Chemistry, Faculty of Chemistry and Pharmacy, University of Sofia 'St. Kl. Ohridski', Sofia, Bulgaria
| | - Marco Lattuada
- Department of Chemistry, Fribourg University, Fribourg, Switzerland
| | - Vasil Simeonov
- Department of Inorganic Chemistry, Faculty of Chemistry and Pharmacy, University of Sofia 'St. Kl. Ohridski', Sofia, Bulgaria
| |
Collapse
|
4
|
Thomas NS, Scalzo RL, Wellberg EA. Diabetes mellitus in breast cancer survivors: metabolic effects of endocrine therapy. Nat Rev Endocrinol 2024; 20:16-26. [PMID: 37783846 PMCID: PMC11487546 DOI: 10.1038/s41574-023-00899-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/01/2023] [Indexed: 10/04/2023]
Abstract
Breast cancer is the most common invasive malignancy in the world, with millions of survivors living today. Type 2 diabetes mellitus (T2DM) is also a globally prevalent disease that is a widely studied risk factor for breast cancer. Most breast tumours express the oestrogen receptor and are treated with systemic therapies designed to disrupt oestrogen-dependent signalling. Since the advent of targeted endocrine therapy six decades ago, the mortality from breast cancer has steadily declined; however, during the past decade, an elevated risk of T2DM after breast cancer treatment has been reported, particularly for those who received endocrine therapy. In this Review, we highlight key events in the history of endocrine therapies, beginning with the development of tamoxifen. We also summarize the sequence of reported adverse metabolic effects, which include dyslipidaemia, hepatic steatosis and impaired glucose tolerance. We discuss the limitations of determining a causal role for breast cancer treatments in T2DM development from epidemiological data and describe informative preclinical studies that suggest complex mechanisms through which endocrine therapy might drive T2DM risk and progression. We also reinforce the life-saving benefits of endocrine therapy and highlight the need for better predictive biomarkers of T2DM risk and preventive strategies for the growing population of breast cancer survivors.
Collapse
Affiliation(s)
- Nisha S Thomas
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, Oklahoma City, OK, USA
- Harold Hamm Diabetes Center, Oklahoma City, OK, USA
| | - Rebecca L Scalzo
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Rocky Mountain Regional VA Medical Center, Aurora, CO, USA
| | - Elizabeth A Wellberg
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Stephenson Cancer Center, Oklahoma City, OK, USA.
- Harold Hamm Diabetes Center, Oklahoma City, OK, USA.
| |
Collapse
|
5
|
Fang Z, Xu H, Duan J, Ruan B, Liu J, Song P, Ding J, Xu C, Li Z, Dou K, Wang L. Short-term tamoxifen administration improves hepatic steatosis and glucose intolerance through JNK/MAPK in mice. Signal Transduct Target Ther 2023; 8:94. [PMID: 36864030 PMCID: PMC9981902 DOI: 10.1038/s41392-022-01299-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 11/30/2022] [Accepted: 12/22/2022] [Indexed: 03/04/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) which is a leading cause of chronic liver diseases lacks effective treatment. Tamoxifen has been proven to be the first-line chemotherapy for several solid tumors in clinics, however, its therapeutic role in NAFLD has never been elucidated before. In vitro experiments, tamoxifen protected hepatocytes against sodium palmitate-induced lipotoxicity. In male and female mice fed with normal diets, continuous tamoxifen administration inhibited lipid accumulation in liver, and improved glucose and insulin intolerance. Short-term tamoxifen administration largely improved hepatic steatosis and insulin resistance, however, the phenotypes manifesting inflammation and fibrosis remained unchanged in abovementioned models. In addition, mRNA expressions of genes related to lipogenesis, inflammation, and fibrosis were downregulated by tamoxifen treatment. Moreover, the therapeutic effect of tamoxifen on NAFLD was not gender or ER dependent, as male and female mice with metabolic disorders shared no difference in response to tamoxifen and ER antagonist (fulvestrant) did not abolish its therapeutic effect as well. Mechanistically, RNA sequence of hepatocytes isolated from fatty liver revealed that JNK/MAPK signaling pathway was inactivated by tamoxifen. Pharmacological JNK activator (anisomycin) partially deprived the therapeutic role of tamoxifen in treating hepatic steatosis, proving tamoxifen improved NAFLD in a JNK/MAPK signaling-dependent manner.
Collapse
Affiliation(s)
- Zhiqiang Fang
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Hao Xu
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Juanli Duan
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Bai Ruan
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, Fourth Military Medical University, Xi'an, 710032, China.,Center of Clinical Aerospace Medicine & Department of Aviation Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Jingjing Liu
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Ping Song
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Jian Ding
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Chen Xu
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Zhiwen Li
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Kefeng Dou
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| | - Lin Wang
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
6
|
Abstract
Signaling via extracellular regulated kinase 1/2 (ERK1/2) and p90 ribosomal S6 kinase (RSK), a downstream effector, mediates numerous processes. For example, ERK1/2-RSK signaling is essential for estrogen homeostasis in the mammary gland and uterus to maintain physiological responsiveness. This review will focus on the coordination of ERK1/2-RSK2 and estrogen signaling through estrogen receptor alpha (ERα). The interrelationship and the feedback mechanisms between these pathways occurs at the level of transcription, translation, and posttranslational modification. Identifying how ERK1/2-RSK2 and estrogen signaling cooperate in homeostasis and disease may lead to novel therapeutic approaches in estrogen-dependent disorders.
Collapse
Affiliation(s)
- Deborah A Lannigan
- Correspondence: Deborah A. Lannigan, PhD, Department of Pathology, Microbiology & Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| |
Collapse
|
7
|
Bulut G, Atci N. Sarcopenic obesity in early breast cancer patients with metabolic syndrome: a cross-sectional study. Future Oncol 2022; 18:2489-2498. [PMID: 35543029 DOI: 10.2217/fon-2022-0155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: The importance of sarcopenic obesity (SO) in malignancy has recently been recognized. The authors aimed to investigate the clinical and body composition data of early-stage breast cancer (BC) patients by focusing on the components of metabolic syndrome (MetS) and SO. Methods: In this cross-sectional study with early-stage BC patients, the clinical and laboratory results were evaluated for diagnosing MetS. Bioimpedance analysis and muscle radiation attenuation (Hounsfield unit average calculation [HUAC]) using acquired computed tomography images were used to assess SO. Results: The age at diagnosis, BMI, visceral fat ratio, hemoglobin A1c and carcinoembryonic antigen levels in patients with MetS were significantly higher (p < 0.05). A cutoff value of 39.4 Hounsfield units was significantly related to MetS with 76.5% sensitivity and 62% specificity. The mean HUAC in patients with MetS (36.9) was significantly lower than in those without MetS (41.4; p = 0.017). Conclusion: HUAC value predicted SO in early-stage BC patients with MetS.
Collapse
Affiliation(s)
- Gulcan Bulut
- International Medicana Hospital, Department of Medical Oncology, Izmir, 35170, Turkey
| | - Nesrin Atci
- Mozaik Hospital, Department of Radiology, Hatay, 31070, Turkey
| |
Collapse
|
8
|
Estrogenic Action in Stress-Induced Neuroendocrine Regulation of Energy Homeostasis. Cells 2022; 11:cells11050879. [PMID: 35269500 PMCID: PMC8909319 DOI: 10.3390/cells11050879] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/16/2022] [Accepted: 02/28/2022] [Indexed: 01/27/2023] Open
Abstract
Estrogens are among important contributing factors to many sex differences in neuroendocrine regulation of energy homeostasis induced by stress. Research in this field is warranted since chronic stress-related psychiatric and metabolic disturbances continue to be top health concerns, and sex differences are witnessed in these aspects. For example, chronic stress disrupts energy homeostasis, leading to negative consequences in the regulation of emotion and metabolism. Females are known to be more vulnerable to the psychological consequences of stress, such as depression and anxiety, whereas males are more vulnerable to the metabolic consequences of stress. Sex differences that exist in the susceptibility to various stress-induced disorders have led researchers to hypothesize that gonadal hormones are regulatory factors that should be considered in stress studies. Further, estrogens are heavily recognized for their protective effects on metabolic dysregulation, such as anti-obesogenic and glucose-sensing effects. Perturbations to energy homeostasis using laboratory rodents, such as physiological stress or over-/under- feeding dietary regimen prevalent in today’s society, offer hints to the underlying mechanisms of estrogenic actions. Metabolic effects of estrogens primarily work through estrogen receptor α (ERα), which is differentially expressed between the sexes in hypothalamic nuclei regulating energy metabolism and in extrahypothalamic limbic regions that are not typically associated with energy homeostasis. In this review, we discuss estrogenic actions implicated in stress-induced sex-distinct metabolic disorders.
