1
|
Kim SO, Albrecht ED, Pepe GJ. Estrogen promotes fetal skeletal muscle mitochondrial distribution and ATP synthase activity important for insulin sensitivity in offspring. Endocrine 2024; 85:417-427. [PMID: 38478198 PMCID: PMC11246263 DOI: 10.1007/s12020-024-03764-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 02/25/2024] [Indexed: 07/14/2024]
Abstract
PURPOSE We previously showed that offspring delivered to baboons in which levels of estradiol (E2) were suppressed during the second half of gestation exhibit insulin resistance. Mitochondria are essential for the production of ATP as the main source of energy for intracellular metabolic pathways, and skeletal muscle of type 2 diabetics exhibit mitochondrial abnormalities. Mitochondria express estrogen receptor β and E2 enhances mitochondrial function in adults. Therefore, the current study ascertained whether exposure of the fetus to E2 is essential for mitochondrial development. METHODS Levels of ATP synthase and citrate synthase and the morphology of mitochondria were determined in fetal skeletal muscle obtained near term from baboons untreated or treated daily with the aromatase inhibitor letrozole or letrozole plus E2. RESULTS Specific activity and amount of ATP synthase were 2-fold lower (P < 0.05) in mitochondria from skeletal muscle of E2 suppressed letrozole-treated fetuses and restored to normal by treatment with letrozole plus E2. Immunocytochemistry showed that in contrast to the punctate formation of mitochondria in myocytes of untreated and letrozole plus E2 treated animals, mitochondria appeared to be diffuse in myocytes of estrogen-suppressed fetuses. However, citrate synthase activity and levels of proteins that control mitochondrial fission/fusion were similar in estrogen replete and suppressed animals. CONCLUSION We suggest that estrogen is essential for fetal skeletal muscle mitochondrial development and thus glucose homeostasis in adulthood.
Collapse
Affiliation(s)
- Soon Ok Kim
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Eugene D Albrecht
- Departments of Obstetrics/Gynecology/Reproductive Sciences and Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Gerald J Pepe
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA, USA.
| |
Collapse
|
2
|
Lin P, Zhang X, Zhu B, Gao J, Yin D, Zeng J, Kang Z. Naringenin protects pancreatic β cells in diabetic rat through activation of estrogen receptor β. Eur J Pharmacol 2023; 960:176115. [PMID: 37866740 DOI: 10.1016/j.ejphar.2023.176115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 09/27/2023] [Accepted: 10/12/2023] [Indexed: 10/24/2023]
Abstract
Naringenin is a citrus flavonoid that potently improves metabolic parameters in animal models of metabolic disorders, such as type 2 diabetes. Estrogen receptor (ER) activation promotes β cell function and survival, thereby improving systemic glucose metabolism. In this study, we used a luciferase reporter assay, isolated rat islets and a diabetic rat model to investigate the effects of naringenin on ER signaling and the underlying mechanism of naringenin-mediated improvement of islet function in diabetes. Naringenin specifically activated ERβ without affecting the activity of ERα, G protein-coupled estrogen receptor (GPER) or estrogen-related receptor (ERR) α/β/γ. Additionally, treatment with naringenin enhanced glucose-stimulated insulin secretion in isolated rat islets. This effect was abrogated by PHTPP, an ERβ antagonist. Transcriptomic analysis revealed that naringenin upregulated the expression of genes, such as Pdx1 and Mafa, which are closely linked to improved β-cell function. In consistence, single administration of naringenin to normal rats elevated plasma insulin levels and improved glucose responses. These beneficial effects were blocked by PHTPP. In streptozocin-nicotinamide induced diabetic rats, treatment for 2 weeks with naringenin alone, but not in combination with PHTPP, significantly restored pancreatic β cell mass and improved glucose metabolism. Collectively, these data support that naringenin specifically activate ERβ to improve insulin secretion in the primary rat islets. Furthermore, naringenin administration also protected β cell function and reversed glucose dysregulation in diabetic rats. These beneficial effects are at least partially dependent on the ERβ pathway.
Collapse
Affiliation(s)
- Peibin Lin
- Department of Basic Medical Research, Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Xiaojing Zhang
- Department of Pharmacy, Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Baoyi Zhu
- Department of Urology, Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, Guangdong, China; Guangdong Engineering Research Center of Urinary Continence and Reproductive Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Jun Gao
- Department of Basic Medical Research, Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Dazhong Yin
- Department of Basic Medical Research, Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Jianwen Zeng
- Department of Urology, Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, Guangdong, China; Guangdong Engineering Research Center of Urinary Continence and Reproductive Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, Guangdong, China.
| | - Zhanfang Kang
- Department of Basic Medical Research, Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, Guangdong, China; Guangdong Engineering Research Center of Urinary Continence and Reproductive Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, Guangdong, China.
| |
Collapse
|
3
|
Inada A, Yasunami Y, Yoshiki A, Nabeshima YI, Inada O. Greb1 Transiently Accelerates Pancreatic β-Cell Proliferation in Diabetic Mice Exposed to Estradiol. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:1081-1100. [PMID: 37516458 DOI: 10.1016/j.ajpath.2023.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/14/2023] [Accepted: 04/04/2023] [Indexed: 07/31/2023]
Abstract
Decrease of pancreatic β cells leads to diabetes. In an inducible cAMP early suppressor (ICER-Iγ) transgenic mouse model of severe type 2 diabetes with reduced insulin production and depleted β cells, supplementation with high concentrations of 17β-estradiol (E2) markedly enhances β-cell proliferation and normalizes glucose levels. The current study explored the underlying mechanisms leading to a dynamic increase of β cells and pathologic changes in diabetic mice exposed to E2. Gene expression profiling of pancreatic islets of 6-month-old ICER-transgenic mice recovering from diabetes due to elevated E2 levels identified growth regulation by estrogen in breast cancer 1 (Greb1) as a gene significantly up-regulated during the recovery phase. To substantiate this, β-cell-specific Greb1-deficient mice were generated, and Greb1 was shown to be essential for recovery of depleted β cells in diabetic mice. Graft growth and glucose lowering were observed in 50 islets with increased Greb1 expression transplanted adjacent to E2 pellets beneath the kidney capsule of streptozotocin-induced diabetic mice. Greb1 expression due to a drastic increase in exogenous or endogenous E2 was transient and closely correlated with changes in E2-related and some cell cycle-related genes. These findings provide new insights into in vivo proliferation of deficient β cells and suggest the possibility of new therapeutic approaches targeting pancreatic β cells that could revolutionize the concept of diabetes treatment, which has been considered difficult to cure completely.
Collapse
Affiliation(s)
- Akari Inada
- Clinical Research Department, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan; Laboratory of Molecular Life Science, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan; Diabetes and Genes, Advanced Medical Initiatives, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | | | - Atsushi Yoshiki
- Experimental Animal Division, RIKEN BioResource Research Center, Tsukuba, Japan
| | - Yo-Ichi Nabeshima
- Laboratory of Molecular Life Science, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
| | - Oogi Inada
- Diabetes and Genes, Advanced Medical Initiatives, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
4
|
Abdel-Karim TR, Hodges JS, Pruett TL, Ramanathan KV, Hering BJ, Dunn TB, Kirchner VA, Beilman GJ, Bellin MD. A randomized controlled pilot trial of etanercept and alpha-1 antitrypsin to improve autologous islet engraftment. Pancreatology 2023; 23:57-64. [PMID: 36443174 PMCID: PMC9839597 DOI: 10.1016/j.pan.2022.11.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/17/2022] [Accepted: 11/12/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND In total pancreatectomy with islet auto-transplantation, successful diabetes outcomes are limited by islet loss from the instant blood mediated inflammatory response. We hypothesized that blockade of the inflammatory response with either etanercept or alpha-1-antitrypsin would improve islet function and insulin independence. METHODS We randomized 43 participants to receive A1AT (90 mg/kg x 6 doses, n = 13), or etanercept (50 mg then 25 mg x 5 doses, n = 14), or standard care (n = 16), aiming to reduce detrimental effects of innate inflammation on early islet survival. Islet graft function was assessed using mixed meal tolerance testing, intravenous glucose tolerance testing, glucose-potentiated arginine-induced insulin secretion studies, HbA1c, and insulin dose 3 months and 1 year post-TPIAT. RESULTS We observed the most robust acute insulin response (AIRglu) and acute C-peptide response to glucose (ACRglu) at 3 months after TPIAT in the etanercept-treated group (p ≤ 0.02), but no differences in other efficacy measures. The groups did not differ overall at 1 year but when adjusted by sex, there was a trend towards a sex-specific treatment effect in females (AIRglu p = 0.05, ACRglu p = 0.06), with insulin secretion measures highest in A1AT-treated females. CONCLUSION Our randomized trial supports a potential role for etanercept in optimizing early islet engraftment but it is unclear whether this benefit is sustained. Further studies are needed to evaluate possible sex-specific responses to either treatment. CLINICAL TRIAL NOTATION This study was performed under an Investigational New Drug Application (IND #119828) from the Food and Drug Administration and was registered on clinicaltrials.gov (NCT#02713997).
Collapse
Affiliation(s)
| | - James S Hodges
- Department of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Timothy L Pruett
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA
| | | | - Bernhard J Hering
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA
| | - Ty B Dunn
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA; Department of Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Varvara A Kirchner
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA; Department of Surgery, Stanford University, Palo Alto, CA, USA
| | - Gregory J Beilman
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA
| | - Melena D Bellin
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA; Department of Surgery, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
5
|
Muhammad A, Forcados GE, Yusuf AP, Abubakar MB, Sadiq IZ, Elhussin I, Siddique MAT, Aminu S, Suleiman RB, Abubakar YS, Katsayal BS, Yates CC, Mahavadi S. Comparative G-Protein-Coupled Estrogen Receptor (GPER) Systems in Diabetic and Cancer Conditions: A Review. Molecules 2022; 27:molecules27248943. [PMID: 36558071 PMCID: PMC9786783 DOI: 10.3390/molecules27248943] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/05/2022] [Accepted: 12/08/2022] [Indexed: 12/23/2022] Open
Abstract
For many patients, diabetes Mellitus and Malignancy are frequently encountered comorbidities. Diabetes affects approximately 10.5% of the global population, while malignancy accounts for 29.4 million cases each year. These troubling statistics indicate that current treatment approaches for these diseases are insufficient. Alternative therapeutic strategies that consider unique signaling pathways in diabetic and malignancy patients could provide improved therapeutic outcomes. The G-protein-coupled estrogen receptor (GPER) is receiving attention for its role in disease pathogenesis and treatment outcomes. This review aims to critically examine GPER' s comparative role in diabetes mellitus and malignancy, identify research gaps that need to be filled, and highlight GPER's potential as a therapeutic target for diabetes and malignancy management. There is a scarcity of data on GPER expression patterns in diabetic models; however, for diabetes mellitus, altered expression of transport and signaling proteins has been linked to GPER signaling. In contrast, GPER expression in various malignancy types appears to be complex and debatable at the moment. Current data show inconclusive patterns of GPER expression in various malignancies, with some indicating upregulation and others demonstrating downregulation. Further research should be conducted to investigate GPER expression patterns and their relationship with signaling pathways in diabetes mellitus and various malignancies. We conclude that GPER has therapeutic potential for chronic diseases such as diabetes mellitus and malignancy.