Collapse
|
9
|
Wang L, Wang B, Gasek NS, Zhou Y, Cohn RL, Martin DE, Zuo W, Flynn WF, Guo C, Jellison ER, Kim T, Prata LGPL, Palmer AK, Li M, Inman CL, Barber LS, Al-Naggar IMA, Zhou Y, Du W, Kshitiz, Kuchel GA, Meves A, Tchkonia T, Kirkland JL, Robson P, Xu M. Targeting p21 Cip1 highly expressing cells in adipose tissue alleviates insulin resistance in obesity. Cell Metab 2022; 34:75-89.e8. [PMID: 34813734 PMCID: PMC8732323 DOI: 10.1016/j.cmet.2021.11.002] [Citation(s) in RCA: 92] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 09/30/2021] [Accepted: 11/03/2021] [Indexed: 01/07/2023]
Abstract
Insulin resistance is a pathological state often associated with obesity, representing a major risk factor for type 2 diabetes. Limited mechanism-based strategies exist to alleviate insulin resistance. Here, using single-cell transcriptomics, we identify a small, critically important, but previously unexamined cell population, p21Cip1 highly expressing (p21high) cells, which accumulate in adipose tissue with obesity. By leveraging a p21-Cre mouse model, we demonstrate that intermittent clearance of p21high cells can both prevent and alleviate insulin resistance in obese mice. Exclusive inactivation of the NF-κB pathway within p21high cells, without killing them, attenuates insulin resistance. Moreover, fat transplantation experiments establish that p21high cells within fat are sufficient to cause insulin resistance in vivo. Importantly, a senolytic cocktail, dasatinib plus quercetin, eliminates p21high cells in human fat ex vivo and mitigates insulin resistance following xenotransplantation into immuno-deficient mice. Our findings lay the foundation for pursuing the targeting of p21high cells as a new therapy to alleviate insulin resistance.
Collapse
Affiliation(s)
- Lichao Wang
- UConn Center on Aging, UConn Health, Farmington, CT 06030, USA; Department of Genetics and Genome Sciences, UConn Health, Farmington, CT 06030, USA
| | - Binsheng Wang
- UConn Center on Aging, UConn Health, Farmington, CT 06030, USA; Department of Genetics and Genome Sciences, UConn Health, Farmington, CT 06030, USA
| | - Nathan S Gasek
- UConn Center on Aging, UConn Health, Farmington, CT 06030, USA; Department of Genetics and Genome Sciences, UConn Health, Farmington, CT 06030, USA
| | - Yueying Zhou
- Xiangya Stomatological Hospital, Central South University, Changsha, 86-410000, China; Center for Regenerative Medicine and Skeletal Development, UConn Health, Farmington, CT 06030, USA
| | - Rachel L Cohn
- UConn Center on Aging, UConn Health, Farmington, CT 06030, USA; Department of Genetics and Genome Sciences, UConn Health, Farmington, CT 06030, USA
| | - Dominique E Martin
- UConn Center on Aging, UConn Health, Farmington, CT 06030, USA; Biomedical Science Graduate Program, UConn Health, Farmington, CT 06030, USA
| | - Wulin Zuo
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06030, USA
| | - William F Flynn
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06030, USA
| | - Chun Guo
- UConn Center on Aging, UConn Health, Farmington, CT 06030, USA
| | - Evan R Jellison
- Department of Immunology, UConn Health, Farmington, CT 06030, USA
| | - Taewan Kim
- UConn Center on Aging, UConn Health, Farmington, CT 06030, USA; Biomedical Science Graduate Program, UConn Health, Farmington, CT 06030, USA
| | | | - Allyson K Palmer
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
| | - Ming Li
- Department of Dermatology, Mayo Clinic, Rochester, MN 55905, USA
| | - Christina L Inman
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
| | - Lauren S Barber
- UConn Center on Aging, UConn Health, Farmington, CT 06030, USA
| | | | - Yanjiao Zhou
- Department of Medicine, UConn Health, Farmington, CT 06030, USA
| | - Wenqiang Du
- Department of Biomedical Engineering, UConn Health, Farmington, CT 06030, USA
| | - Kshitiz
- Department of Biomedical Engineering, UConn Health, Farmington, CT 06030, USA
| | - George A Kuchel
- UConn Center on Aging, UConn Health, Farmington, CT 06030, USA
| | - Alexander Meves
- Department of Dermatology, Mayo Clinic, Rochester, MN 55905, USA
| | - Tamar Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
| | - Paul Robson
- Department of Genetics and Genome Sciences, UConn Health, Farmington, CT 06030, USA; The Jackson Laboratory for Genomic Medicine, Farmington, CT 06030, USA
| | - Ming Xu
- UConn Center on Aging, UConn Health, Farmington, CT 06030, USA; Department of Genetics and Genome Sciences, UConn Health, Farmington, CT 06030, USA.
| |
Collapse
|
10
|
Adlanmerini M, Fontaine C, Gourdy P, Arnal JF, Lenfant F. Segregation of nuclear and membrane-initiated actions of estrogen receptor using genetically modified animals and pharmacological tools. Mol Cell Endocrinol 2022; 539:111467. [PMID: 34626731 DOI: 10.1016/j.mce.2021.111467] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 09/06/2021] [Accepted: 09/28/2021] [Indexed: 11/23/2022]
Abstract
Estrogen receptor alpha (ERα) and beta (ERβ) are members of the nuclear receptor superfamily, playing widespread functions in reproductive and non-reproductive tissues. Beside the canonical function of ERs as nuclear receptors, in this review, we summarize our current understanding of extra-nuclear, membrane-initiated functions of ERs with a specific focus on ERα. Over the last decade, in vivo evidence has accumulated to demonstrate the physiological relevance of this ERα membrane-initiated-signaling from mouse models to selective pharmacological tools. Finally, we discuss the perspectives and future challenges opened by the integration of extra-nuclear ERα signaling in physiology and pathology of estrogens.
Collapse
Affiliation(s)
- Marine Adlanmerini
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U1297, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France
| | - Coralie Fontaine
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U1297, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France
| | - Pierre Gourdy
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U1297, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France
| | - Jean-François Arnal
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U1297, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France
| | - Françoise Lenfant
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U1297, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France.
| |
Collapse
|
11
|
Daniels Gatward LF, Kennard MR, Smith LIF, King AJF. The use of mice in diabetes research: The impact of physiological characteristics, choice of model and husbandry practices. Diabet Med 2021; 38:e14711. [PMID: 34614258 DOI: 10.1111/dme.14711] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 10/04/2021] [Indexed: 12/24/2022]
Abstract
Diabetes mellitus is characterised by hyperglycaemia, which results from an absolute or relative lack of insulin. Chronic and acute hyperglycaemia are associated with a range of health complications and an overall increased risk of mortality. Mouse models are vital in understanding the pathogenesis of this disease and its complications, as well as for developing new diabetes therapeutics. However, for experimental questions to be suitably tested, it is critical that factors inherent to the animal model are considered, as these can have profound impacts on experimental outcome, data reproducibility and robustness. In this review, we discuss key considerations relating to model choice, physiological characteristics (such as age, sex and genetic background) and husbandry practices and explore the impact of these on common experimental readouts used in preclinical diabetes research.
Collapse
|
12
|
Alemany M. Estrogens and the regulation of glucose metabolism. World J Diabetes 2021; 12:1622-1654. [PMID: 34754368 PMCID: PMC8554369 DOI: 10.4239/wjd.v12.i10.1622] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/10/2021] [Accepted: 04/14/2021] [Indexed: 02/06/2023] Open
Abstract
The main estrogens: estradiol, estrone, and their acyl-esters have been studied essentially related to their classical estrogenic and pharmacologic functions. However, their main effect in the body is probably the sustained control of core energy metabolism. Estrogen nuclear and membrane receptors show an extraordinary flexibility in the modulation of metabolic responses, and largely explain gender and age differences in energy metabolism: part of these mechanisms is already sufficiently known to justify both. With regard to energy, the estrogen molecular species act essentially through four key functions: (1) Facilitation of insulin secretion and control of glucose availability; (2) Modulation of energy partition, favoring the use of lipid as the main energy substrate when more available than carbohydrates; (3) Functional protection through antioxidant mechanisms; and (4) Central effects (largely through neural modulation) on whole body energy management. Analyzing the different actions of estrone, estradiol and their acyl esters, a tentative classification based on structure/effects has been postulated. Either separately or as a group, estrogens provide a comprehensive explanation that not all their quite diverse actions are related solely to specific molecules. As a group, they constitute a powerful synergic action complex. In consequence, estrogens may be considered wardens of energy homeostasis.
Collapse
Affiliation(s)
- Marià Alemany
- Faculty of Biology, University of Barcelona, Barcelona 08028, Catalonia, Spain
| |
Collapse
|
13
|
Estrada-Meza J, Videlo J, Bron C, Saint-Béat C, Silva M, Duboeuf F, Peyruchaud O, Rajas F, Mithieux G, Gautier-Stein A. Tamoxifen Treatment in the Neonatal Period Affects Glucose Homeostasis in Adult Mice in a Sex-Dependent Manner. Endocrinology 2021; 162:6277101. [PMID: 33999998 DOI: 10.1210/endocr/bqab098] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Indexed: 12/11/2022]
Abstract
Tamoxifen is a selective estrogen receptor modulator used to activate the CREERT2 recombinase, allowing tissue-specific and temporal control of the somatic mutagenesis to generate transgenic mice. Studies integrating development and metabolism require a genetic modification induced by a neonatal tamoxifen administration. Here, we investigate the effects of a neonatal tamoxifen administration on energy homeostasis in adult male and female C57BL/6J mice. C57BL/6J male and female mouse pups received a single injection of tamoxifen 1 day after birth (NTT) and were fed a high-fat/high-sucrose diet at 6 weeks of age. We measured weight, body composition, glucose and insulin tolerance, basal metabolism, and tibia length and weight in adult mice. The neonatal tamoxifen administration exerted long-term, sex-dependent effects on energy homeostasis. NTT female mice became overweight and developed impaired glucose control in comparison to vehicle-treated littermates. NTT females exhibited 60% increased fat mass, increased food intake, decreased physical activity and energy expenditure, impaired glucose and insulin tolerance, and fasting hyperglycemia and hyperinsulinemia. In contrast, NTT male mice exhibited a modest amelioration of glucose and insulin tolerance and long-term decreased lean mass linked to decreased bone weight. These results suggest that the neonatal tamoxifen administration exerted a marked and sex-dependent influence on adult energy homeostasis and bone weight and must therefore be used with caution for the development of transgenic mouse models regarding studies on energy homeostasis and bone biology.
Collapse
Affiliation(s)
- Judith Estrada-Meza
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, Lyon, France
| | - Jasmine Videlo
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, Lyon, France
| | - Clara Bron
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, Lyon, France
| | - Cécile Saint-Béat
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, Lyon, France
| | - Marine Silva
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, Lyon, France
| | - François Duboeuf
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1033, Lyon, France
| | - Olivier Peyruchaud
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1033, Lyon, France
| | - Fabienne Rajas
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, Lyon, France
| | - Gilles Mithieux
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, Lyon, France
| | | |
Collapse
|
14
|
Chen Q, Wang B, Wang S, Qian X, Li X, Zhao J, Zhang H, Chen W, Wang G. Modulation of the Gut Microbiota Structure with Probiotics and Isoflavone Alleviates Metabolic Disorder in Ovariectomized Mice. Nutrients 2021; 13:1793. [PMID: 34070274 PMCID: PMC8225012 DOI: 10.3390/nu13061793] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/12/2021] [Accepted: 05/21/2021] [Indexed: 02/06/2023] Open
Abstract
The decrease in ovarian hormone secretion that occurs during menopause results in an increase in body weight and adipose tissue mass. Probiotics and soy isoflavones (SIFs) could affect the gut microbiota and exert anti-obesity effects. The objective of this study was to investigate the effects of probiotics and a diet containing SIF (SIF diet) on ovariectomized mice with menopausal obesity, including the gut microbiome. The results demonstrate that Bifidobacterium longum 15M1 can reverse menopausal obesity, whilst the combination of Lactobacillus plantarum 30M5 and a SIF diet was more effective in alleviating menopausal lipid metabolism disorder than either components alone. Probiotics and SIFs play different anti-obesity roles in menopausal mice. Furthermore, 30M5 alters the metabolites of the gut microbiota that increase the circulating estrogen level, upregulates the expression of estrogen receptor α in abdominal adipose tissue and improves the production of short-chain fatty acids (SCFAs). A SIF diet can significantly alter the structure of the fecal bacterial community and enrich the pathways related to SCFAs production. Moreover, 30M5 and a SIF diet acted synergistically to effectively resolve abnormal serum lipid levels in ovariectomized mice, and these effects appear to be associated with regulation of the diversity and structure of the intestinal microbiota to enhance SCFAs production and promote estrogen circulation.
Collapse
Affiliation(s)
- Qian Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (Q.C.); (B.W.); (S.W.); (X.Q.); (X.L.); (J.Z.); (H.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Botao Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (Q.C.); (B.W.); (S.W.); (X.Q.); (X.L.); (J.Z.); (H.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Shunhe Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (Q.C.); (B.W.); (S.W.); (X.Q.); (X.L.); (J.Z.); (H.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Xin Qian
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (Q.C.); (B.W.); (S.W.); (X.Q.); (X.L.); (J.Z.); (H.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Xiu Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (Q.C.); (B.W.); (S.W.); (X.Q.); (X.L.); (J.Z.); (H.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (Q.C.); (B.W.); (S.W.); (X.Q.); (X.L.); (J.Z.); (H.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi 214122, China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (Q.C.); (B.W.); (S.W.); (X.Q.); (X.L.); (J.Z.); (H.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
- Wuxi Translational Medicine Research Center and Jiangsu Translational Medicine Research Institute Wuxi Branch, Wuxi 214122, China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (Q.C.); (B.W.); (S.W.); (X.Q.); (X.L.); (J.Z.); (H.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Gang Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (Q.C.); (B.W.); (S.W.); (X.Q.); (X.L.); (J.Z.); (H.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi 214122, China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, China
| |
Collapse
|
15
|
Zahreddine R, Davezac M, Smirnova N, Buscato M, Lhuillier E, Lupieri A, Solinhac R, Vinel A, Vessieres E, Henrion D, Renault MA, Gadeau AP, Flouriot G, Lenfant F, Laffargue M, Métivier R, Arnal JF, Fontaine C. Tamoxifen Accelerates Endothelial Healing by Targeting ERα in Smooth Muscle Cells. Circ Res 2020; 127:1473-1487. [PMID: 33012251 DOI: 10.1161/circresaha.120.317062] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
RATIONALE Tamoxifen prevents the recurrence of breast cancer and is also beneficial against bone demineralization and arterial diseases. It acts as an ER (estrogen receptor) α antagonist in ER-positive breast cancers, whereas it mimics the protective action of 17β-estradiol in other tissues such as arteries. However, the mechanisms of these tissue-specific actions remain unclear. OBJECTIVE Here, we tested whether tamoxifen is able to accelerate endothelial healing and analyzed the underlying mechanisms. METHODS AND RESULTS Using 3 complementary mouse models of carotid artery injury, we demonstrated that both tamoxifen and estradiol accelerated endothelial healing, but only tamoxifen required the presence of the underlying medial smooth muscle cells. Chronic treatment with 17β-estradiol and tamoxifen elicited differential gene expression profiles in the carotid artery. The use of transgenic mouse models targeting either whole ERα in a cell-specific manner or ERα subfunctions (membrane/extranuclear versus genomic/transcriptional) demonstrated that 17β-estradiol-induced acceleration of endothelial healing is mediated by membrane ERα in endothelial cells, while the effect of tamoxifen is mediated by the nuclear actions of ERα in smooth muscle cells. CONCLUSIONS Whereas tamoxifen acts as an antiestrogen and ERα antagonist in breast cancer but also on the membrane ERα of endothelial cells, it accelerates endothelial healing through activation of nuclear ERα in smooth muscle cells, inviting to revisit the mechanisms of action of selective modulation of ERα.
Collapse
Affiliation(s)
- Rana Zahreddine
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, University of Toulouse 3, France (R.Z., M.D., N.S., M.B., E.L., A.L., R.S., A.V., F.L., M.L., J.-F.A., C.F.)
| | - Morgane Davezac
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, University of Toulouse 3, France (R.Z., M.D., N.S., M.B., E.L., A.L., R.S., A.V., F.L., M.L., J.-F.A., C.F.)
| | - Natalia Smirnova
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, University of Toulouse 3, France (R.Z., M.D., N.S., M.B., E.L., A.L., R.S., A.V., F.L., M.L., J.-F.A., C.F.)
| | - Melissa Buscato
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, University of Toulouse 3, France (R.Z., M.D., N.S., M.B., E.L., A.L., R.S., A.V., F.L., M.L., J.-F.A., C.F.)
| | - Emeline Lhuillier
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, University of Toulouse 3, France (R.Z., M.D., N.S., M.B., E.L., A.L., R.S., A.V., F.L., M.L., J.-F.A., C.F.)
| | - Adrien Lupieri
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, University of Toulouse 3, France (R.Z., M.D., N.S., M.B., E.L., A.L., R.S., A.V., F.L., M.L., J.-F.A., C.F.)
| | - Romain Solinhac
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, University of Toulouse 3, France (R.Z., M.D., N.S., M.B., E.L., A.L., R.S., A.V., F.L., M.L., J.-F.A., C.F.)
| | - Alexia Vinel
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, University of Toulouse 3, France (R.Z., M.D., N.S., M.B., E.L., A.L., R.S., A.V., F.L., M.L., J.-F.A., C.F.)
| | - Emilie Vessieres
- MITOVASC Institute, CARFI facility, INSERM U1083, UMR CNRS 6015, University of Angers, France (E.V., D.H.)
| | - Daniel Henrion
- MITOVASC Institute, CARFI facility, INSERM U1083, UMR CNRS 6015, University of Angers, France (E.V., D.H.)
| | - Marie-Ange Renault
- University of Bordeaux, INSERM, Biology of Cardiovascular Diseases, UMR 1034, Pessac, France (M.-A.R., A.-P.G.)
| | - Alain-Pierre Gadeau
- University of Bordeaux, INSERM, Biology of Cardiovascular Diseases, UMR 1034, Pessac, France (M.-A.R., A.-P.G.)
| | - Gilles Flouriot
- University of Rennes, EHESP, IRSET (Institut de Recherche en Santé, Environnement et Travail) - INSERM, UMR_S 1085, France (G.F.)
| | - Françoise Lenfant
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, University of Toulouse 3, France (R.Z., M.D., N.S., M.B., E.L., A.L., R.S., A.V., F.L., M.L., J.-F.A., C.F.)
| | - Muriel Laffargue
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, University of Toulouse 3, France (R.Z., M.D., N.S., M.B., E.L., A.L., R.S., A.V., F.L., M.L., J.-F.A., C.F.)
| | - Raphaël Métivier
- CNRS, Univeristy of Rennes, IGDR (Institut de Génétique De Rennes) - UMR 6290, France (R.M.)
| | - Jean-François Arnal
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, University of Toulouse 3, France (R.Z., M.D., N.S., M.B., E.L., A.L., R.S., A.V., F.L., M.L., J.-F.A., C.F.)