Collapse
Affiliation(s)
- Aliyu Muhammad
- Center for Cancer Research, Department of Biology, Tuskegee University, Tuskegee, AL 36088, USA
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria P.M.B. 1044, Nigeria
| | | | - Abdurrahman Pharmacy Yusuf
- Department of Biochemistry, School of Life Sciences, Federal University of Technology, Minna P.M.B. 65, Nigeria
| | - Murtala Bello Abubakar
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, Usmanu Danfodiyo University, Sokoto P.M.B. 2254, Nigeria
- Centre for Advanced Medical Research & Training (CAMRET), Usmanu Danfodiyo University, Sokoto P.M.B. 2254, Nigeria
| | - Idris Zubairu Sadiq
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria P.M.B. 1044, Nigeria
| | - Isra Elhussin
- Center for Cancer Research, Department of Biology, Tuskegee University, Tuskegee, AL 36088, USA
| | - Md Abu Talha Siddique
- Center for Cancer Research, Department of Biology, Tuskegee University, Tuskegee, AL 36088, USA
| | - Suleiman Aminu
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria P.M.B. 1044, Nigeria
| | - Rabiatu Bako Suleiman
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria P.M.B. 1044, Nigeria
| | - Yakubu Saddeeq Abubakar
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria P.M.B. 1044, Nigeria
| | - Babangida Sanusi Katsayal
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria P.M.B. 1044, Nigeria
| | - Clayton C Yates
- Center for Cancer Research, Department of Biology, Tuskegee University, Tuskegee, AL 36088, USA
| | - Sunila Mahavadi
- Center for Cancer Research, Department of Biology, Tuskegee University, Tuskegee, AL 36088, USA
| |
Collapse
|
6
|
Estrogen as a key regulator of energy homeostasis and metabolic health. Biomed Pharmacother 2022; 156:113808. [DOI: 10.1016/j.biopha.2022.113808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/02/2022] [Accepted: 10/03/2022] [Indexed: 11/23/2022] Open
|
7
|
Mauvais-Jarvis F, Lange CA, Levin ER. Membrane-Initiated Estrogen, Androgen, and Progesterone Receptor Signaling in Health and Disease. Endocr Rev 2022; 43:720-742. [PMID: 34791092 PMCID: PMC9277649 DOI: 10.1210/endrev/bnab041] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Indexed: 12/15/2022]
Abstract
Rapid effects of steroid hormones were discovered in the early 1950s, but the subject was dominated in the 1970s by discoveries of estradiol and progesterone stimulating protein synthesis. This led to the paradigm that steroid hormones regulate growth, differentiation, and metabolism via binding a receptor in the nucleus. It took 30 years to appreciate not only that some cellular functions arise solely from membrane-localized steroid receptor (SR) actions, but that rapid sex steroid signaling from membrane-localized SRs is a prerequisite for the phosphorylation, nuclear import, and potentiation of the transcriptional activity of nuclear SR counterparts. Here, we provide a review and update on the current state of knowledge of membrane-initiated estrogen (ER), androgen (AR) and progesterone (PR) receptor signaling, the mechanisms of membrane-associated SR potentiation of their nuclear SR homologues, and the importance of this membrane-nuclear SR collaboration in physiology and disease. We also highlight potential clinical implications of pathway-selective modulation of membrane-associated SR.
Collapse
Affiliation(s)
- Franck Mauvais-Jarvis
- Department of Medicine, Section of Endocrinology and Metabolism, Tulane University School of Medicine, New Orleans, LA, 70112, USA.,Tulane Center of Excellence in Sex-Based Biology & Medicine, New Orleans, LA, 70112, USA.,Southeast Louisiana Veterans Affairs Medical Center, New Orleans, LA, 70119, USA
| | - Carol A Lange
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA.,Department of Medicine (Division of Hematology, Oncology, and Transplantation), University of Minnesota, Minneapolis, MN 55455, USA.,Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Ellis R Levin
- Division of Endocrinology, Department of Medicine, University of California, Irvine, Irvine, CA, 92697, USA.,Department of Veterans Affairs Medical Center, Long Beach, Long Beach, CA, 90822, USA
| |
Collapse
|
8
|
Basile G, Qadir MMF, Mauvais-Jarvis F, Vetere A, Shoba V, Modell AE, Pastori RL, Russ HA, Wagner BK, Dominguez-Bendala J. Emerging diabetes therapies: Bringing back the β-cells. Mol Metab 2022; 60:101477. [PMID: 35331962 PMCID: PMC8987999 DOI: 10.1016/j.molmet.2022.101477] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 03/11/2022] [Accepted: 03/14/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Stem cell therapies are finally coming of age as a viable alternative to pancreatic islet transplantation for the treatment of insulin-dependent diabetes. Several clinical trials using human embryonic stem cell (hESC)-derived β-like cells are currently underway, with encouraging preliminary results. Remaining challenges notwithstanding, these strategies are widely expected to reduce our reliance on human isolated islets for transplantation procedures, making cell therapies available to millions of diabetic patients. At the same time, advances in our understanding of pancreatic cell plasticity and the molecular mechanisms behind β-cell replication and regeneration have spawned a multitude of translational efforts aimed at inducing β-cell replenishment in situ through pharmacological means, thus circumventing the need for transplantation. SCOPE OF REVIEW We discuss here the current state of the art in hESC transplantation, as well as the parallel quest to discover agents capable of either preserving the residual mass of β-cells or inducing their proliferation, transdifferentiation or differentiation from progenitor cells. MAJOR CONCLUSIONS Stem cell-based replacement therapies in the mold of islet transplantation are already around the corner, but a permanent cure for type 1 diabetes will likely require the endogenous regeneration of β-cells aided by interventions to restore the immune balance. The promise of current research avenues and a strong pipeline of clinical trials designed to tackle these challenges bode well for the realization of this goal.
Collapse
Affiliation(s)
- G Basile
- Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - M M F Qadir
- Tulane University School of Medicine, New Orleans, LA, USA; Southeast Louisiana Veterans Affairs Medical Center, New Orleans, LA, USA
| | - F Mauvais-Jarvis
- Tulane University School of Medicine, New Orleans, LA, USA; Southeast Louisiana Veterans Affairs Medical Center, New Orleans, LA, USA
| | - A Vetere
- Broad Institute, Cambridge, MA, USA
| | - V Shoba
- Broad Institute, Cambridge, MA, USA
| | | | - R L Pastori
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - H A Russ
- Barbara Davis Center for Diabetes, Colorado University Anschutz Medical Campus, Aurora, CO, USA.
| | | | - J Dominguez-Bendala
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
9
|
Fuselier T, Mota de Sa P, Qadir MMF, Xu B, Allard C, Meyers MM, Tiano JP, Yang BS, Gelfanov V, Lindsey SH, Dimarchi RD, Mauvais-Jarvis F. Efficacy of glucagon-like peptide-1 and estrogen dual agonist in pancreatic islets protection and pre-clinical models of insulin-deficient diabetes. Cell Rep Med 2022; 3:100598. [PMID: 35492248 PMCID: PMC9043999 DOI: 10.1016/j.xcrm.2022.100598] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/18/2022] [Accepted: 03/15/2022] [Indexed: 02/06/2023]
Abstract
We study the efficacy of a glucagon-like peptide-1 (GLP-1) and estrogen dual agonist (GLP1-E2) in pancreatic islet protection. GLP1-E2 provides superior protection from insulin-deficient diabetes induced by multiple low-dose streptozotocin (MLD-STZ-diabetes) and by the Akita mutation in mice than a GLP-1 monoagonist. GLP1-E2 does not protect from MLD-STZ-diabetes in estrogen receptor-α (ERα)-deficient mice and fails to prevent diabetes in Akita mice following GLP-1 receptor (GLP-1R) antagonism, demonstrating the requirement of GLP-1R and ERα for GLP1-E2 antidiabetic actions. In the MIN6 β cell model, GLP1-E2 activates estrogen action following clathrin-dependent, GLP-1R-mediated internalization and lysosomal acidification. In cultured human islet, proteomic bioinformatic analysis reveals that GLP1-E2 amplifies the antiapoptotic pathways activated by monoagonists. However, in cultured mouse islets, GLP1-E2 provides antiapoptotic protection similar to monoagonists. Thus, GLP1-E2 promotes GLP-1 and E2 antiapoptotic signals in cultured islets, but in vivo, additional GLP1-E2 actions in non-islet cells expressing GLP-1R are instrumental to prevent diabetes.
Collapse
Affiliation(s)
- Taylor Fuselier
- Deming Department of Medicine, Section of Endocrinology and Metabolism, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA 70112, USA
| | - Paula Mota de Sa
- Deming Department of Medicine, Section of Endocrinology and Metabolism, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA 70112, USA; Tulane Center of Excellence in Sex-Based Biology & Medicine, New Orleans, LA 70112, USA; Southeast Louisiana Veterans Healthcare System Medical Center, New Orleans, LA 70119, USA
| | - M M Fahd Qadir
- Deming Department of Medicine, Section of Endocrinology and Metabolism, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA 70112, USA; Tulane Center of Excellence in Sex-Based Biology & Medicine, New Orleans, LA 70112, USA; Southeast Louisiana Veterans Healthcare System Medical Center, New Orleans, LA 70119, USA
| | - Beibei Xu
- Deming Department of Medicine, Section of Endocrinology and Metabolism, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA 70112, USA
| | - Camille Allard
- Deming Department of Medicine, Section of Endocrinology and Metabolism, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA 70112, USA
| | - Mathew M Meyers
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Joseph P Tiano
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Bin S Yang
- Novo Nordisk Research Center Indianapolis, Indianapolis, IN 46241, USA
| | - Vasily Gelfanov
- Novo Nordisk Research Center Indianapolis, Indianapolis, IN 46241, USA
| | - Sarah H Lindsey
- Tulane Center of Excellence in Sex-Based Biology & Medicine, New Orleans, LA 70112, USA; Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | | | - Franck Mauvais-Jarvis
- Deming Department of Medicine, Section of Endocrinology and Metabolism, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA 70112, USA; Tulane Center of Excellence in Sex-Based Biology & Medicine, New Orleans, LA 70112, USA; Southeast Louisiana Veterans Healthcare System Medical Center, New Orleans, LA 70119, USA.
| |
Collapse
|
10
|
Lemos JRN, Baidal DA, Poggioli R, Fuenmayor V, Chavez C, Alvarez A, Linetsky E, Mauvais-Jarvis F, Ricordi C, Alejandro R. Prolonged Islet Allograft Function is Associated With Female Sex in Patients After Islet Transplantation. J Clin Endocrinol Metab 2022; 107:e973-e979. [PMID: 34727179 PMCID: PMC8852206 DOI: 10.1210/clinem/dgab787] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Indexed: 12/25/2022]
Abstract
BACKGROUND Islet transplantation (ITx) has proved to be effective in preventing severe hypoglycemia and improving metabolic control in selected subjects with type 1 diabetes. Long-term graft function remains a challenge. Estrogens have been shown to protect β cells from metabolic stresses and improve revascularization of transplanted human islets in the mouse. We aimed to evaluate the influence of sex in allograft survival of ITx recipients. METHODS We analyzed a retrospective cohort of ITx recipients (n = 56) followed-up for up to 20 years. Allograft failure was defined as a stimulated C-peptide <0.3 ng/mL during a mixed-meal tolerance test. Subjects were divided into recipients of at least 1 female donor (group 1) and recipients of male donors only (group 2). RESULTS Group 1 subjects (n = 25) were aged 41.5 ± 8.4 years and group 2 subjects (n = 22) 45.9 ± 7.3 years (P = 0.062). Female recipient frequency was 44.8% (n = 13) in group 1 and 55.2% (n = 16) in group 2 (P = 0.145). Group 2 developed graft failure earlier than group 1 (680 [286-1624] vs 1906 [756-3256] days, P = 0.038). We performed additional analyses on female recipients only from each group (group 1, n = 16; group 2, n = 20). Female recipients in group 1 exhibited prolonged allograft function compared with group 2, after adjustment for confounders (odds ratio, 28.6; 95% CI, 1.3-619.1; P < 0.05). CONCLUSION Recipients of islets from at least 1 female donor exhibited prolonged graft survival compared with recipients of islets from exclusively male donors. In addition, female recipients exhibited prolonged survival compared with male recipients following ITx of at least 1 female donor.
Collapse
Affiliation(s)
- Joana R N Lemos
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - David A Baidal
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Tulane Center of Excellence in Sex Based Biology & Medicine, New Orleans, LA 70112, USA
| | - Raffaella Poggioli
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Virginia Fuenmayor
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Carmen Chavez
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Ana Alvarez
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Elina Linetsky
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Franck Mauvais-Jarvis
- Tulane Center of Excellence in Sex Based Biology & Medicine, New Orleans, LA 70112, USA
- Diabetes Discovery Research & Sex-Based Medicine Laboratory, New Orleans, LA 70112, USA
- Section of Endocrinology and Metabolism, Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA
- Southeast Louisiana Veterans Health Care System, New Orleans, LA 70119, USA
| | - Camillo Ricordi
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Division of Cellular Transplantation, Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Rodolfo Alejandro
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
11
|
Xu W, Schiffer L, Qadir MMF, Zhang Y, Hawley J, Mota De Sa P, Keevil BG, Wu H, Arlt W, Mauvais-Jarvis F. Intracrine Testosterone Activation in Human Pancreatic β-Cells Stimulates Insulin Secretion. Diabetes 2020; 69:2392-2399. [PMID: 32855171 PMCID: PMC7576567 DOI: 10.2337/db20-0228] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 08/21/2020] [Indexed: 02/06/2023]
Abstract
Testosterone (T) affects β-cell function in men and women. T is a prohormone that undergoes intracrine conversion in target tissues to the potent androgen dihydrotestosterone (DHT) via the enzyme 5α-reductase (5α-R) or to the active estrogen 17β-estradiol (E2) via the aromatase enzyme. Using male and female human pancreas sections, we show that the 5α-R type 1 isoform (SRD5A1) and aromatase are expressed in male and female β-cells. We show that cultured male and female human islets exposed to T produce DHT and downstream metabolites. In these islets, exposure to the 5α-R inhibitors finasteride and dutasteride inhibited T conversion into DHT. We did not detect T conversion into E2 from female islets. However, we detected T conversion into E2 in islets from two out of four male donors. In these donors, exposure to the aromatase inhibitor anastrozole inhibited E2 production. Notably, in cultured male and female islets, T enhanced glucose-stimulated insulin secretion (GSIS). In these islets, exposure to 5α-R inhibitors or the aromatase inhibitor both inhibited T enhancement of GSIS. In conclusion, male and female human islets convert T into DHT and E2 via the intracrine activities of SRD5A1 and aromatase. This process is necessary for T enhancement of GSIS.
Collapse
Affiliation(s)
- Weiwei Xu
- Section of Endocrinology and Metabolism, Department of Medicine, Tulane University Health Sciences Center, New Orleans, LA
| | - Lina Schiffer
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, U.K
| | - M M Fahd Qadir
- Section of Endocrinology and Metabolism, Department of Medicine, Tulane University Health Sciences Center, New Orleans, LA
| | - Yanqing Zhang
- Section of Endocrinology and Metabolism, Department of Medicine, Tulane University Health Sciences Center, New Orleans, LA
| | - James Hawley
- Department of Clinical Biochemistry, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester, U.K
| | - Paula Mota De Sa
- Section of Endocrinology and Metabolism, Department of Medicine, Tulane University Health Sciences Center, New Orleans, LA
| | - Brian G Keevil
- Department of Clinical Biochemistry, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester, U.K
| | - Hongju Wu
- Section of Endocrinology and Metabolism, Department of Medicine, Tulane University Health Sciences Center, New Orleans, LA
| | - Wiebke Arlt
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, U.K
- National Institute for Health Research Birmingham Biomedical Research Centre, University of Birmingham and University Hospitals Birmingham NHS Foundation Trust, Birmingham, U.K
| | - Franck Mauvais-Jarvis
- Section of Endocrinology and Metabolism, Department of Medicine, Tulane University Health Sciences Center, New Orleans, LA
- Southeast Louisiana Veterans Health Care System, New Orleans, LA
| |
Collapse
|
12
|
Salvoza NC, Giraudi PJ, Tiribelli C, Rosso N. Sex differences in non-alcoholic fatty liver disease: hints for future management of the disease. EXPLORATION OF MEDICINE 2020. [DOI: 10.37349/emed.2020.00005] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) remains a major cause of chronic liver disease worldwide. Despite extensive studies, the heterogeneity of the risk factors as well as different disease mechanisms complicate the goals toward effective diagnosis and management. Recently, it has been shown that sex differences play a role in the prevalence and progression of NAFLD. In vitro, in vivo, and clinical studies revealed that the lower prevalence of NAFLD in premenopausal as compared to postmenopausal women and men is mainly due to the protective effects of estrogen and body fat distribution. It has been also described that males and females present differential pathogenic features in terms of biochemical profiles and histological characteristics. However, the exact molecular mechanisms for the gender differences that exist in the pathogenesis of NAFLD are still elusive. Lipogenesis, oxidative stress, and inflammation play a key role in the progression of NAFLD. For NAFLD, only a few studies characterized these mechanisms at the molecular level. Therefore, we aim to review the reported differential molecular mechanisms that trigger such different pathogenesis in both sexes. Differences in lipid metabolism, glucose homeostasis, oxidative stress, inflammation, and fibrosis were discussed based on the evidence reported in recent publications. In conclusion, with this review, we hope to provide a new perspective for the development of future practice guidelines as well as a new avenue for the management of the disease.
Collapse
Affiliation(s)
- Noel C. Salvoza
- Fondazione Italiana Fegato ONLUS, Area Science Park Basovizza SS14 km 163.5, 34149 Trieste, Italy; Philippine Council for Health Research and Development, DOST Compound, Bicutan Taguig City 1631, Philippines
| | - Pablo J. Giraudi
- Fondazione Italiana Fegato ONLUS, Area Science Park Basovizza SS14 km 163.5, 34149 Trieste, Italy
| | - Claudio Tiribelli
- Fondazione Italiana Fegato ONLUS, Area Science Park Basovizza SS14 km 163.5, 34149 Trieste, Italy
| | - Natalia Rosso
- Fondazione Italiana Fegato ONLUS, Area Science Park Basovizza SS14 km 163.5, 34149 Trieste, Italy
| |
Collapse
|
13
|
A Selective Ligand for Estrogen Receptor Proteins Discriminates Rapid and Genomic Signaling. Cell Chem Biol 2019; 26:1692-1702.e5. [PMID: 31706983 DOI: 10.1016/j.chembiol.2019.10.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 09/05/2019] [Accepted: 10/18/2019] [Indexed: 12/27/2022]
Abstract
Estrogen exerts extensive and diverse effects throughout the body of women. In addition to the classical nuclear estrogen receptors (ERα and ERβ), the G protein-coupled estrogen receptor GPER is an important mediator of estrogen action. Existing ER-targeted therapeutic agents act as GPER agonists. Here, we report the identification of a small molecule, named AB-1, with the previously unidentified activity of high selectivity for binding classical ERs over GPER. AB-1 also possesses a unique functional activity profile as an agonist of transcriptional activity but an antagonist of rapid signaling through ERα. Our results define a class of small molecules that discriminate between the classical ERs and GPER, as well as between modes of signaling within the classical ERs. Such an activity profile, if developed into an ER antagonist, could represent an opportunity for the development of first-in-class nuclear hormone receptor-targeted therapeutics for breast cancer exhibiting reduced acquired and de novo resistance.
Collapse
|
14
|
Vantyghem MC, Chetboun M, Gmyr V, Jannin A, Espiard S, Le Mapihan K, Raverdy V, Delalleau N, Machuron F, Hubert T, Frimat M, Van Belle E, Hazzan M, Pigny P, Noel C, Caiazzo R, Kerr-Conte J, Pattou F. Ten-Year Outcome of Islet Alone or Islet After Kidney Transplantation in Type 1 Diabetes: A Prospective Parallel-Arm Cohort Study. Diabetes Care 2019; 42:2042-2049. [PMID: 31615852 DOI: 10.2337/dc19-0401] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 08/03/2019] [Indexed: 02/03/2023]
Abstract
OBJECTIVE The long-term outcome of allogenic islet transplantation is unknown. The aim of this study was to evaluate the 10-year outcome of islet transplantation in patients with type 1 diabetes and hypoglycemia unawareness and/or a functioning kidney graft. RESEARCH DESIGN AND METHODS We enrolled in this prospective parallel-arm cohort study 28 subjects with type 1 diabetes who received islet transplantation either alone (ITA) or after a kidney graft (IAK). Islet transplantation consisted of two or three intraportal infusions of allogenic islets administered within (median [interquartile range]) 68 days (43-92). Immunosuppression was induced with interleukin-2 receptor antibodies and maintained with sirolimus and tacrolimus. The primary outcome was insulin independence with A1C ≤6.5% (48 mmol/mol). Secondary outcomes were patient and graft survival, severe hypoglycemic events (SHEs), metabolic control, and renal function. RESULTS The primary outcome was met by (Kaplan-Meier estimates [95% CI]) 39% (22-57) and 28% (13-45) of patients 5 and 10 years after islet transplantation, respectively. Graft function persisted in 82% (62-92) and 78% (57-89) of case subjects after 5 and 10 years, respectively, and was associated with improved glucose control, reduced need for exogenous insulin, and a marked decrease of SHEs. ITA and IAK had similar outcomes. Primary graft function, evaluated 1 month after the last islet infusion, was significantly associated with the duration of graft function and insulin independence. CONCLUSIONS Islet transplantation with the Edmonton protocol can provide 10-year markedly improved metabolic control without SHEs in three-quarters of patients with type 1 diabetes, kidney transplanted or not.
Collapse
Affiliation(s)
- Marie-Christine Vantyghem
- University of Lille, U1190-EGID, Lille, France .,Department of Endocrinology, Diabetology, and Metabolism, Centre Hospitalier Universitaire de Lille, Lille, France.,Inserm, U1190, Lille, France
| | - Mikael Chetboun
- University of Lille, U1190-EGID, Lille, France.,Inserm, U1190, Lille, France.,Department of General and Endocrine Surgery, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Valéry Gmyr
- University of Lille, U1190-EGID, Lille, France.,Inserm, U1190, Lille, France
| | - Arnaud Jannin
- Department of Endocrinology, Diabetology, and Metabolism, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Stéphanie Espiard
- Department of Endocrinology, Diabetology, and Metabolism, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Kristell Le Mapihan
- Department of Endocrinology, Diabetology, and Metabolism, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Violeta Raverdy
- University of Lille, U1190-EGID, Lille, France.,Inserm, U1190, Lille, France
| | - Nathalie Delalleau
- University of Lille, U1190-EGID, Lille, France.,Inserm, U1190, Lille, France
| | - François Machuron
- Department of Methodology, Biostatistics, and Data Management, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Thomas Hubert
- University of Lille, U1190-EGID, Lille, France.,Inserm, U1190, Lille, France
| | - Marie Frimat
- Department of Nephrology, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Eric Van Belle
- Department of Cardiology, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Marc Hazzan
- Department of Nephrology, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Pascal Pigny
- Department of Biochemistry and Hormonology, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Christian Noel
- Department of Nephrology, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Robert Caiazzo
- University of Lille, U1190-EGID, Lille, France.,Inserm, U1190, Lille, France.,Department of General and Endocrine Surgery, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Julie Kerr-Conte
- University of Lille, U1190-EGID, Lille, France.,Inserm, U1190, Lille, France
| | - François Pattou
- University of Lille, U1190-EGID, Lille, France .,Inserm, U1190, Lille, France.,Department of General and Endocrine Surgery, Centre Hospitalier Universitaire de Lille, Lille, France
| | | |
Collapse
|
15
|
Ahn SH, Granger A, Rankin MM, Lam CJ, Cox AR, Kushner JA. Tamoxifen suppresses pancreatic β-cell proliferation in mice. PLoS One 2019; 14:e0214829. [PMID: 31490929 PMCID: PMC6731016 DOI: 10.1371/journal.pone.0214829] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 08/11/2019] [Indexed: 12/13/2022] Open
Abstract
Tamoxifen is a mixed agonist/antagonist estrogen analogue that is frequently used to induce conditional gene deletion in mice using Cre-loxP mediated gene recombination. Tamoxifen is routinely employed in extremely high-doses relative to typical human doses to induce efficient gene deletion in mice. Although tamoxifen has been widely assumed to have no influence upon β-cells, the acute developmental and functional consequences of high-dose tamoxifen upon glucose homeostasis and adult β-cells are largely unknown. We tested if tamoxifen influences glucose homeostasis in male mice of various genetic backgrounds. We then carried out detailed histomorphometry studies of mouse pancreata. We also performed gene expression studies with islets of tamoxifen-treated mice and controls. Tamoxifen had modest effects upon glucose homeostasis of mixed genetic background (F1 B6129SF1/J) mice, with fasting hyperglycemia and improved glucose tolerance but without overt effects on fed glucose levels or insulin sensitivity. Tamoxifen inhibited proliferation of β-cells in a dose-dependent manner, with dramatic reductions in β-cell turnover at the highest dose (decreased by 66%). In sharp contrast, tamoxifen did not reduce proliferation of pancreatic acinar cells. β-cell proliferation was unchanged by tamoxifen in 129S2 mice but was reduced in C57Bl6 genetic background mice (decreased by 59%). Gene expression studies revealed suppression of RNA for cyclins D1 and D2 within islets of tamoxifen-treated mice. Tamoxifen has a cytostatic effect on β-cells, independent of changes in glucose homeostasis, in mixed genetic background and also in C57Bl6 mice. Tamoxifen should be used judiciously to inducibly inactivate genes in studies of glucose homeostasis.