| | - Coralie Fontaine
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, University of Toulouse 3, France (R.Z., M.D., N.S., M.B., E.L., A.L., R.S., A.V., F.L., M.L., J.-F.A., C.F.)
| |
Collapse
|
16
|
DiStefano JK. NAFLD and NASH in Postmenopausal Women: Implications for Diagnosis and Treatment. Endocrinology 2020; 161:5890353. [PMID: 32776116 PMCID: PMC7473510 DOI: 10.1210/endocr/bqaa134] [Citation(s) in RCA: 134] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 08/03/2020] [Indexed: 12/13/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) prevalence in women is increasing worldwide. Women of reproductive age have lower rates of NAFLD compared with men; however, this protection is lost following the menopausal transition when NAFLD prevalence in postmenopausal women becomes similar to or surpasses that in age-matched male counterparts. Ongoing epidemiological, clinical, and experimental studies indicate greater NAFLD risk and higher rates of severe hepatic fibrosis in postmenopausal women relative to premenopausal women, and that older women with NAFLD experience greater mortality than men. Investigations involving ovariectomized animal models demonstrate a causal relationship between estrogen deficiency and heightened susceptibility to the development of fatty liver and steatohepatitis, although dietary factors may exacerbate this complex relationship. The accumulated findings suggest that a better understanding of the interplay among menopausal status, metabolic comorbidities, and sex steroids in NAFLD pathogenesis is needed. Further, the mechanisms underlying the difference in NAFLD risk between postmenopausal and premenopausal women remain incompletely understood. The goals of this review are to summarize studies of NAFLD risk in postmenopausal women, discuss results from animal models of estrogen deficiency, and explore the development of NAFD within the context of altered sex hormone profiles resulting from the menopausal transition. Potential implications for the prevention, diagnosis, and treatment of NAFLD in this relatively understudied cohort are also addressed.
Collapse
Affiliation(s)
- Johanna K DiStefano
- Diabetes and Fibrotic Disease Research Unit, Translational Genomics Research Institute, Phoenix, Arizona
- Correspondence: Johanna K. DiStefano, Diabetes and Fibrotic Disease Research Unit, Translational Genomics Research Institute, Phoenix, AZ, USA. E-mail:
| |
Collapse
|
17
|
Hepatic lipid metabolism in adult rats using early weaning models: sex-related differences. J Dev Orig Health Dis 2020; 11:499-508. [PMID: 32519629 DOI: 10.1017/s2040174420000495] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Non-pharmacological early weaning (NPEW) induces liver damage in male progeny at adulthood; however, pharmacological early weaning (PEW) does not cause this dysfunction. To elucidate this difference in liver dysfunction between these two models and determine the phenotype of female offspring, de novo lipogenesis, β-oxidation, very low-density lipoprotein (VLDL) export, and gluconeogenesis in both sexes were investigated in the adult Wistar rats that were weaned after a normal period of lactation (control group) or early weaned either by restriction of access to the dams' teats (NPEW group) or by reduction of dams' milk production with bromocriptine (PEW group). The offspring received standard diet from weaning to euthanasia (PN180). NPEW males had higher plasma triglycerides and TyG index, liver triglycerides, and cholesterol by de novo lipogenesis, which leads to intracellular lipids accumulation. As expected, hepatic morphology was preserved in PEW males, but they showed increased liver triglycerides. The only molecular difference between PEW and NPEW males was in acetyl-CoA carboxylase-1 (ACC-1) and stearoyl-CoA desaturase-1 (SCD-1), which were lower in PEW animals. Both early weaning (EW) females had no changes in liver cholesterol and triglyceride contents, and the hepatic cytoarchitecture was preserved. The expression of microsomal triglyceride transfer protein was increased in both the female EW groups, which could constitute a protective factor. The changes in hepatic lipid metabolism in EW offspring were less marked in females. EW impacted in the hepatic cytoarchitecture only in NPEW males, which showed higher ACC-1 and SCD-1 when compared to the PEW group. As these enzymes are lipogenic, it could explain a worsened liver function in NPEW males.
Collapse
|
18
|
The Disruption of Liver Metabolic Circadian Rhythms by a Cafeteria Diet Is Sex-Dependent in Fischer 344 Rats. Nutrients 2020; 12:nu12041085. [PMID: 32295282 PMCID: PMC7230270 DOI: 10.3390/nu12041085] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 04/10/2020] [Accepted: 04/11/2020] [Indexed: 11/30/2022] Open
Abstract
Circadian rhythms are ~24 h fluctuations of different biological processes that are regulated by the circadian clock system. They exert a major influence on most of the metabolism, such as the hepatic metabolism. This rhythmicity can be disrupted by obesogenic diets, fact that is considered to be a risk factor for the development of metabolic diseases. Nevertheless, obesogenic diets do not affect both genders in the same manner. We hypothesized that the circadian rhythms disruption of the hepatic metabolism, caused by obesogenic diets, is gender-dependent. Male and female Fischer 344 rats were fed either a standard diet or a cafeteria diet and sacrificed at two different moments, at zeitgeber 3 and 15. Only female rats maintained the circadian variations of the hepatic metabolism under a cafeteria diet. Most of those metabolites were related with the very low density lipoprotein (VLDL) synthesis, such as choline, betaine or phosphatidylcholine. Most of these metabolites were found to be increased at the beginning of the dark period. On the other hand, male animals did not show these time differences. These findings suggest that females might be more protected against the circadian disruption of the hepatic metabolism caused by a cafeteria diet through the increase of the VLDL synthesis at the beginning of the feeding time.
Collapse
|
19
|
Fontaine C, Buscato M, Vinel A, Giton F, Raymond-Letron I, Kim SH, Katzenellenbogen BS, Katzenellenbogen JA, Gourdy P, Milon A, Flouriot G, Ohlsson C, Lenfant F, Arnal JF. The tissue-specific effects of different 17β-estradiol doses reveal the key sensitizing role of AF1 domain in ERα activity. Mol Cell Endocrinol 2020; 505:110741. [PMID: 32004676 DOI: 10.1016/j.mce.2020.110741] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 01/23/2020] [Accepted: 01/25/2020] [Indexed: 12/31/2022]
Abstract
17β-Estradiol (E2) action can be mediated by the full-length estrogen receptor alpha (ERα66), and also by the AF1 domain-deficient ERα (ERα46) isoform, but their respective sensitivity to E2 is essentially unknown. We first performed a dose response study using subcutaneous home-made pellets mimicking either metestrus, proestrus or a pharmacological doses of E2, which resulted in plasma concentrations around 3, 30 and 600 pM, respectively. Analysis of the uterus, vagina and bone after chronic exposure to E2 demonstrated dose-dependent effects, with a maximal response reached at the proestrus-dose in wild type mice expressing mainly ERα66. In contrast, in transgenic mice harbouring only an ERα deleted in AF1, these effects of E2 were either strongly shifted rightward (10-100-fold) and/or attenuated, depending on the tissue studied. Finally, experiments in different cell lines transfected with ERα66 or ERα46 also delineated varying profiles of ERα AF1 sensitivity to E2. Altogether, this work emphasizes the importance of dose in the tissue-specific actions of E2 and demonstrates the key sensitizing role of AF1 in ERα activity.
Collapse
Affiliation(s)
- Coralie Fontaine
- INSERM U1048, Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse - UPS, Toulouse, France
| | - Melissa Buscato
- INSERM U1048, Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse - UPS, Toulouse, France
| | - Alexia Vinel
- INSERM U1048, Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse - UPS, Toulouse, France
| | - Frank Giton
- INSERM IMRB U955 Eq07, Créteil, France; AP-HP, Pôle Biologie-Pathologie Henri Mondor, Créteil, France
| | | | - Sung Hoon Kim
- Departments of Molecular and Integrative Biology, and of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Benita S Katzenellenbogen
- Departments of Molecular and Integrative Biology, and of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - John A Katzenellenbogen
- Departments of Molecular and Integrative Biology, and of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Pierre Gourdy
- INSERM U1048, Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse - UPS, Toulouse, France
| | - Alain Milon
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Gilles Flouriot
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR 1085, Rennes, France
| | - Claes Ohlsson
- Center for Bone and Arthritis Research, Institute of Medicine, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Françoise Lenfant
- INSERM U1048, Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse - UPS, Toulouse, France.
| | - Jean-François Arnal
- INSERM U1048, Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse - UPS, Toulouse, France
| |
Collapse
|
20
|
Farhadi Z, Khaksari M, Azizian H, Mortazaeizadeh A, Shabani M, Shahrokhi N. Beneficial effects of tamoxifen on leptin sensitivity in young mice fed a high fat diet: Role of estrogen receptor α and cytokines. Life Sci 2020; 246:117384. [PMID: 32061672 DOI: 10.1016/j.lfs.2020.117384] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 01/26/2020] [Accepted: 01/28/2020] [Indexed: 01/02/2023]
Affiliation(s)
- Zeinab Farhadi
- Department of Physiology, and Endocrinology and Metabolism Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Khaksari
- Neuroscience Research, and Physiology Research Centers, Kerman University of Medical Sciences, Kerman, Iran.