Collapse
Affiliation(s)
- Surl-Hee Ahn
- Pediatric Endocrinology and Diabetes, University of Pennsylvania School of Medicine, Philadelphia, PA, United States of America
| | - Anne Granger
- Pediatric Endocrinology and Diabetes, University of Pennsylvania School of Medicine, Philadelphia, PA, United States of America
| | - Matthew M. Rankin
- Pediatric Endocrinology and Diabetes, University of Pennsylvania School of Medicine, Philadelphia, PA, United States of America
| | - Carol J. Lam
- Pediatric Endocrinology and Diabetes, University of Pennsylvania School of Medicine, Philadelphia, PA, United States of America
- McNair Medical Institute, Baylor College of Medicine, Houston, TX, United States of America
| | - Aaron R. Cox
- McNair Medical Institute, Baylor College of Medicine, Houston, TX, United States of America
| | - Jake A. Kushner
- Pediatric Endocrinology and Diabetes, University of Pennsylvania School of Medicine, Philadelphia, PA, United States of America
- McNair Medical Institute, Baylor College of Medicine, Houston, TX, United States of America
- * E-mail:
| |
Collapse
|
16
|
Handgraaf S, Philippe J. The Role of Sexual Hormones on the Enteroinsular Axis. Endocr Rev 2019; 40:1152-1162. [PMID: 31074764 DOI: 10.1210/er.2019-00004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Accepted: 05/03/2019] [Indexed: 12/17/2022]
Abstract
Sex steroid estrogens, androgens, and progesterone, produced by the gonads, which have long been considered as endocrine glands, are implicated in sexual differentiation, puberty, and reproduction. However, the impact of sex hormones goes beyond these effects through their role on energy metabolism. Indeed, sex hormones are important physiological regulators of glucose homeostasis and, in particular, of the enteroinsular axis. In this review, we describe the roles of estrogens, androgens, and progesterone on glucose homeostasis through their effects on pancreatic α- and β-cells, as well as on enteroendocrine L-cells, and their implications in hormonal biosynthesis and secretion. The analysis of their mechanisms of action with the dissection of the receptors implicated in the several protective effects could provide some new aspects of the fine-tuning of hormonal secretion under the influence of the sex. This knowledge paves the way to the understanding of transgender physiology and new potential therapeutics in the field of type 2 diabetes.
Collapse
Affiliation(s)
- Sandra Handgraaf
- Laboratory of Molecular Diabetes, Division of Endocrinology, Diabetes, Hypertension, and Nutrition, University Hospital/Diabetes Center/University of Geneva Medical School, Geneva, Switzerland
| | - Jacques Philippe
- Laboratory of Molecular Diabetes, Division of Endocrinology, Diabetes, Hypertension, and Nutrition, University Hospital/Diabetes Center/University of Geneva Medical School, Geneva, Switzerland
| |
Collapse
|
17
|
Lovre D, Peacock E, Katalenich B, Moreau C, Xu B, Tate C, Utzschneider KM, Gautier JF, Fonseca V, Mauvais-Jarvis F. Conjugated Estrogens and Bazedoxifene Improve β Cell Function in Obese Menopausal Women. J Endocr Soc 2019; 3:1583-1594. [PMID: 31384719 PMCID: PMC6676076 DOI: 10.1210/js.2019-00074] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 05/31/2019] [Indexed: 12/21/2022] Open
Abstract
CONTEXT Studies suggest that menopausal hormone therapy (MHT) prevents type 2 diabetes (T2D). The combination of conjugated estrogens (CE) with the selective estrogen receptor modulator bazedoxifene (BZA) is an MHT that improves obesity and T2D in preclinical models of menopausal metabolic syndrome. The effect of CE/BZA on adiposity and glucose homeostasis in obese postmenopausal women is unknown. OBJECTIVE To investigate the effect of CE/BZA on body composition, glucose homeostasis, and markers of inflammation in obese postmenopausal women. RESEARCH DESIGN INTERVENTION AND PARTICIPANTS Randomized, double-blind, placebo-controlled pilot trial of 12 obese menopausal women assigned to 12-week treatment with CE 0.45 mg/BZA 20 mg (n = 7) or placebo (n = 5). At baseline and after 12 weeks, we assessed body composition (dual-energy X-ray absorptiometry), glucose homeostasis (IV glucose tolerance test), and inflammation biomarkers. RESULTS Women treated with CE/BZA exhibited increased β cell function using homeostatic model assessment-B [median (interquartile range) CE/BZA vs placebo: 18.5 (-0.9 to 320.6) μU/mM vs -25.5 (-39.9 to -0.1) μU/mM; P = 0.045], and decreased basal glucose concentrations (Gb) [-5.2 (-9.2 to -1.7) mg/dL vs 2.7 (0.9 to 4.9) mg/dL; P = 0.029]. Insulin sensitivity was higher in the placebo arm [1.35 (1.12 to 1.82) (μU/mL) min-1 vs -0.24 (-1.50 to 0.19) (μU/mL) min-1; P = 0.029]. No changes between treatment groups were observed for the acute insulin response to glucose (AIRg), the disposition index (DI), body composition, and inflammatory biomarkers. CONCLUSIONS A 12-week treatment of obese postmenopausal women with CEs/BZA improves fasting β cell function and glucose concentrations without change in AIRg, HOMA-IR, DI, body composition, or markers of inflammation.
Collapse
Affiliation(s)
- Dragana Lovre
- Section of Endocrinology and Metabolism, Tulane University Health Sciences Center, New Orleans, Louisiana
- Section of Endocrinology, Southeast Louisiana Veterans Healthcare System, New Orleans, Louisiana
| | - Erin Peacock
- Department of Medicine, Tulane University Health Sciences Center, New Orleans, Louisiana
| | - Bonnie Katalenich
- Clinical Translational Unit, School of Medicine, Tulane University, New Orleans, Louisiana
| | - Cynthia Moreau
- Clinical Translational Unit, School of Medicine, Tulane University, New Orleans, Louisiana
| | - Beibei Xu
- Section of Endocrinology and Metabolism, Tulane University Health Sciences Center, New Orleans, Louisiana
| | - Chandra Tate
- Section of Endocrinology and Metabolism, Tulane University Health Sciences Center, New Orleans, Louisiana
| | - Kristina M Utzschneider
- Division of Metabolism, Endocrinology and Nutrition, VA Puget Sound Health Care System and University of Washington, Seattle, Washington
| | - Jean-François Gautier
- Department of Diabetes and Endocrinology, Assistance Publique-Hôpitaux de Paris, Lariboisière Hospital, University Paris-Diderot Paris-7, Paris, France
- INSERM UMRS 1138, Cordeliers Research Center, Paris, France
| | - Vivian Fonseca
- Section of Endocrinology and Metabolism, Tulane University Health Sciences Center, New Orleans, Louisiana
- Section of Endocrinology, Southeast Louisiana Veterans Healthcare System, New Orleans, Louisiana
| | - Franck Mauvais-Jarvis
- Section of Endocrinology and Metabolism, Tulane University Health Sciences Center, New Orleans, Louisiana
- Section of Endocrinology, Southeast Louisiana Veterans Healthcare System, New Orleans, Louisiana
| |
Collapse
|
18
|
Henstridge DC, Abildgaard J, Lindegaard B, Febbraio MA. Metabolic control and sex: A focus on inflammatory-linked mediators. Br J Pharmacol 2019; 176:4193-4207. [PMID: 30820935 DOI: 10.1111/bph.14642] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 12/05/2018] [Accepted: 01/21/2019] [Indexed: 12/15/2022] Open
Abstract
Men and women have many differing biological and physiological characteristics. Thus, it is no surprise that the control of metabolic processes and the mechanisms underlying metabolic-related diseases have sex-specific components. There is a clear metabolic sexual dimorphism in that up until midlife, men have a far greater likelihood of acquiring cardio-metabolic disease than women. Following menopause, however, this difference is reduced, suggestive of a protective role of the female sex hormones. Inflammatory processes have been implicated in the pathogenesis of cardio-metabolic disease with human studies correlating metabolic disease acquisition or risk with levels of various inflammatory markers. Rodent studies employing genetic modifications or novel pharmacological approaches have provided mechanistic insight into the role of these inflammatory mediators. Sex differences impact inflammatory processes and the subsequent biological response. As a consequence, this may affect how inflammation alters metabolic processes between the sexes. Recently, some of our work in the field of inflammatory genes and metabolic control identified a sexual dimorphism in a preclinical model and caused us to question the frequency and scale of such findings in the literature. This review concentrates on inflammatory-related signalling in relation to obesity, insulin resistance, and type 2 diabetes and highlights the differences observed between males and females. Differences in the activation and signalling of various inflammatory genes and proteins present another reason why studying both male and female patients or animals is important in the context of understanding and finding therapeutics for metabolic-related disease. LINKED ARTICLES: This article is part of a themed section on The Importance of Sex Differences in Pharmacology Research. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.21/issuetoc.
Collapse
Affiliation(s)
- Darren C Henstridge
- Molecular Metabolism & Aging Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.,School of Health Sciences, University of Tasmania, Launceston, Tasmania, Australia
| | - Julie Abildgaard
- The Centre of Inflammation and Metabolism and the Centre for Physical Activity Research, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Birgitte Lindegaard
- The Centre of Inflammation and Metabolism and the Centre for Physical Activity Research, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.,Department of Pulmonary and Infectious Diseases, Nordsjaellands Hospital, Hillerød, Denmark
| | - Mark A Febbraio
- Division of Diabetes & Metabolism, Garvan Institute of Medical Research, Sydney, New South Wales, Australia.,Drug Discover Biology, Monash Institute of Pharmaceutical Sciences, Melbourne, Victoria, Australia
| |
Collapse
|
19
|
Sekido T, Nishio SI, Ohkubo Y, Sekido K, Kitahara J, Miyamoto T, Komatsu M. Repression of insulin gene transcription by indirect genomic signaling via the estrogen receptor in pancreatic beta cells. In Vitro Cell Dev Biol Anim 2019; 55:226-236. [PMID: 30790128 PMCID: PMC6443913 DOI: 10.1007/s11626-019-00328-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 01/27/2019] [Indexed: 12/19/2022]
Abstract
The mechanism whereby 17β-estradiol (E2) mediates insulin gene transcription has not been fully elucidated. In this study, exposure of hamster insulinoma (HIT-T15) cells to 5 × 10-9 to 1 × 10-7 M E2 led to a concentration-dependent decrease of insulin mRNA levels. Transient expression of the estrogen receptor (ER) in HIT-T15 cells revealed that estrogen receptor α (ERα) repressed transcription of the rat insulin II promoter in both ligand-dependent and ligand-independent manners. The N-terminal A/B domain of ERα was not required for either activity. However, the repression was absent with mutated ER lacking the DNA-binding domain. Moreover, introducing mutations in the D-box and P-box of the zinc finger of ER (C227S, C202L) also abolished the repression. Deletion of the insulin promoter region revealed that nucleotide positions - 238 to - 144 (relative to the transcriptional start site) were needed for ER repression of the rat insulin II gene. PDX1- and BETA2-binding sites were required for the repression, but an estrogen response element-like sequence or an AP1 site in the promoter was not involved. In conclusion, we found that estrogen repressed insulin mRNA expression in a beta cell line. In addition, the ER suppressed insulin gene transcription in a ligand-independent matter. These observations suggest ER may regulate insulin transcription by indirect genomic signaling.
Collapse
Affiliation(s)
- Takashi Sekido
- Division of Diabetes, Endocrinology and Metabolism, Department of Internal Medicine, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
| | - Shin-Ichi Nishio
- Division of Diabetes, Endocrinology and Metabolism, Department of Internal Medicine, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan.
| | - Yohsuke Ohkubo
- Division of Diabetes, Endocrinology and Metabolism, Department of Internal Medicine, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
| | - Keiko Sekido
- Division of Diabetes, Endocrinology and Metabolism, Department of Internal Medicine, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
| | - Junichiro Kitahara
- Division of Diabetes, Endocrinology and Metabolism, Department of Internal Medicine, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
| | | | - Mitsuhisa Komatsu
- Division of Diabetes, Endocrinology and Metabolism, Department of Internal Medicine, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
| |
Collapse
|
20
|
Bian C, Bai B, Gao Q, Li S, Zhao Y. 17β-Estradiol Regulates Glucose Metabolism and Insulin Secretion in Rat Islet β Cells Through GPER and Akt/mTOR/GLUT2 Pathway. Front Endocrinol (Lausanne) 2019; 10:531. [PMID: 31447779 PMCID: PMC6691154 DOI: 10.3389/fendo.2019.00531] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 07/17/2019] [Indexed: 12/14/2022] Open
Abstract
Aims: To explore the molecular mechanism by which 17β-estradiol (estrogen 2, E2) regulates glucose transporter 2 (GLUT2) and insulin secretion in islet β cells through G protein-coupled estrogen receptor (GPER) via Akt/mTOR pathway. Methods: SPF-grade SD male rats were used to establish an in vivo type 2 diabetes model treated with E2. Rat insulinoma cells (INS-1) were cultured in normal or high glucose media with or without E2. Immunofluorescence double staining was used to detect GPER, GLUT2, insulin, and glucagon immunolocalization in rat islet tissues. Western blot was used to detect GPER, Akt, mTOR, and GLUT2 protein immunocontent. Real-time PCR detected Slc2a2 and glucose kinase (GK) content, and ELISA was used to detect insulin levels. Glucose uptake, GK activity and pyruvate dehydrogenase (PDH) activity were analyzed with glucose detection, GK activity and PDH activity assay kit. Results: Immunofluorescence double staining confocal indicated that E2 treatment up-regulated expression levels of GPER, GLUT2, and insulin, while down-regulated glucagon. Western blot results revealed E2 increased GPER, Akt/mTOR pathway, and GLUT2 protein immunocontent. Real-time PCR showed E2 elevated Slc2a2, GK content. Moreover, E2 improved insulin secretion, glucose uptake, GK activity, and PDH activity. Conclusion: Our findings indicated that exogenous E2 up-regulated GPER via the Akt/mTOR pathway to increase GLUT2 protein content and insulin secretion in islet β cells.