| | - Hossein Azizian
- Neurobiomedical Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Abbas Mortazaeizadeh
- Pathology and Stem Cell Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Shabani
- Neuroscience Research, and Physiology Research Centers, Kerman University of Medical Sciences, Kerman, Iran
| | - Nader Shahrokhi
- Department of Physiology, and Endocrinology and Metabolism Research Center, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
21
|
Zidon TM, Padilla J, Fritsche KL, Welly RJ, McCabe LT, Stricklin OE, Frank A, Park Y, Clegg DJ, Lubahn DB, Kanaley JA, Vieira-Potter VJ. Effects of ERβ and ERα on OVX-induced changes in adiposity and insulin resistance. J Endocrinol 2020; 245:165-178. [PMID: 32053493 PMCID: PMC7391131 DOI: 10.1530/joe-19-0321] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 02/13/2020] [Indexed: 12/12/2022]
Abstract
Loss of ovarian hormones leads to increased adiposity and insulin resistance (IR), increasing the risk for cardiovascular and metabolic diseases. The purpose of this study was to investigate whether the molecular mechanism behind the adverse systemic and adipose tissue-specific metabolic effects of ovariectomy requires loss of signaling through estrogen receptor alpha (ERα) or estrogen receptor β (ERβ). We examined ovariectomized (OVX) and ovary-intactwild-type (WT), ERα-null (αKO), and ERβ-null (βKO) female mice (age ~49 weeks; n = 7-12/group). All mice were fed a phytoestrogen-free diet (<15 mg/kg) and either remained ovary-intact (INT) or were OVX and followed for 12 weeks. Body composition, energy expenditure, glucose tolerance, and adipose tissue gene and protein expression were analyzed. INT αKO were ~25% fatter with reduced energy expenditure compared to age-matched INT WT controls and βKO mice (all P < 0.001). Following OVX, αKO mice did not increase adiposity or experience a further increase in IR, unlike WT and βKO, suggesting that loss of signaling through ERα mediates OVX-induced metabolic dysfunction. In fact, OVX in αKO mice (i.e., signaling through ERβ in the absence of ERα) resulted in reduced adiposity, adipocyte size, and IR (P < 0.05 for all). βKO mice responded adversely to OVX in terms of increased adiposity and development of IR. Together, these findings challenge the paradigm that ERα mediates metabolic protection over ERβ in all settings. These findings lead us to suggest that, following ovarian hormone loss, ERβ may mediate protective metabolic benefits.
Collapse
Affiliation(s)
- Terese M. Zidon
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia MO 65211
| | - Jaume Padilla
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia MO 65211
- Dalton Cardiovascular Research Center, University of Missouri, Columbia MO
| | - Kevin L. Fritsche
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia MO 65211
| | - Rebecca J. Welly
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia MO 65211
| | - Leighton T. McCabe
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia MO 65211
| | - Olivia E. Stricklin
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia MO 65211
| | - Aaron Frank
- Department of Biomedical Sciences, Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048
| | - Youngmin Park
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia MO 65211
- Department of Exercise and Health Science, Incheon National University, South Korea
| | - Deborah J. Clegg
- College of Nursing and Health Professions, Drexel University, Philadelphia, PA
| | | | - Jill A. Kanaley
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia MO 65211
| | | |
Collapse
|
22
|
Kim SD, Kim Y, Kim M, Jeong H, Choi SH, Ryu HW, Oh SR, Lee SW, Li WY, Wu HH, Zhu Y, Wang X, Chang M, Song YS. Estrogenic properties of Prunus cerasoides extract and its constituents in MCF-7 cell and evaluation in estrogen-deprived rodent models. Phytother Res 2020; 34:1347-1357. [PMID: 31908073 DOI: 10.1002/ptr.6604] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 11/26/2019] [Accepted: 12/04/2019] [Indexed: 12/18/2022]
Abstract
Prunus cerasoides (PC) products contain relatively high levels of flavones and isoflavones and may be potential sources of phytoestrogens for postmenopausal symptom relief. We assessed the PC extract (PCE) and its representative constituents in vitro with assays for estrogen receptor alpha binding, estrogen response element transcriptional activity, cell proliferation, and gene expression changes for pS2 in MCF-7 cells. PCE and its compounds showed strong estrogen receptor binding affinities and estrogen response element induction. A previously undescribed compound (designated as compound 18), now identified as being gentisic acid, 5-O-β-D-(6'-O-trans-4-coumaroyl)-glucopyranoside, also showed potent estrogenic properties and induced proliferation of MCF-7 cells. PCE was evaluated for its in vivo uterotrophic effects in immature female rats as well as for its lipid lowering effects in estrogen-deprived animals. For ovariectomized rats and aged female mice, PCE-treated groups had lower plasma triglyceride levels compared with control and, for the same comparison, had reduced serum levels of liver stress/damage markers. Our results point to strong estrogenic activities and beneficial metabolic effects for PCE, with properties that put PC and its extracts as promising sources of phytoestrogens for symptom relief in menopausal and postmenopausal cases.
Collapse
Affiliation(s)
- So-Dam Kim
- College of Pharmacy, Drug Information Research Institute, Sookmyung Women's University, Seoul, Republic of Korea
| | - Yeojin Kim
- College of Pharmacy, Drug Information Research Institute, Sookmyung Women's University, Seoul, Republic of Korea
| | - Mingee Kim
- College of Pharmacy, Drug Information Research Institute, Sookmyung Women's University, Seoul, Republic of Korea
| | - Hyesoo Jeong
- Department of Biological Sciences, College of Science, Sookmyung Women's University, Seoul, South Korea
| | - Sang Ho Choi
- International Biological Material Research Center, KRIBB, Daejeon, Republic of Korea
| | - Hyung Won Ryu
- Natural Medicine Research Center, KRIBB, ChungBuk, Republic of Korea
| | - Sei-Ryang Oh
- Natural Medicine Research Center, KRIBB, ChungBuk, Republic of Korea
| | - Sang Woo Lee
- International Biological Material Research Center, KRIBB, Daejeon, Republic of Korea.,Institute of Medicinal Plants, Yunnan Academy of Agricultural Sciences, Yunnan, China
| | - Wan Yi Li
- Institute of Medicinal Plants, Yunnan Academy of Agricultural Sciences, Yunnan, China
| | - Hong-Hua Wu
- College of Pharmacy, Drug Information Research Institute, Sookmyung Women's University, Seoul, Republic of Korea.,Natural Medicine Research Center, KRIBB, ChungBuk, Republic of Korea.,Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin Key Laboratory of Chemistry and Analysis of Traditional Chinese Medicine, Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yan Zhu
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin Key Laboratory of Chemistry and Analysis of Traditional Chinese Medicine, Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaoying Wang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin Key Laboratory of Chemistry and Analysis of Traditional Chinese Medicine, Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Clinical Neuroscience Reseaerch Center, Department of Neurosurgery and Neurology, Tulane, University School of Medicine, New Orleans, LA
| | - Minsun Chang
- Department of Biological Sciences, College of Science, Sookmyung Women's University, Seoul, South Korea
| | - Yun Seon Song
- College of Pharmacy, Drug Information Research Institute, Sookmyung Women's University, Seoul, Republic of Korea
| |
Collapse
|
23
|
Smith LIF, Hill TG, Bowe JE. Generating Beta-Cell-Specific Transgenic Mice Using the Cre-Lox System. Methods Mol Biol 2020; 2128:181-205. [PMID: 32180194 DOI: 10.1007/978-1-0716-0385-7_13] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Beta-cell-specific transgenic mice provide an invaluable model for dissecting the direct signaling mechanisms involved in regulating beta-cell structure and function. Furthermore, generating novel transgenic models is now easier and more cost-effective than ever, thanks to exciting novel approaches such as CRISPR.Here, we describe the commonly used approaches for generating and maintaining beta-cell-specific transgenic models and some of the considerations involved in their use. This includes the use of different beta-cell-specific promoters (e.g., pancreatic and duodenal homeobox factor 1 (Pdx1), rat insulin 2 promoter (RIP), and mouse insulin 1 promoter (MIP)) to drive site-specific recombinase technology. Important considerations during selection include level and uniformity of expression in the beta-cell population, ectopic transgene expression, and the use of inducible models.This chapter provides a guide to the procurement, generation, and maintenance of a beta-cell-specific transgene colony from preexisting Cre and loxP mouse strains, providing methods for crossbreeding and genotyping, as well as subsequent maintenance and, in the case of inducible models, transgenic induction.
Collapse
Affiliation(s)
- Lorna I F Smith
- Department of Diabetes, School of Life Course Sciences, King's College London, London, UK.
| | - Thomas G Hill
- Department of Diabetes, School of Life Course Sciences, King's College London, London, UK
| | - James E Bowe
- Department of Diabetes, School of Life Course Sciences, King's College London, London, UK
| |
Collapse
|
24
|
King SE, Nilsson E, Beck D, Skinner MK. Adipocyte epigenetic alterations and potential therapeutic targets in transgenerationally inherited lean and obese phenotypes following ancestral exposures. Adipocyte 2019; 8:362-378. [PMID: 31755359 PMCID: PMC6948971 DOI: 10.1080/21623945.2019.1693747] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 10/30/2019] [Accepted: 11/11/2019] [Indexed: 01/01/2023] Open
Abstract
The incidence of obesity has increased dramatically over the past two decades with a prevalence of approximately 40% of the adult population within the United States. The current study examines the potential for transgenerational adipocyte (fat cell) epigenetic alterations. Adipocytes were isolated from the gonadal fat pad of the great-grand offspring F3 generation 1-year old rats ancestrally exposed to DDT (dichlorodiphenyltrichloroethane), atrazine, or vehicle control in order to obtain adipocytes for DNA methylation analysis. Observations indicate that there were differential DNA methylated regions (DMRs) in the adipocytes with the lean or obese phenotypes compared to control normal (non-obese or lean) populations. The comparison of epigenetic alterations indicated that there were substantial overlaps between the different treatment lineage groups for both the lean and obese phenotypes. Novel correlated genes and gene pathways associated with DNA methylation were identified, and may aid in the discovery of potential therapeutic targets for metabolic diseases such as obesity. Observations indicate that ancestral exposures during critical windows of development can induce the epigenetic transgenerational inheritance of DNA methylation changes in adipocytes that ultimately may contribute to an altered metabolic phenotype.