Collapse
Affiliation(s)
- Che Bian
- Department of Endocrinology, The First Affiliated Hospital of China Medical University, Shenyang, China
- Department of Endocrinology and Metabolism, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Bowen Bai
- Department of Endocrinology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Qian Gao
- Department of Endocrinology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Siyi Li
- Department of Endocrinology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yuyan Zhao
- Department of Endocrinology, The First Affiliated Hospital of China Medical University, Shenyang, China
- *Correspondence: Yuyan Zhao
| |
Collapse
|
21
|
Kaliannan K, Robertson RC, Murphy K, Stanton C, Kang C, Wang B, Hao L, Bhan AK, Kang JX. Estrogen-mediated gut microbiome alterations influence sexual dimorphism in metabolic syndrome in mice. MICROBIOME 2018; 6:205. [PMID: 30424806 PMCID: PMC6234624 DOI: 10.1186/s40168-018-0587-0] [Citation(s) in RCA: 143] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Accepted: 10/30/2018] [Indexed: 05/17/2023]
Abstract
BACKGROUND Understanding the mechanism of the sexual dimorphism in susceptibility to obesity and metabolic syndrome (MS) is important for the development of effective interventions for MS. RESULTS Here we show that gut microbiome mediates the preventive effect of estrogen (17β-estradiol) on metabolic endotoxemia (ME) and low-grade chronic inflammation (LGCI), the underlying causes of MS and chronic diseases. The characteristic profiles of gut microbiome observed in female and 17β-estradiol-treated male and ovariectomized mice, such as decreased Proteobacteria and lipopolysaccharide biosynthesis, were associated with a lower susceptibility to ME, LGCI, and MS in these animals. Interestingly, fecal microbiota-transplant from male mice transferred the MS phenotype to female mice, while antibiotic treatment eliminated the sexual dimorphism in MS, suggesting a causative role of the gut microbiome in this condition. Moreover, estrogenic compounds such as isoflavones exerted microbiome-modulating effects similar to those of 17β-estradiol and reversed symptoms of MS in the male mice. Finally, both expression and activity of intestinal alkaline phosphatase (IAP), a gut microbiota-modifying non-classical anti-microbial peptide, were upregulated by 17β-estradiol and isoflavones, whereas inhibition of IAP induced ME and LGCI in female mice, indicating a critical role of IAP in mediating the effects of estrogen on these parameters. CONCLUSIONS In summary, we have identified a previously uncharacterized microbiome-based mechanism that sheds light upon sexual dimorphism in the incidence of MS and that suggests novel therapeutic targets and strategies for the management of obesity and MS in males and postmenopausal women.
Collapse
Affiliation(s)
- Kanakaraju Kaliannan
- Laboratory of Lipid Medicine and Technology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, 149 -13th Street, Boston, MA 02129 USA
| | - Ruairi C. Robertson
- School of Microbiology, University College Cork, Cork, Ireland
- Teagasc Moorepark Food Research Centre, Fermoy, Co., Cork, Ireland
- APC Microbiome Institute, University College Cork, Cork, Ireland
| | - Kiera Murphy
- School of Microbiology, University College Cork, Cork, Ireland
| | - Catherine Stanton
- School of Microbiology, University College Cork, Cork, Ireland
- Teagasc Moorepark Food Research Centre, Fermoy, Co., Cork, Ireland
- APC Microbiome Institute, University College Cork, Cork, Ireland
| | - Chao Kang
- Research Center for Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing Key Laboratory of Nutrition and Food Safety, Chongqing Medical Nutrition Research Center, Chongqing, People’s Republic of China
| | - Bin Wang
- Research Center for Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing Key Laboratory of Nutrition and Food Safety, Chongqing Medical Nutrition Research Center, Chongqing, People’s Republic of China
| | - Lei Hao
- Laboratory of Lipid Medicine and Technology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, 149 -13th Street, Boston, MA 02129 USA
| | - Atul K. Bhan
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114 USA
| | - Jing X. Kang
- Laboratory of Lipid Medicine and Technology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, 149 -13th Street, Boston, MA 02129 USA
| |
Collapse
|
22
|
Ren G, Wu C, Teng C, Yao Y. Synergistic effect of combined protopanaxatiol and ginsenoside Rh2 on antiproliferative activity in MDA-MB-231 human breast cancer cells in vitro. FOOD AGR IMMUNOL 2018. [DOI: 10.1080/09540105.2018.1490700] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Affiliation(s)
- Guixing Ren
- College of Pharmacy and Biological Engineering, Chengdu University, Chengdu City, People’s Republic of China
| | - Caie Wu
- College of Light Industry and Food Engineering, Nanjing Forestry University, Nanjing, People’s Republic of China
| | - Cong Teng
- Chinese Academy of Agricultural Sciences, Institute of Crop Science, Beijing, People’s Republic of China
| | - Yang Yao
- Chinese Academy of Agricultural Sciences, Institute of Crop Science, Beijing, People’s Republic of China
| |
Collapse
|
23
|
Kooptiwut S, Kaewin S, Semprasert N, Sujjitjoon J, Junking M, Suksri K, Yenchitsomanus PT. Estradiol Prevents High Glucose-Induced β-cell Apoptosis by Decreased BTG2 Expression. Sci Rep 2018; 8:12256. [PMID: 30115961 PMCID: PMC6095866 DOI: 10.1038/s41598-018-30698-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 08/03/2018] [Indexed: 01/20/2023] Open
Abstract
Hyperglycemia stimulates several pathways to induce pancreatic β-cell apoptosis. In our previous study by mRNA analysis, we demonstrated that B-cell translocation gene 2 (BTG2) expression was up-regulated in INS-1 cells cultured under high glucose conditions, but this effect was reversed by estrogen. In the present study, we demonstrated that BTG2 mRNA and protein expressions in both INS-1 cells and mouse pancreatic islets increased under high glucose conditions compared to those cultured under basal glucose conditions, while in the presence of estrogen, the BTG2 mRNA and protein expressions decreased. SiRNA-BTG2 significantly reduced cell apoptosis, cleaved-caspase 3, and Bax, compared to the siRNA-control in INS-1 cultured under high glucose conditions. We further demonstrated that BTG2 promoter activity was activated under high glucose conditions whereas estrogen significantly reduced it. The effects of estrogen on BTG2 expression were inhibited by estrogen receptor inhibitors. Also, under high glucose conditions, p53 and Bax mRNA and protein expressions increased, but they decreased in the presence of estrogen. Again, the effect of estrogen on p53 and Bax expression was inhibited by estrogen receptor inhibitors. Taken together, this study demonstrates that estrogen reduces pancreatic β-cell apoptosis under high glucose conditions via suppression of BTG2, p53, and Bax expressions.
Collapse
Affiliation(s)
- Suwattanee Kooptiwut
- Department of Physiology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand.
| | - Suchada Kaewin
- Department of Physiology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Namoiy Semprasert
- Department of Physiology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Jatuporn Sujjitjoon
- Division of Molecular Medicine, Research Department, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Mutita Junking
- Division of Molecular Medicine, Research Department, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Kanchana Suksri
- Department of Physiology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Pa-Thai Yenchitsomanus
- Division of Molecular Medicine, Research Department, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| |
Collapse
|
24
|
Xu B, Allard C, Alvarez-Mercado AI, Fuselier T, Kim JH, Coons LA, Hewitt SC, Urano F, Korach KS, Levin ER, Arvan P, Floyd ZE, Mauvais-Jarvis F. Estrogens Promote Misfolded Proinsulin Degradation to Protect Insulin Production and Delay Diabetes. Cell Rep 2018; 24:181-196. [PMID: 29972779 PMCID: PMC6092934 DOI: 10.1016/j.celrep.2018.06.019] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 05/11/2018] [Accepted: 06/01/2018] [Indexed: 02/06/2023] Open
Abstract
Conjugated estrogens (CE) delay the onset of type 2 diabetes (T2D) in postmenopausal women, but the mechanism is unclear. In T2D, the endoplasmic reticulum (ER) fails to promote proinsulin folding and, in failing to do so, promotes ER stress and β cell dysfunction. We show that CE prevent insulin-deficient diabetes in male and in female Akita mice using a model of misfolded proinsulin. CE stabilize the ER-associated protein degradation (ERAD) system and promote misfolded proinsulin proteasomal degradation. This involves activation of nuclear and membrane estrogen receptor-α (ERα), promoting transcriptional repression and proteasomal degradation of the ubiquitin-conjugating enzyme and ERAD degrader, UBC6e. The selective ERα modulator bazedoxifene mimics CE protection of β cells in females but not in males.
Collapse
Affiliation(s)
- Beibei Xu
- Diabetes Discovery Research and Gender Medicine Laboratory, Department of Medicine, Section of Endocrinology and Metabolism, School of Medicine, Tulane University Health Sciences Center, New Orleans, LA 70112, USA
| | - Camille Allard
- Diabetes Discovery Research and Gender Medicine Laboratory, Department of Medicine, Section of Endocrinology and Metabolism, School of Medicine, Tulane University Health Sciences Center, New Orleans, LA 70112, USA
| | - Ana I Alvarez-Mercado
- Diabetes Discovery Research and Gender Medicine Laboratory, Department of Medicine, Section of Endocrinology and Metabolism, School of Medicine, Tulane University Health Sciences Center, New Orleans, LA 70112, USA
| | - Taylor Fuselier
- Diabetes Discovery Research and Gender Medicine Laboratory, Department of Medicine, Section of Endocrinology and Metabolism, School of Medicine, Tulane University Health Sciences Center, New Orleans, LA 70112, USA; Southeast Louisiana Veterans Healthcare System Medical Center, New Orleans, LA 70112, USA
| | - Jun Ho Kim
- Department of Food Science and Biotechnology, Andong National University, Andong, Gyeongsangbuk-do 36729, South Korea
| | - Laurel A Coons
- Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, Durham, NC 27709, USA; Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Sylvia C Hewitt
- Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, Durham, NC 27709, USA
| | - Fumihiko Urano
- Department of Medicine, Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Kenneth S Korach
- Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, Durham, NC 27709, USA
| | - Ellis R Levin
- Division of Endocrinology, Veterans Affairs Medical Center, Long Beach, CA 90822, USA; Departments of Medicine and Biochemistry, University of California, Irvine, Irvine, CA 92717, USA
| | - Peter Arvan
- Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical School, Ann Arbor, MI 48105, USA
| | - Z Elizabeth Floyd
- Ubiquitin Lab, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70803, USA
| | - Franck Mauvais-Jarvis
- Diabetes Discovery Research and Gender Medicine Laboratory, Department of Medicine, Section of Endocrinology and Metabolism, School of Medicine, Tulane University Health Sciences Center, New Orleans, LA 70112, USA; Southeast Louisiana Veterans Healthcare System Medical Center, New Orleans, LA 70112, USA.
| |
Collapse
|
25
|
Mauvais-Jarvis F, Le May C, Tiano JP, Liu S, Kilic-Berkmen G, Kim JH. The Role of Estrogens in Pancreatic Islet Physiopathology. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1043:385-399. [PMID: 29224104 DOI: 10.1007/978-3-319-70178-3_18] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
In rodent models of insulin-deficient diabetes, 17β-estradiol (E2) protects pancreatic insulin-producing β-cells against oxidative stress, amyloid polypeptide toxicity, gluco-lipotoxicity, and apoptosis. Three estrogen receptors (ERs)-ERα, ERβ, and the G protein-coupled ER (GPER)-have been identified in rodent and human β-cells. This chapter describes recent advances in our understanding of the role of ERs in islet β-cell function, nutrient homeostasis, survival from pro-apoptotic stimuli, and proliferation. We discuss why and how ERs represent potential therapeutic targets for the maintenance of functional β-cell mass.
Collapse
Affiliation(s)
- Franck Mauvais-Jarvis
- Department of Medicine, Section of Endocrinology and Metabolism, Tulane University Health Sciences Center, School of Medicine, New Orleans, LA, USA.
| | - Cedric Le May
- L'institut du Thorax, INSERM-CNRS, University of Nantes, Nantes, France
| | - Joseph P Tiano
- Diabetes, Endocrinology, and Obesity Branch, NIDDK, Bethesda, MD, USA
| | - Suhuan Liu
- Xiamen Diabetes Institute, the First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Gamze Kilic-Berkmen
- Department of Pediatric, Emory University School of Medicine, Atlanta, GA, USA
| | - Jun Ho Kim
- Department of Food and Biotechnology, Korea University, Sejong, South Korea
| |
Collapse
|
26
|
Gannon M, Kulkarni RN, Tse HM, Mauvais-Jarvis F. Sex differences underlying pancreatic islet biology and its dysfunction. Mol Metab 2018; 15:82-91. [PMID: 29891438 PMCID: PMC6066785 DOI: 10.1016/j.molmet.2018.05.017] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 05/25/2018] [Indexed: 12/30/2022] Open
Abstract
Background The sex of an individual affects glucose homeostasis and the pathophysiology, incidence, and prevalence of diabetes as well as the response to therapy. Scope of the review This review focuses on clinical and experimental sex differences in islet cell biology and dysfunction during development and in adulthood in human and animal models. We discuss sex differences in β-cell and α-cell function, heterogeneity, and dysfunction. We cover sex differences in communication between gonads and islets and islet-cell immune interactions. Finally, we discuss sex differences in β-cell programming by nutrition and other environmental factors during pregnancy. Major conclusions Important sex differences exist in islet cell function and susceptibility to failure. These differences represent sex-related biological factors that can be harnessed for gender-based prevention of and therapy for diabetes.
Collapse
Affiliation(s)
- Maureen Gannon
- Department of Medicine, Vanderbilt University Medical Center, Nashville, USA; Department of Veterans Affairs, Tennessee Valley Health Authority, Nashville, TN, USA
| | - Rohit N Kulkarni
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, USA; Department of Medicine, Harvard Medical School, Boston, USA; Harvard Stem Cell Institute, Boston, MA, USA
| | - Hubert M Tse
- Department of Microbiology, Birmingham, USA; Comprehensive Diabetes Center, University of Alabama at Birmingham School of Medicine, Birmingham, AL, USA
| | - Franck Mauvais-Jarvis
- Department of Medicine, Section of Endocrinology and Metabolism, Tulane University Health Sciences Center School of Medicine, New Orleans, USA; Southeast Louisiana Veterans Healthcare System Medical Center, New Orleans, LA, USA.
| |
Collapse
|
27
|
Handgraaf S, Dusaulcy R, Visentin F, Philippe J, Gosmain Y. 17-β Estradiol regulates proglucagon-derived peptide secretion in mouse and human α- and L cells. JCI Insight 2018; 3:98569. [PMID: 29618657 DOI: 10.1172/jci.insight.98569] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 02/28/2018] [Indexed: 01/11/2023] Open
Abstract
Clinical and experimental data indicate a beneficial effect of estrogens on energy and glucose homeostasis associated with improved insulin sensitivity and positive effects on insulin secretion. The aim of the study was to investigate the impact of estrogens on proglucagon-producing cells, pancreatic α cells, and enteroendocrine L cells. The consequences of sexual hormone deprivation were evaluated in ovariectomized mice (ovx). Ovx mice exhibited impaired glucose tolerance during oral glucose tolerance tests (OGTT), which was associated with decreased GLP-1 intestinal and pancreatic secretion and content, an effect that was reversed by estradiol (E2) treatment. Indeed, E2 increased oral glucose-induced GLP-1 secretion in vivo and GLP-1 secretion from primary culture of mouse and human α cells through the activation of all 3 estrogen receptors (ERs), whereas E2-induced GLP-1 secretion from mouse and human intestinal explants occurred only by ERβ activation. Underlying the implication of ERβ, its selective agonist WAY20070 was able to restore glucose tolerance in ovx mice at least partly through plasma GLP-1 increase. We conclude that E2 directly controls both α- and L cells to increase GLP-1 secretion, in addition to its effects on insulin and glucagon secretion, highlighting the potential beneficial role of the estrogenic pathway and, more particularly, of ERβ agonists to prevent type 2 diabetes.
Collapse
|
28
|
Zhou Z, Ribas V, Rajbhandari P, Drew BG, Moore TM, Fluitt AH, Reddish BR, Whitney KA, Georgia S, Vergnes L, Reue K, Liesa M, Shirihai O, van der Bliek AM, Chi NW, Mahata SK, Tiano JP, Hewitt SC, Tontonoz P, Korach KS, Mauvais-Jarvis F, Hevener AL. Estrogen receptor α protects pancreatic β-cells from apoptosis by preserving mitochondrial function and suppressing endoplasmic reticulum stress. J Biol Chem 2018; 293:4735-4751. [PMID: 29378845 PMCID: PMC5880140 DOI: 10.1074/jbc.m117.805069] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 11/24/2017] [Indexed: 12/25/2022] Open
Abstract
Estrogen receptor α (ERα) action plays an important role in pancreatic β-cell function and survival; thus, it is considered a potential therapeutic target for the treatment of type 2 diabetes in women. However, the mechanisms underlying the protective effects of ERα remain unclear. Because ERα regulates mitochondrial metabolism in other cell types, we hypothesized that ERα may act to preserve insulin secretion and promote β-cell survival by regulating mitochondrial-endoplasmic reticulum (EndoRetic) function. We tested this hypothesis using pancreatic islet-specific ERα knockout (PERαKO) mice and Min6 β-cells in culture with Esr1 knockdown (KD). We found that Esr1-KD promoted reactive oxygen species production that associated with reduced fission/fusion dynamics and impaired mitophagy. Electron microscopy showed mitochondrial enlargement and a pro-fusion phenotype. Mitochondrial cristae and endoplasmic reticulum were dilated in Esr1-KD compared with ERα replete Min6 β-cells. Increased expression of Oma1 and Chop was paralleled by increased oxygen consumption and apoptosis susceptibility in ERα-KD cells. In contrast, ERα overexpression and ligand activation reduced both Chop and Oma1 expression, likely by ERα binding to consensus estrogen-response element sites in the Oma1 and Chop promoters. Together, our findings suggest that ERα promotes β-cell survival and insulin secretion through maintenance of mitochondrial fission/fusion-mitophagy dynamics and EndoRetic function, in part by Oma1 and Chop repression.
Collapse
Affiliation(s)
- Zhenqi Zhou
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, Los Angeles, California 90095
| | - Vicent Ribas
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, Los Angeles, California 90095
| | - Prashant Rajbhandari
- Department of Pathology and Laboratory Medicine and the Howard Hughes Research Institute, Los Angeles, California 90095
| | - Brian G Drew
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, Los Angeles, California 90095
| | - Timothy M Moore
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, Los Angeles, California 90095
| | - Amy H Fluitt
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, Los Angeles, California 90095
| | - Britany R Reddish
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, Los Angeles, California 90095
| | - Kate A Whitney
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, Los Angeles, California 90095
| | - Senta Georgia
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, Los Angeles, California 90095
| | - Laurent Vergnes
- Departments of Human Genetics, Los Angeles, California 90095
| | - Karen Reue
- Departments of Human Genetics, Los Angeles, California 90095
| | - Marc Liesa
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, Los Angeles, California 90095
| | - Orian Shirihai
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, Los Angeles, California 90095
| | | | - Nai-Wen Chi
- Department of Medicine, University of California, San Diego, La Jolla, California 92037
| | - Sushil K Mahata
- Department of Medicine, University of California, San Diego, La Jolla, California 92037; Veterans Affairs San Diego Healthcare System, San Diego, California 92161
| | - Joseph P Tiano
- Department of Medicine and Pharmacology, Tulane University Health Sciences Center, New Orleans, Louisiana 70112
| | - Sylvia C Hewitt
- Receptor Biology Section, NIEHS, National Institutes of Health, Research Triangle Park, North Carolina 27709
| | - Peter Tontonoz
- Department of Pathology and Laboratory Medicine and the Howard Hughes Research Institute, Los Angeles, California 90095
| | - Kenneth S Korach
- Receptor Biology Section, NIEHS, National Institutes of Health, Research Triangle Park, North Carolina 27709
| | - Franck Mauvais-Jarvis
- Department of Medicine and Pharmacology, Tulane University Health Sciences Center, New Orleans, Louisiana 70112
| | - Andrea L Hevener
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, Los Angeles, California 90095; Iris Cantor UCLA Women's Health Research Center, Los Angeles, California 90095.
| |
Collapse
|
29
|
Involvement of estrogen receptors in silibinin protection of pancreatic β-cells from TNFα- or IL-1β-induced cytotoxicity. Biomed Pharmacother 2018; 102:344-353. [PMID: 29571019 DOI: 10.1016/j.biopha.2018.01.128] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 01/26/2018] [Accepted: 01/28/2018] [Indexed: 01/09/2023] Open
Abstract
Silibinin is a polyphenolic flavonoid that exhibits anticarcinogenic, anti-inflammatory and cytoprotective effects. The effect of silibinin on pancreatic islet β-cell is yet largely unknown in spite of well documented-hepatoprotective effects. Protecting the functional insulin-producing β-cells in the pancreas is a major therapeutic challenge in the patients with type 1 (T1DM) or type 2 diabetes mellitus (T2DM). This study reports the effect of silibinin on the rat pancreatic β-cell line, INS-1, damaged with pro-inflammatory cytokine, TNFα or IL-1β. Exposure to TNFα or IL-1β for 48 h caused INS-1 cells to reduce the production of insulin as well as cell viability. These actions of TNFα or IL-1β are associated with suppression of the expression of estrogen receptors (ERs). Further study revealed that silibinin protected the suppression in the expression of both ERα and ERβ that were involved in insulin synthesis and release, respectively. Furthermore, evidence is obtained that silibinin may impede the loss of critical INS-1 cells by promoting autophagy and preventing apoptosis. Direct cytoprotective effects of silibinin on INS-1 cells suggest that silibinin may be therapeutically beneficial for diabetes.
Collapse
|
30
|
Wang T, Liu Y, Huang C, Mansai HAA, Wei W, Zhang X, Li X, Liu S, Yang S. Puerarin promotes MIN6 cell survival by reducing cellular reactive oxygen species. Mol Med Rep 2018; 17:7281-7286. [PMID: 29568901 DOI: 10.3892/mmr.2018.8731] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Accepted: 07/25/2017] [Indexed: 11/05/2022] Open
Abstract
Type 1 diabetes is caused by destruction of the pancreatic β‑cells and, to date, no cure has been developed. Promoting the survival of pancreatic β‑cells may be beneficial for patients with type 1 diabetes. Puerarin is an estrogen analogue that been demonstrated in previous studies to be able to decreased blood glucose in patients with type 1 diabetes. Similar results were demonstrated in previous studies which additionally demonstrated that puerarin was able to decreased blood glucose in type 1 diabetic mice by protecting pancreatic β‑cells. However, the mechanism underlying the function of puerarin in pancreatic β‑cells remains unclear. Therefore, the present study sought to investigate the detailed function of puerarin in pancreatic β‑cells. In the present study, H2O2 was used to induce apoptosis. It was observed that puerarin significantly decreased H2O2‑induced apoptosis in mouse insulinoma MIN6 cells. It was additionally observed that puerarin decreased the levels of intracellular reactive oxygen species and mitochondrial superoxide in MIN6 cells. The protective effect of puerarin was markedly decreased by 6‑aminonicotinamide, an inhibitor of glucose‑6‑phosphate dehydrogenase (G6PD). In conclusion, the results of the present study suggested that puerarin may increase the activity of G6PD, decreased the level of oxidative stress in MIN6 cells, protect mitochondria and promote MIN6 cell survival. Investigating the mechanism underlying the effect of puerarin in MIN6 cells may provide a novel approach for development of a cure for type 1 diabetes.
Collapse
Affiliation(s)
- Tianxi Wang
- Xiamen Diabetes Institute, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, P.R. China
| | - Yijie Liu
- Division of Gastroenterology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, P.R. China
| | - Caoxin Huang
- Xiamen Diabetes Institute, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, P.R. China
| | - Hussen Amir Ahmed Mansai
- Xiamen Diabetes Institute, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, P.R. China
| | - Wenjing Wei
- Xiamen Diabetes Institute, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, P.R. China
| | - Xiaofang Zhang
- Xiamen Diabetes Institute, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, P.R. China
| | - Xuejun Li
- Xiamen Diabetes Institute, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, P.R. China
| | - Suhuan Liu
- Xiamen Diabetes Institute, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, P.R. China
| | - Shuyu Yang
- Xiamen Diabetes Institute, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, P.R. China
| |
Collapse
|
31
|
Sharma G, Mauvais-Jarvis F, Prossnitz ER. Roles of G protein-coupled estrogen receptor GPER in metabolic regulation. J Steroid Biochem Mol Biol 2018; 176:31-37. [PMID: 28223150 PMCID: PMC5563497 DOI: 10.1016/j.jsbmb.2017.02.012] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 02/13/2017] [Accepted: 02/17/2017] [Indexed: 12/19/2022]
Abstract
Metabolic homeostasis is differentially regulated in males and females. The lower incidence of obesity and associated diseases in pre-menopausal females points towards the beneficial role of the predominant estrogen, 17β-estradiol (E2). The actions of E2 are elicited by nuclear and extra-nuclear estrogen receptor (ER) α and ERβ, as well as the G protein-coupled estrogen receptor (GPER, previously termed GPR30). The roles of GPER in the regulation of metabolism are only beginning to emerge and much remains unclear. The present review highlights recent advances implicating the importance of GPER in metabolic regulation. Assessment of the specific metabolic roles of GPER employing GPER-deficient mice and highly selective GPER-targeted pharmacological agents, agonist G-1 and antagonists G-15 and G36, is also presented. Evidence from in vitro and in vivo studies involving either GPER deficiency or selective activation suggests that GPER is involved in body weight regulation, glucose and lipid homeostasis as well as inflammation. The therapeutic potential of activating GPER signaling through selective ligands for the treatment of obesity and diabetes is also discussed.