Collapse
Affiliation(s)
- Stephanie E. King
- Center for Reproductive Biology, School of Biological Sciences, Washington State University, Pullman, WA, USA
| | - Eric Nilsson
- Center for Reproductive Biology, School of Biological Sciences, Washington State University, Pullman, WA, USA
| | - Daniel Beck
- Center for Reproductive Biology, School of Biological Sciences, Washington State University, Pullman, WA, USA
| | - Michael K. Skinner
- Center for Reproductive Biology, School of Biological Sciences, Washington State University, Pullman, WA, USA
| |
Collapse
|
25
|
Guillaume M, Riant E, Fabre A, Raymond-Letron I, Buscato M, Davezac M, Tramunt B, Montagner A, Smati S, Zahreddine R, Palierne G, Valera MC, Guillou H, Lenfant F, Unsicker K, Metivier R, Fontaine C, Arnal JF, Gourdy P. Selective Liver Estrogen Receptor α Modulation Prevents Steatosis, Diabetes, and Obesity Through the Anorectic Growth Differentiation Factor 15 Hepatokine in Mice. Hepatol Commun 2019; 3:908-924. [PMID: 31304450 PMCID: PMC6601326 DOI: 10.1002/hep4.1363] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 03/28/2019] [Indexed: 12/17/2022] Open
Abstract
Hepatocyte estrogen receptor α (ERα) was recently recognized as a relevant molecular target for nonalcoholic fatty liver disease (NAFLD) prevention. The present study defined to what extent hepatocyte ERα could be involved in preserving metabolic homeostasis in response to a full (17β-estradiol [E2]) or selective (selective estrogen receptor modulator [SERM]) activation. Ovariectomized mice harboring a hepatocyte-specific ERα deletion (LERKO mice) and their wild-type (WT) littermates were fed a high-fat diet (HFD) and concomitantly treated with E2, tamoxifen (TAM; the most used SERM), or vehicle. As expected, both E2 and TAM prevented all HFD-induced metabolic disorders in WT mice, and their protective effects against steatosis were abolished in LERKO mice. However, while E2 still prevented obesity and glucose intolerance in LERKO mice, hepatocyte ERα deletion also abrogated TAM-mediated control of food intake as well as its beneficial actions on adiposity, insulin sensitivity, and glucose homeostasis, suggesting a whole-body protective role for liver-derived circulating factors. Moreover, unlike E2, TAM induced a rise in plasma concentration of the anorectic hepatokine growth differentiation factor 15 (Gdf15) through a transcriptional mechanism dependent on hepatocyte ERα activation. Accordingly, ERα was associated with specific binding sites in the Gdf15 regulatory region in hepatocytes from TAM-treated mice but not under E2 treatment due to specific epigenetic modifications. Finally, all the protective effects of TAM were abolished in HFD-fed GDF15-knockout mice. Conclusion: We identified the selective modulation of hepatocyte ERα as a pharmacologic strategy to induce sufficient anorectic hepatokine Gdf15 to prevent experimental obesity, type 2 diabetes, and NAFLD.
Collapse
Affiliation(s)
- Maeva Guillaume
- Institut des Maladies Métaboliques et Cardiovasculaires Unité Médicale de Recherche 1048, Institut National de la Santé et de le Recherche Médicale (INSERM), Université de Toulouse III Toulouse France.,Service d'Hépato-gastro-entérologie Centre Hospitalier Universitaire de Toulouse Toulouse France
| | - Elodie Riant
- Institut des Maladies Métaboliques et Cardiovasculaires Unité Médicale de Recherche 1048, Institut National de la Santé et de le Recherche Médicale (INSERM), Université de Toulouse III Toulouse France
| | - Aurélie Fabre
- Institut des Maladies Métaboliques et Cardiovasculaires Unité Médicale de Recherche 1048, Institut National de la Santé et de le Recherche Médicale (INSERM), Université de Toulouse III Toulouse France
| | - Isabelle Raymond-Letron
- STROMALab, Centre National de la Recherche Scientifique ERL5311 Etablissement Français du Sang, Ecole Nationale Vétérinaire de Toulouse, Institut National de la Santé et de le Recherche Médicale (INSERM) U1031, Université de Toulouse III Toulouse France
| | - Melissa Buscato
- Institut des Maladies Métaboliques et Cardiovasculaires Unité Médicale de Recherche 1048, Institut National de la Santé et de le Recherche Médicale (INSERM), Université de Toulouse III Toulouse France
| | - Morgane Davezac
- Institut des Maladies Métaboliques et Cardiovasculaires Unité Médicale de Recherche 1048, Institut National de la Santé et de le Recherche Médicale (INSERM), Université de Toulouse III Toulouse France
| | - Blandine Tramunt
- Institut des Maladies Métaboliques et Cardiovasculaires Unité Médicale de Recherche 1048, Institut National de la Santé et de le Recherche Médicale (INSERM), Université de Toulouse III Toulouse France
| | - Alexandra Montagner
- Institut des Maladies Métaboliques et Cardiovasculaires Unité Médicale de Recherche 1048, Institut National de la Santé et de le Recherche Médicale (INSERM), Université de Toulouse III Toulouse France
| | - Sarra Smati
- Institut des Maladies Métaboliques et Cardiovasculaires Unité Médicale de Recherche 1048, Institut National de la Santé et de le Recherche Médicale (INSERM), Université de Toulouse III Toulouse France.,Institut National de La Recherche Agronomique Unité Médicale de Recherche 1331, ToxAlim, Université de Toulouse Toulouse France
| | - Rana Zahreddine
- Institut des Maladies Métaboliques et Cardiovasculaires Unité Médicale de Recherche 1048, Institut National de la Santé et de le Recherche Médicale (INSERM), Université de Toulouse III Toulouse France
| | - Gaëlle Palierne
- Equipe SP@RTE, Unité Médicale de Recherche 6290, Institut de Genétique et Développement de Rennes Université de Rennes 1 Rennes France
| | - Marie-Cécile Valera
- Institut des Maladies Métaboliques et Cardiovasculaires Unité Médicale de Recherche 1048, Institut National de la Santé et de le Recherche Médicale (INSERM), Université de Toulouse III Toulouse France
| | - Hervé Guillou
- Institut National de La Recherche Agronomique Unité Médicale de Recherche 1331, ToxAlim, Université de Toulouse Toulouse France
| | - Françoise Lenfant
- Institut des Maladies Métaboliques et Cardiovasculaires Unité Médicale de Recherche 1048, Institut National de la Santé et de le Recherche Médicale (INSERM), Université de Toulouse III Toulouse France
| | - Klaus Unsicker
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology University of Freiburg Freiburg Germany
| | - Raphaël Metivier
- Equipe SP@RTE, Unité Médicale de Recherche 6290, Institut de Genétique et Développement de Rennes Université de Rennes 1 Rennes France
| | - Coralie Fontaine
- Institut des Maladies Métaboliques et Cardiovasculaires Unité Médicale de Recherche 1048, Institut National de la Santé et de le Recherche Médicale (INSERM), Université de Toulouse III Toulouse France
| | - Jean-François Arnal
- Institut des Maladies Métaboliques et Cardiovasculaires Unité Médicale de Recherche 1048, Institut National de la Santé et de le Recherche Médicale (INSERM), Université de Toulouse III Toulouse France
| | - Pierre Gourdy
- Institut des Maladies Métaboliques et Cardiovasculaires Unité Médicale de Recherche 1048, Institut National de la Santé et de le Recherche Médicale (INSERM), Université de Toulouse III Toulouse France.,Service de Diabétologie Maladies Métaboliques et Nutrition, Centre Hospitalier Universitaire de Toulouse Toulouse France
| |
Collapse
|
26
|
Ceasrine AM, Ruiz-Otero N, Lin EE, Lumelsky DN, Boehm ED, Kuruvilla R. Tamoxifen Improves Glucose Tolerance in a Delivery-, Sex-, and Strain-Dependent Manner in Mice. Endocrinology 2019; 160:782-790. [PMID: 30759201 PMCID: PMC6424092 DOI: 10.1210/en.2018-00985] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 02/07/2019] [Indexed: 12/19/2022]
Abstract
Tamoxifen, a selective estrogen-receptor modulator, is widely used in mouse models to temporally control gene expression but is also known to affect body composition. We report that tamoxifen has significant and sustained effects on glucose tolerance, independent of effects on insulin sensitivity, in mice. IP, but not oral, tamoxifen delivery improved glucose tolerance in three inbred mouse strains. The extent and persistence of tamoxifen-induced effects were sex and strain dependent. These findings highlight the need to revise commonly used tamoxifen-based protocols for gene manipulation in mice by including longer chase periods after injection, oral delivery, and the use of tamoxifen-treated littermate controls.