Collapse
Affiliation(s)
- Geetanjali Sharma
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, 87131, United States
| | - Franck Mauvais-Jarvis
- Diabetes Discovery and Gender Medicine Laboratory, Section of Endocrinology and Metabolism, Department of Medicine,Tulane University Health Sciences Center, School of Medicine, New Orleans, LA, 70112, United States
| | - Eric R Prossnitz
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, 87131, United States; University of New Mexico Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, United States.
| |
Collapse
|
32
|
Mauvais-Jarvis F. Are estrogens promoting immune modulation and islet protection in type 1 diabetes? J Diabetes Complications 2017; 31:1563-1564. [PMID: 28890307 DOI: 10.1016/j.jdiacomp.2017.07.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 07/26/2017] [Indexed: 12/24/2022]
Affiliation(s)
- Franck Mauvais-Jarvis
- Section of Endocrinology & Metabolism, Department of Medicine, Tulane University Health Sciences Center, New Orleans, LA 70112, USA.
| |
Collapse
|
33
|
Huang G, Xu J, Lefever DE, Glenn TC, Nagy T, Guo TL. Genistein prevention of hyperglycemia and improvement of glucose tolerance in adult non-obese diabetic mice are associated with alterations of gut microbiome and immune homeostasis. Toxicol Appl Pharmacol 2017; 332:138-148. [PMID: 28412308 PMCID: PMC5592136 DOI: 10.1016/j.taap.2017.04.009] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 03/25/2017] [Accepted: 04/10/2017] [Indexed: 12/17/2022]
Abstract
Although studies have linked soy phytoestrogen 4,7,4-trihydroxyisoflavone genistein (GEN) to reduced type 1 diabetes (T1D) risk, the mechanism of dietary GEN on T1D remains unknown. In our studies, adult non-obese diabetic (NOD) mouse model was employed to investigate the effects of GEN exposure on blood glucose level (BGL), glucose tolerance, gut microbiome, and immune responses. Adult male and female NOD mice were fed with either soy-based or casein-based diet, and received GEN at 20mg/kg body weight by gavage daily. The BGL and immune responses (represented by serum antibodies, cytokines and chemokines, and histopathology) were monitored, while the fecal gut microbiome was sequenced for 16S ribosomal RNA to reveal any alterations in gut microbial communities. A significantly reduced BGL was found in NOD males fed with soy-based diet on day 98 after initial dosing, and an improved glucose tolerance was observed on both diets. In addition, an anti-inflammatory response (suggested by reduced IgG2b and cytokine/chemokine levels, and alterations in the microbial taxonomy) was accompanied by an altered β-diversity in gut microbial species. Among the NOD females exposed to GEN, a later onset of T1D was observed. However, the profiles of gut microbiome, antibodies and cytokines/chemokines were all indicative of pro-inflammation. This study demonstrated an association among GEN exposure, gut microbiome alteration, and immune homeostasis in NOD males. Although the mechanisms underlying the protective effects of GEN in NOD mice need to be explored further, the current study suggested a GEN-induced sex-specific effect in inflammatory status and gut microbiome.
Collapse
Affiliation(s)
- Guannan Huang
- Department of Environmental Health Sciences, College of Public Health, United States
| | - Joella Xu
- Department of Veterinary Biosciences and Diagnostic Imaging, College of Veterinary Medicine, United States
| | - Daniel E Lefever
- Department of Veterinary Biosciences and Diagnostic Imaging, College of Veterinary Medicine, United States
| | - Travis C Glenn
- Department of Environmental Health Sciences, College of Public Health, United States
| | - Tamas Nagy
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, United States
| | - Tai L Guo
- Department of Veterinary Biosciences and Diagnostic Imaging, College of Veterinary Medicine, United States.
| |
Collapse
|
34
|
Mauvais-Jarvis F, Manson JE, Stevenson JC, Fonseca VA. Menopausal Hormone Therapy and Type 2 Diabetes Prevention: Evidence, Mechanisms, and Clinical Implications. Endocr Rev 2017; 38:173-188. [PMID: 28323934 PMCID: PMC5460681 DOI: 10.1210/er.2016-1146] [Citation(s) in RCA: 181] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Accepted: 03/02/2017] [Indexed: 12/14/2022]
Abstract
Type 2 diabetes has reached epidemic proportions in the United States. Large, randomized controlled trials suggest that menopausal hormone therapy (MHT) delays the onset of type 2 diabetes in women. However, the mechanisms and clinical implications of this association are still a matter of controversy. This review provides an up-to-date analysis and integration of epidemiological, clinical, and basic studies, and proposes a mechanistic explanation for the effect of menopause and MHT on type 2 diabetes development and prevention. We discuss the beneficial effects of endogenous estradiol with respect to insulin secretion, insulin sensitivity, and glucose effectiveness; we also discuss energy expenditure and adipose distribution, both of which are affected by menopause and improved by MHT, which thereby decreases the incidence of type 2 diabetes. We reconcile differences among studies that investigated the effect of menopause and MHT formulations on type 2 diabetes. We argue that discrepancies arise from physiological differences in methods used to assess glucose homeostasis, ranging from clinical indices of insulin sensitivity to steady-state methods to assess insulin action. We also discuss the influence of the route of estrogen administration and the addition of progestogens. We conclude that, although MHT is neither approved nor appropriate for the prevention of type 2 diabetes due to its complex balance of risks and benefits, it should not be withheld from women with increased risk of type 2 diabetes who seek treatment for menopausal symptoms.
Collapse
Affiliation(s)
- Franck Mauvais-Jarvis
- Department of Medicine, Division of Endocrinology and Metabolism, School of Medicine, Tulane University Health Sciences Center, New Orleans, Louisiana 70112
| | - JoAnn E Manson
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115.,Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, Massachusetts 02115
| | - John C Stevenson
- National Heart and Lung Institute, Imperial College London, Royal Brompton Hospital, London SW3 6NP, United Kingdom
| | - Vivian A Fonseca
- Department of Medicine, Division of Endocrinology and Metabolism, School of Medicine, Tulane University Health Sciences Center, New Orleans, Louisiana 70112
| |
Collapse
|
35
|
GPER-novel membrane oestrogen receptor. Clin Sci (Lond) 2017; 130:1005-16. [PMID: 27154744 DOI: 10.1042/cs20160114] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 03/02/2016] [Indexed: 12/11/2022]
Abstract
The recent discovery of the G protein-coupled oestrogen receptor (GPER) presents new challenges and opportunities for understanding the physiology, pathophysiology and pharmacology of many diseases. This review will focus on the expression and function of GPER in hypertension, kidney disease, atherosclerosis, vascular remodelling, heart failure, reproduction, metabolic disorders, cancer, environmental health and menopause. Furthermore, this review will highlight the potential of GPER as a therapeutic target.
Collapse
|
36
|
Iuamoto LR, Franco AS, Suguita FY, Essu FF, Oliveira LT, Kato JM, Torsani MB, Meyer A, Andraus W, Chaib E, D'Albuquerque LAC. Human islet xenotransplantation in rodents: A literature review of experimental model trends. Clinics (Sao Paulo) 2017; 72:238-243. [PMID: 28492724 PMCID: PMC5401612 DOI: 10.6061/clinics/2017(04)08] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 12/16/2016] [Indexed: 01/19/2023] Open
Abstract
Among the innovations for the treatment of type 1 diabetes, islet transplantation is a less invasive method of treatment, although it is still in development. One of the greatest barriers to this technique is the low number of pancreas donors and the low number of pancreases that are available for transplantation. Rodent models have been chosen in most studies of islet rejection and type 1 diabetes prevention to evaluate the quality and function of isolated human islets and to identify alternative solutions to the problem of islet scarcity. The purpose of this study is to conduct a review of islet xenotransplantation experiments from humans to rodents, to organize and analyze the parameters of these experiments, to describe trends in experimental modeling and to assess the viability of this procedure. In this study, we reviewed recently published research regarding islet xenotransplantation from humans to rodents, and we summarized the findings and organized the relevant data. The included studies were recent reports that involved xenotransplantation using human islets in a rodent model. We excluded the studies that related to isotransplantation, autotransplantation and allotransplantation. A total of 34 studies that related to xenotransplantation were selected for review based on their relevance and current data. Advances in the use of different graft sites may overcome autoimmunity and rejection after transplantation, which may solve the problem of the scarcity of islet donors in patients with type 1 diabetes.
Collapse
Affiliation(s)
- Leandro Ryuchi Iuamoto
- Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
- *Corresponding author. E-mail:
| | | | | | | | | | | | | | - Alberto Meyer
- Departamento de Gastroenterologia, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Wellington Andraus
- Departamento de Gastroenterologia, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Eleazar Chaib
- Departamento de Gastroenterologia, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | | |
Collapse
|
37
|
Mauvais-Jarvis F. Menopause, Estrogens, and Glucose Homeostasis in Women. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1043:217-225. [DOI: 10.1007/978-3-319-70178-3_11] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
38
|
Mauvais-Jarvis F. Role of Sex Steroids in β Cell Function, Growth, and Survival. Trends Endocrinol Metab 2016; 27:844-855. [PMID: 27640750 PMCID: PMC5116277 DOI: 10.1016/j.tem.2016.08.008] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 08/27/2016] [Accepted: 08/29/2016] [Indexed: 01/08/2023]
Abstract
The gonads have long been considered endocrine glands, producing sex steroids such as estrogens, androgens, and progesterone (P4) for the sole purpose of sexual differentiation, puberty, and reproduction. Reproduction and energy metabolism are tightly linked, however, and gonadal steroids play an important role in sex-specific aspects of energy metabolism in various physiological conditions. In that respect, gonadal steroids also influence the secretion of insulin in a sex-specific manner. This review presents a perspective on the physiological roles of estrogens, androgens, and P4 via their receptors in pancreatic β cells in the gender-specific tuning of insulin secretion. I also discuss potential gender-specific therapeutic avenues that this knowledge may open in the future.
Collapse
Affiliation(s)
- Franck Mauvais-Jarvis
- Diabetes Discovery and Gender Medicine Laboratory, Section of Endocrinology and Metabolism, Department of Medicine, Tulane University Health Sciences Center, School of Medicine, New Orleans, LA, USA.
| |
Collapse
|
39
|
Karakose M, Pinarli FA, Arslan MS, Boyuk G, Boztok B, Albayrak A, Ulus AT, Cakal E, Delibasi T. Comparison of the Ovary and Kidney as Sites for Islet Transplantation in Diabetic Rats. Transplant Proc 2016; 48:2216-20. [PMID: 27569973 DOI: 10.1016/j.transproceed.2016.04.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 03/30/2016] [Accepted: 04/25/2016] [Indexed: 11/29/2022]
Abstract
BACKGROUND Currently, the most commonly used site for clinical islet transplantation is the liver although it is far from being an ideal site. Low oxygen tension and the induction of an inflammatory response impair islet implantation and lead to significant early loss of islet. The present study aimed to investigate and compare the efficacy of islet transplantation to the ovary and kidney subcapsule in diabetic rats. METHODS The study was performed with 3 groups of rats (control, ovary, and kidney subcapsule) including 6 Sprague female rats each. Diabetes model was created with the use of streptozotocin, and blood glucose levels of the rats were measured after 72 hours. Thirty days after the transplantation, blood samples were obtained from the rats, and then pancreas, kidney, and ovary specimens were fixed in 10% formaldehyde and the experiment completed. After staining with hematoxylin and eosin, the tissue samples were morphologically evaluated by a specialist histopathologist. RESULTS Changes in mean blood glucose and C-peptide levels were statistically significant in the ovary and kidney subcapsule groups. Histologic examination revealed that granulosus insulin-bearing cells were detected in the islet grafts of both ovary and kidney subcapsule groups. The renal subcapsule group had inflammation signs on histologic examination. The islet cells of both ovary and renal subcapsule groups had no vacuolization. CONCLUSIONS We showed that the ovary might be a new site for islet transplantation. Further research should be done on whether the initial results of this study can be reproduced in larger numbers of animal models and eventually in humans.