Collapse
Affiliation(s)
- Alexis M Ceasrine
- Department of Biology, Johns Hopkins University, Baltimore, Maryland
- Correspondence: Alexis M. Ceasrine, PhD, Johns Hopkins University, 3400 N. Charles Street, Mudd Hall 200, Baltimore, Maryland 21218. E-mail:
| | | | - Eugene E Lin
- Department of Biology, Johns Hopkins University, Baltimore, Maryland
| | - David N Lumelsky
- Department of Biology, Johns Hopkins University, Baltimore, Maryland
| | - Erica D Boehm
- Department of Biology, Johns Hopkins University, Baltimore, Maryland
| | - Rejji Kuruvilla
- Department of Biology, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
27
|
Woo ARE, Sze SK, Chung HH, Lin VCL. Delineation of critical amino acids in activation function 1 of progesterone receptor for recruitment of transcription coregulators. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2019; 1862:522-533. [DOI: 10.1016/j.bbagrm.2019.01.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 01/15/2019] [Accepted: 01/30/2019] [Indexed: 12/17/2022]
|
28
|
Mishra S, Nyomba BLG. Prohibitin: A hypothetical target for sex-based new therapeutics for metabolic and immune diseases. Exp Biol Med (Maywood) 2019; 244:157-170. [PMID: 30717609 PMCID: PMC6405819 DOI: 10.1177/1535370219828362] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
IMPACT STATEMENT Traditional sex-related biases in research are now obsolete, and it is important to identify the sex of humans, animals, and even cells in research protocols, due to the role of sex as a fundamental facet of biology, predisposition to disease, and response to therapy. Genetic sex, epigenetics and hormonal regulations, generate sex-dimorphisms. Recent investigations acknowledge sex differences in metabolic and immune health as well as chronic diseases. Prohibitin, an evolutionarily conserved molecule, has pleotropic functions in mitochondrial housekeeping, plasma membrane signaling, and nuclear genetic transcription. Studies in adipocytes, macrophages, and transgenic mice indicate that prohibitin interacts with sex steroids and plays a role in mediating sex differences in adipose tissues and immune cell types. Prohibitin may, depending on context, modulate predisposition to chronic metabolic diseases and malignancy and, because of these attributes, could be a target for sex-based therapies of metabolic and immune-related diseases as well as cancer.
Collapse
Affiliation(s)
- Suresh Mishra
- Department of Internal Medicine, University of Manitoba,
Manitoba R3A1R9, Canada
- Department of Physiology & Pathophysiology, University of
Manitoba, Manitoba R3E0J9, Canada
| | - BL Grégoire Nyomba
- Department of Internal Medicine, University of Manitoba,
Manitoba R3A1R9, Canada
| |
Collapse
|
29
|
Valéra MC, Fontaine C, Noirrit-Esclassan E, Boudou F, Buscato M, Adlanmerini M, Trémollières F, Gourdy P, Lenfant F, Arnal JF. [Towards an optimization of the modulation of the estrogen receptor during menopausal hormonal therapy]. Med Sci (Paris) 2019; 34:1056-1062. [PMID: 30623764 DOI: 10.1051/medsci/2018297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Women now live more than a third of their lives after the onset of menopause. The decline in endogenous estrogen production during this period is accompanied by functional disorders that affect quality of life. These symptoms may be relieved by menopausal hormone therapy (MHT) initially based on the administration of equine conjugated estrogens (mainly in the United States, oral route) or the natural estrogen, 17β-estradiol (in Europe, transdermal route). Estrogen receptor α (ERα), but not ERβ, mediates most of the physiological effects of estrogens. ERα belongs to the superfamily of nuclear receptors and regulates the transcription of genes via its activation functions AF1 and AF2. In addition to these classical genomic actions, estrogens can activate a subpopulation of ERα present at the cell membrane and thereby induce rapid signals. In this review, we will summarize the evolution of MHTs in last decades, as well as treatments that use various selective estrogen receptor modulators (SERMs). Next, we will describe recent advances in the understanding of the mechanisms of estrogen action, in particular the respective roles of nuclear and membrane ERα as well as the potential implications for future therapies.
Collapse
Affiliation(s)
- Marie-Cécile Valéra
- Inserm U1048 et université Toulouse III, I2MC, CHU Rangueil, BP 84225, 31432 Toulouse Cedex 4, France
| | - Coralie Fontaine
- Inserm U1048 et université Toulouse III, I2MC, CHU Rangueil, BP 84225, 31432 Toulouse Cedex 4, France
| | | | - Frédéric Boudou
- Inserm U1048 et université Toulouse III, I2MC, CHU Rangueil, BP 84225, 31432 Toulouse Cedex 4, France
| | - Melissa Buscato
- Inserm U1048 et université Toulouse III, I2MC, CHU Rangueil, BP 84225, 31432 Toulouse Cedex 4, France
| | - Marine Adlanmerini
- Inserm U1048 et université Toulouse III, I2MC, CHU Rangueil, BP 84225, 31432 Toulouse Cedex 4, France
| | - Florence Trémollières
- Inserm U1048 et université Toulouse III, I2MC, CHU Rangueil, BP 84225, 31432 Toulouse Cedex 4, France
| | - Pierre Gourdy
- Inserm U1048 et université Toulouse III, I2MC, CHU Rangueil, BP 84225, 31432 Toulouse Cedex 4, France
| | - Françoise Lenfant
- Inserm U1048 et université Toulouse III, I2MC, CHU Rangueil, BP 84225, 31432 Toulouse Cedex 4, France
| | - Jean-François Arnal
- Inserm U1048 et université Toulouse III, I2MC, CHU Rangueil, BP 84225, 31432 Toulouse Cedex 4, France
| |
Collapse
|
30
|
Wu S, Xue P, Grayson N, Bland JS, Wolfe A. Bitter Taste Receptor Ligand Improves Metabolic and Reproductive Functions in a Murine Model of PCOS. Endocrinology 2019; 160:143-155. [PMID: 30418546 DOI: 10.1210/en.2018-00711] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 11/05/2018] [Indexed: 12/13/2022]
Abstract
Polycystic ovary syndrome (PCOS) results from functional ovarian hyperandrogenism due to dysregulation of androgen secretion. Cultured theca cells from polycystic ovaries of women with the most common form of PCOS overexpress most androgen producing enzymes, particularly CYP450c17. In this study, a murine model was used of PCOS induced by chronic feeding with a high-fat diet that exhibits the reproductive, hyperandrogenic, and metabolic constellation of PCOS symptoms seen in women. Oral administration of KDT501, a hops-derived bitter taste receptor (Tas2R 108) isohumulone ligand resulted in resolution of PCOS-associated endocrine and metabolic disturbances and restored reproductive function. Pioglitazone, a PPARγ agonist, also improved metabolic and reproductive function, though not to the same degree as KDT501. Specifically, treatment of the murine PCOS model with KDT501 resulted in reduced testosterone and androstenedione levels in the absence of significant changes in LH or FSH, improved glucose tolerance and lipid metabolism, and reduced hepatic lipid infiltration and adiposity. There was significant improvement in estrous cyclicity and an increase in the number of ovarian corpora lutea, indicative of improved reproductive function after exposure to KDT501. Finally, ex vivo exposure of murine ovaries to KDT501 attenuated androgen production and ovarian expression of CYP450c17. Interestingly, the ovaries expressed Tas2R 108, suggesting a potential regulation of ovarian steroidogenesis through this chemosensory receptor family. In summary, a therapeutic strategy for PCOS possibly could include direct influences on ovarian steroidogenesis that are independent of gonadotrophic hormone regulation.
Collapse
Affiliation(s)
- Sheng Wu
- Division of Endocrinology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ping Xue
- Division of Endocrinology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Neile Grayson
- Kindex Pharmaceutical, Bainbridge Island, Washington
| | | | - Andrew Wolfe
- Division of Endocrinology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
31
|
Arao Y, Hamilton KJ, Lierz SL, Korach KS. N-terminal transactivation function, AF-1, of estrogen receptor alpha controls obesity through enhancement of energy expenditure. Mol Metab 2018; 18:68-78. [PMID: 30287090 PMCID: PMC6308972 DOI: 10.1016/j.molmet.2018.09.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 09/03/2018] [Accepted: 09/17/2018] [Indexed: 01/28/2023] Open
Abstract
Objective Studies using the estrogen receptor alpha (ERα) knock-out (αERKO) mice have demonstrated that ERα plays a crucial role in various estrogen-mediated metabolic regulations. ERα is a ligand dependent transcription regulator and its activity is regulated by estrogenic compounds. ERα consists of two transcriptional activation domains, AF-1 and AF-2. The activities of these domains are regulated through different mechanisms; however, the specific physiological role in metabolic regulation by these domains is still unclear. Methods We utilized an ERα AF-2 mutant knock-in mouse (AF2ERKI) to evaluate the physiological functionality of ERα transactivation domains. Due to the estrogen insensitive AF-2 mutation, the phenotypes of AF2ERKI mice are seemingly identical to the global αERKO including obesity in the females. Distinct from the αERKO, the AF-1 function of AF2ERKI mice can be activated by tamoxifen (Tam). Ovariectomized (OVX) AF2ERKI and WT females were treated with Tam and fed a high-fat diet (HFD) for 10 weeks. Additionally, indirect calorimetric analysis was performed using metabolic chambers with food intake and locomotor activity recorded for Tam-treated AF2ERKI and αERKO females. Results Obesity in HFD-fed AF2ERKI females was prevented by Tam treatment; particularly, inguinal fat accumulation was strongly blocked by Tam treatment. Alterations in fat metabolism genes, however, were not found in either inguinal fat nor visceral fat to be Tam-regulated, even though fat accumulation was strongly reduced by Tam treatment. Indirect calorimetric analysis revealed that without alteration of food intake and locomotor activity Tam treatment increased energy expenditure in AF2ERKI but not αERKO females. Conclusions These results suggest that the activation of ERα AF-1 prevents fat accumulation. The prevention of obesity through AF-1 is mediated by induction of energy expenditure rather than ERα AF-1 functionality of lipid metabolism gene regulation in fat tissues. AF-2 disrupted ERα mutant (AF2ERKI) females are obese with a pre-diabetic condition. Activation of ERα AF-1 prevented AF2ERKI obesity. Inguinal fat accumulation altered more than visceral fat in AF2ERKI females. AF-1 activation improved energy expenditure without changing activity and feeding.
Collapse
Affiliation(s)
- Yukitomo Arao
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences/NIH, Research Triangle Park, NC, 27709, USA.
| | - Katherine J Hamilton
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences/NIH, Research Triangle Park, NC, 27709, USA
| | - Sydney L Lierz
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences/NIH, Research Triangle Park, NC, 27709, USA
| | - Kenneth S Korach
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences/NIH, Research Triangle Park, NC, 27709, USA.
| |
Collapse
|
32
|
Gourdy P, Guillaume M, Fontaine C, Adlanmerini M, Montagner A, Laurell H, Lenfant F, Arnal JF. Estrogen receptor subcellular localization and cardiometabolism. Mol Metab 2018; 15:56-69. [PMID: 29807870 PMCID: PMC6066739 DOI: 10.1016/j.molmet.2018.05.009] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 05/09/2018] [Accepted: 05/10/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND In addition to their crucial role in reproduction, estrogens are key regulators of energy and glucose homeostasis and they also exert several cardiovascular protective effects. These beneficial actions are mainly mediated by estrogen receptor alpha (ERα), which is widely expressed in metabolic and vascular tissues. As a member of the nuclear receptor superfamily, ERα was primarily considered as a transcription factor that controls gene expression through the activation of its two activation functions (ERαAF-1 and ERαAF-2). However, besides these nuclear actions, a pool of ERα is localized in the vicinity of the plasma membrane, where it mediates rapid signaling effects called membrane-initiated steroid signals (MISS) that have been well described in vitro, especially in endothelial cells. SCOPE OF THE REVIEW This review aims to summarize our current knowledge of the mechanisms of nuclear vs membrane ERα activation that contribute to the cardiometabolic protection conferred by estrogens. Indeed, new transgenic mouse models (affecting either DNA binding, activation functions or membrane localization), together with the use of novel pharmacological tools that electively activate membrane ERα effects recently allowed to begin to unravel the different modes of ERα signaling in vivo. CONCLUSION Altogether, available data demonstrate the prominent role of ERα nuclear effects, and, more specifically, of ERαAF-2, in the preventive effects of estrogens against obesity, diabetes, and atheroma. However, membrane ERα signaling selectively mediates some of the estrogen endothelial/vascular effects (NO release, reendothelialization) and could also contribute to the regulation of energy balance, insulin sensitivity, and glucose metabolism. Such a dissection of ERα biological functions related to its subcellular localization will help to understand the mechanism of action of "old" ER modulators and to design new ones with an optimized benefit/risk profile.
Collapse
Affiliation(s)
- Pierre Gourdy
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1048/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France; Service de Diabétologie, Maladies Métaboliques et Nutrition, CHU de Toulouse, Toulouse, France.
| | - Maeva Guillaume
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1048/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France; Service d'Hépatologie et Gastro-Entérologie, CHU de Toulouse, Toulouse, France
| | - Coralie Fontaine
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1048/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France
| | - Marine Adlanmerini
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1048/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France
| | - Alexandra Montagner
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1048/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France
| | - Henrik Laurell
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1048/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France
| | - Françoise Lenfant
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1048/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France
| | - Jean-François Arnal
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1048/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France
| |
Collapse
|
33
|
Valéra MC, Fontaine C, Dupuis M, Noirrit-Esclassan E, Vinel A, Guillaume M, Gourdy P, Lenfant F, Arnal JF. Towards optimization of estrogen receptor modulation in medicine. Pharmacol Ther 2018; 189:123-129. [PMID: 29730442 DOI: 10.1016/j.pharmthera.2018.05.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Women now spend more than one-third of their lives in the postmenopausal years, and the decline of endogenous estrogen production during menopause is accompanied by a series of functional disorders that affect the quality of life. These symptoms could be alleviated or even totally suppressed by menopausal hormone therapy (MHT), initially based on natural estrogens extracted from the urine of pregnant mares (mainly in the USA, using the oral route) and later from the synthesis of the natural estrogen, 17β-estradiol (mainly in Europe, in particular using the transdermal route). Estrogen receptor (ER) α is the main receptor mediating the physiological effects of estrogens. ERα belongs to the nuclear receptor superfamily and activates gene transcription in a time and tissue-specific manner through two distinct activation functions (AF), AF1 and AF2. In addition to these classical genomic actions, ERα also mediates membrane initiated signaling enabling rapid actions of estrogen, potentially along or in interaction with other receptors. Here, we provide a brief historical overview of MHT, and we then highlight recent advances in the characterization of new treatments based on the association of estrogens with selective estrogen receptor modulators (SERMs) or on the modulation of nuclear or membrane ERα.
Collapse
Affiliation(s)
- Marie-Cécile Valéra
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048 and Université Toulouse III, I2MC, Toulouse, France
| | - Coralie Fontaine
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048 and Université Toulouse III, I2MC, Toulouse, France
| | - Marion Dupuis
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048 and Université Toulouse III, I2MC, Toulouse, France
| | - Emmanuelle Noirrit-Esclassan
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048 and Université Toulouse III, I2MC, Toulouse, France
| | - Alexia Vinel
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048 and Université Toulouse III, I2MC, Toulouse, France
| | - Maeva Guillaume
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048 and Université Toulouse III, I2MC, Toulouse, France
| | - Pierre Gourdy
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048 and Université Toulouse III, I2MC, Toulouse, France
| | - Françoise Lenfant
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048 and Université Toulouse III, I2MC, Toulouse, France
| | - Jean-François Arnal
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048 and Université Toulouse III, I2MC, Toulouse, France.
| |
Collapse
|
34
|
Passeri GI, Trisciuzzi D, Alberga D, Siragusa L, Leonetti F, Mangiatordi GF, Nicolotti O. Strategies of Virtual Screening in Medicinal Chemistry. ACTA ACUST UNITED AC 2018. [DOI: 10.4018/ijqspr.2018010108] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Virtual screening represents an effective computational strategy to rise-up the chances of finding new bioactive compounds by accelerating the time needed to move from an initial intuition to market. Classically, the most pursued approaches rely on ligand- and structure-based studies, the former employed when structural data information about the target is missing while the latter employed when X-ray/NMR solved or homology models are instead available for the target. The authors will focus on the most advanced techniques applied in this area. In particular, they will survey the key concepts of virtual screening by discussing how to properly select chemical libraries, how to make database curation, how to applying and- and structure-based techniques, how to wisely use post-processing methods. Emphasis will be also given to the most meaningful databases used in VS protocols. For the ease of discussion several examples will be presented.
Collapse
Affiliation(s)
| | - Daniela Trisciuzzi
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari “Aldo Moro”, Bari, Italy
| | - Domenico Alberga
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari “Aldo Moro”, Bari, Italy
| | - Lydia Siragusa
- Molecular Discovery Ltd., Pinner, Middlesex, London, United Kingdom
| | - Francesco Leonetti
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari “Aldo Moro”, Bari, Italy
| | - Giuseppe F. Mangiatordi
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari “Aldo Moro”, Bari, Italy
| | | |
Collapse
|
35
|
Nuclear and Membrane Actions of Estrogen Receptor Alpha: Contribution to the Regulation of Energy and Glucose Homeostasis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1043:401-426. [PMID: 29224105 DOI: 10.1007/978-3-319-70178-3_19] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Estrogen receptor alpha (ERα) has been demonstrated to play a key role in reproduction but also to exert numerous functions in nonreproductive tissues. Accordingly, ERα is now recognized as a key regulator of energy homeostasis and glucose metabolism and mediates the protective effects of estrogens against obesity and type 2 diabetes. This chapter attempts to summarize our current understanding of the mechanisms of ERα activation and their involvement in the modulation of energy balance and glucose metabolism. We first focus on the experimental studies that constitute the basis of the understanding of ERα as a nuclear receptor and more specifically on the key roles played by its two activation functions (AFs). We depict the consequences of the selective inactivation of these AFs in mouse models, which further underline the prominent role of nuclear ERα in the prevention of obesity and diabetes, as on the reproductive tract and the vascular system. Besides these nuclear actions, a fraction of ERα is associated with the plasma membrane and activates nonnuclear signaling from this site. Such rapid effects, called membrane-initiated steroid signals (MISS), have been characterized in a variety of cell lines and in particular in endothelial cells. The development of selective pharmacological tools that specifically activate MISS as well as the generation of mice expressing an ERα protein impeded for membrane localization has just begun to unravel the physiological role of MISS in vivo and their contribution to ERα-mediated metabolic protection. Finally, we discuss novel perspectives for the design of tissue-selective ER modulators.
Collapse
|