Collapse
Affiliation(s)
- M Karakose
- Department of Endocrinology and Metabolism, Diskapi Yildirim Beyazit Training and Research Hospital, Ankara, Turkey.
| | - F A Pinarli
- Department of Genetic and Medical Research Center, Diskapi Yildirim Beyazit Training and Research Hospital, Ankara, Turkey
| | - M S Arslan
- Department of Endocrinology and Metabolism, Diskapi Yildirim Beyazit Training and Research Hospital, Ankara, Turkey
| | - G Boyuk
- Adacell Laboratory, Pancreas Islet Cell Research Center, Diskapi Yildirim Beyazit Training and Research Hospital, Ankara, Turkey
| | - B Boztok
- Adacell Laboratory, Pancreas Islet Cell Research Center, Diskapi Yildirim Beyazit Training and Research Hospital, Ankara, Turkey
| | - A Albayrak
- Department of Pathology, Diskapi Yildirim Beyazit Training and Research Hospital, Ankara, Turkey
| | - A T Ulus
- Department of Cardiovascular Surgery, Hacettepe University School of Medicine, Ankara, Turkey
| | - E Cakal
- Department of Endocrinology and Metabolism, Diskapi Yildirim Beyazit Training and Research Hospital, Ankara, Turkey
| | - T Delibasi
- Department of Internal Medicine, Kastamonu University School of Medicine, Kastamonu, Turkey
| |
Collapse
|
40
|
Meyer MR, Barton M. Estrogens and Coronary Artery Disease: New Clinical Perspectives. ADVANCES IN PHARMACOLOGY 2016; 77:307-60. [PMID: 27451102 DOI: 10.1016/bs.apha.2016.05.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In premenopausal women, endogenous estrogens are associated with reduced prevalence of arterial hypertension, coronary artery disease, myocardial infarction, and stroke. Clinical trials conducted in the 1990s such as HERS, WHI, and WISDOM have shown that postmenopausal treatment with horse hormone mixtures (so-called conjugated equine estrogens) and synthetic progestins adversely affects female cardiovascular health. Our understanding of rapid (nongenomic) and chronic (genomic) estrogen signaling has since advanced considerably, including identification of a new G protein-coupled estrogen receptor (GPER), which like the "classical" receptors ERα and ERβ is highly abundant in the cardiovascular system. Here, we discuss the role of estrogen receptors in the pathogenesis of coronary artery disease and review natural and synthetic ligands of estrogen receptors as well as their effects in physiology, on cardiovascular risk factors, and atherosclerotic vascular disease. Data from preclinical and clinical studies using nonselective compounds activating GPER, which include selective estrogen receptor modulators such as tamoxifen or raloxifene, selective estrogen receptor downregulators such as Faslodex™ (fulvestrant/ICI 182,780), vitamin B3 (niacin), green tea catechins, and soy flavonoids such as genistein or resveratrol, strongly suggest that activation of GPER may afford therapeutic benefit for primary and secondary prevention in patients with or at risk for coronary artery disease. Evidence from preclinical studies suggest similar efficacy profiles for selective small molecule GPER agonists such as G-1 which are devoid of uterotrophic activity. Further clinical research in this area is warranted to provide opportunities for future cardiovascular drug development.
Collapse
Affiliation(s)
- M R Meyer
- Triemli City Hospital, Zürich, Switzerland.
| | - M Barton
- Molecular Internal Medicine, University of Zürich, Zürich, Switzerland.
| |
Collapse
|
41
|
Gourdy P, Bourgeois EA, Levescot A, Pham L, Riant E, Ahui ML, Damotte D, Gombert JM, Bayard F, Ohlsson C, Arnal JF, Herbelin A. Estrogen Therapy Delays Autoimmune Diabetes and Promotes the Protective Efficiency of Natural Killer T-Cell Activation in Female Nonobese Diabetic Mice. Endocrinology 2016; 157:258-67. [PMID: 26485613 DOI: 10.1210/en.2015-1313] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Therapeutic strategies focused on restoring immune tolerance remain the main avenue to prevent type 1 diabetes (T1D). Because estrogens potentiate FoxP3+ regulatory T cells (Treg) and invariant natural killer T (iNKT) cells, two regulatory lymphocyte populations that are functionally deficient in nonobese diabetic (NOD) mice, we investigated whether estradiol (E2) therapy influences the course of T1D in this model. To this end, female NOD mice were sc implanted with E2- or placebo-delivering pellets to explore the course of spontaneous and cyclophosphamide-induced diabetes. Treg-depleted and iNKT-cell-deficient (Jα18(-/-)) NOD mice were used to assess the respective involvement of these lymphocyte populations in E2 effects. Early E2 administration (from 4 wk of age) was found to preserve NOD mice from both spontaneous and cyclophosphamide-induced diabetes, and a complete protection was also observed throughout treatment when E2 treatment was initiated after the onset of insulitis (from 12 wk of age). This delayed E2 treatment remained fully effective in Treg-depleted mice but failed to entirely protect Jα18(-/-) mice. Accordingly, E2 administration was shown to restore the cytokine production of iNKT cells in response to in vivo challenge with the cognate ligand α-galactosylceramide. Finally, transient E2 administration potentiated the previously described protective action of α-galactosylceramide treatment in NOD females. This study provides original evidence that E2 therapy strongly protects NOD mice from T1D and reveals the estrogen/iNKT cell axis as a new effective target to counteract diabetes onset at the stage of insulitis. Estrogen-based therapy should thus be considered for T1D prevention.
Collapse
MESH Headings
- Animals
- Autoimmune Diseases/drug therapy
- Autoimmune Diseases/immunology
- Autoimmune Diseases/metabolism
- Autoimmune Diseases/prevention & control
- Cytokines/blood
- Cytokines/metabolism
- Diabetes Mellitus, Type 1/drug therapy
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/prevention & control
- Drug Implants
- Estradiol/administration & dosage
- Estradiol/therapeutic use
- Estrogen Replacement Therapy
- Estrogens/administration & dosage
- Estrogens/therapeutic use
- Female
- Galactosylceramides/agonists
- Galactosylceramides/pharmacology
- Galactosylceramides/therapeutic use
- Immune Tolerance/drug effects
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Lymphocyte Activation/drug effects
- Lymphocyte Depletion/adverse effects
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Mice, Mutant Strains
- Ovariectomy/adverse effects
- Prediabetic State/drug therapy
- Prediabetic State/immunology
- Prediabetic State/metabolism
- Prediabetic State/prevention & control
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
Collapse
Affiliation(s)
- Pierre Gourdy
- INSERM Unité 1048 (P.G., E.R., F.B., J.-F.A.), Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; Toulouse University (P.G., J.-F.A.), 31059 Toulouse, France; Department of Diabetology (P.G.), Toulouse University Hospital, 31403 Toulouse, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 8147 (E.A.B., L.P., M.-L.A.), Necker Hospital, 75015 Paris, France; Paris Descartes University, Necker Hospital (E.A.B., L.P., M.-L.A., A.H.), 75014 Paris, France; INSERM Unité 1082 (A.L., A.H.), 86022 Poitiers, France; Paris-Sud-11 University (A.L.), 91405 Orsay, France; Department of Anatomy and Cytology (A.L., D.D.), Hôtel Dieu, 49033 Paris, France; Laboratory of Immunology (J.-M.G.), Poitiers, and Poitiers University (J.-M.G., A.H.), 86000 Poitiers, France; Centre Hospitalo-Universitaire de Poitiers (J.-M.G., A.H.), 86021 Poitiers, France; and Centre for Bone and Arthritis Research (C.O.), University of Gothenburg, S-405 30 Gothenburg, Sweden
| | - Elvire A Bourgeois
- INSERM Unité 1048 (P.G., E.R., F.B., J.-F.A.), Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; Toulouse University (P.G., J.-F.A.), 31059 Toulouse, France; Department of Diabetology (P.G.), Toulouse University Hospital, 31403 Toulouse, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 8147 (E.A.B., L.P., M.-L.A.), Necker Hospital, 75015 Paris, France; Paris Descartes University, Necker Hospital (E.A.B., L.P., M.-L.A., A.H.), 75014 Paris, France; INSERM Unité 1082 (A.L., A.H.), 86022 Poitiers, France; Paris-Sud-11 University (A.L.), 91405 Orsay, France; Department of Anatomy and Cytology (A.L., D.D.), Hôtel Dieu, 49033 Paris, France; Laboratory of Immunology (J.-M.G.), Poitiers, and Poitiers University (J.-M.G., A.H.), 86000 Poitiers, France; Centre Hospitalo-Universitaire de Poitiers (J.-M.G., A.H.), 86021 Poitiers, France; and Centre for Bone and Arthritis Research (C.O.), University of Gothenburg, S-405 30 Gothenburg, Sweden
| | - Anaïs Levescot
- INSERM Unité 1048 (P.G., E.R., F.B., J.-F.A.), Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; Toulouse University (P.G., J.-F.A.), 31059 Toulouse, France; Department of Diabetology (P.G.), Toulouse University Hospital, 31403 Toulouse, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 8147 (E.A.B., L.P., M.-L.A.), Necker Hospital, 75015 Paris, France; Paris Descartes University, Necker Hospital (E.A.B., L.P., M.-L.A., A.H.), 75014 Paris, France; INSERM Unité 1082 (A.L., A.H.), 86022 Poitiers, France; Paris-Sud-11 University (A.L.), 91405 Orsay, France; Department of Anatomy and Cytology (A.L., D.D.), Hôtel Dieu, 49033 Paris, France; Laboratory of Immunology (J.-M.G.), Poitiers, and Poitiers University (J.-M.G., A.H.), 86000 Poitiers, France; Centre Hospitalo-Universitaire de Poitiers (J.-M.G., A.H.), 86021 Poitiers, France; and Centre for Bone and Arthritis Research (C.O.), University of Gothenburg, S-405 30 Gothenburg, Sweden
| | - Linh Pham
- INSERM Unité 1048 (P.G., E.R., F.B., J.-F.A.), Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; Toulouse University (P.G., J.-F.A.), 31059 Toulouse, France; Department of Diabetology (P.G.), Toulouse University Hospital, 31403 Toulouse, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 8147 (E.A.B., L.P., M.-L.A.), Necker Hospital, 75015 Paris, France; Paris Descartes University, Necker Hospital (E.A.B., L.P., M.-L.A., A.H.), 75014 Paris, France; INSERM Unité 1082 (A.L., A.H.), 86022 Poitiers, France; Paris-Sud-11 University (A.L.), 91405 Orsay, France; Department of Anatomy and Cytology (A.L., D.D.), Hôtel Dieu, 49033 Paris, France; Laboratory of Immunology (J.-M.G.), Poitiers, and Poitiers University (J.-M.G., A.H.), 86000 Poitiers, France; Centre Hospitalo-Universitaire de Poitiers (J.-M.G., A.H.), 86021 Poitiers, France; and Centre for Bone and Arthritis Research (C.O.), University of Gothenburg, S-405 30 Gothenburg, Sweden
| | - Elodie Riant
- INSERM Unité 1048 (P.G., E.R., F.B., J.-F.A.), Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; Toulouse University (P.G., J.-F.A.), 31059 Toulouse, France; Department of Diabetology (P.G.), Toulouse University Hospital, 31403 Toulouse, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 8147 (E.A.B., L.P., M.-L.A.), Necker Hospital, 75015 Paris, France; Paris Descartes University, Necker Hospital (E.A.B., L.P., M.-L.A., A.H.), 75014 Paris, France; INSERM Unité 1082 (A.L., A.H.), 86022 Poitiers, France; Paris-Sud-11 University (A.L.), 91405 Orsay, France; Department of Anatomy and Cytology (A.L., D.D.), Hôtel Dieu, 49033 Paris, France; Laboratory of Immunology (J.-M.G.), Poitiers, and Poitiers University (J.-M.G., A.H.), 86000 Poitiers, France; Centre Hospitalo-Universitaire de Poitiers (J.-M.G., A.H.), 86021 Poitiers, France; and Centre for Bone and Arthritis Research (C.O.), University of Gothenburg, S-405 30 Gothenburg, Sweden
| | - Marie-Louise Ahui
- INSERM Unité 1048 (P.G., E.R., F.B., J.-F.A.), Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; Toulouse University (P.G., J.-F.A.), 31059 Toulouse, France; Department of Diabetology (P.G.), Toulouse University Hospital, 31403 Toulouse, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 8147 (E.A.B., L.P., M.-L.A.), Necker Hospital, 75015 Paris, France; Paris Descartes University, Necker Hospital (E.A.B., L.P., M.-L.A., A.H.), 75014 Paris, France; INSERM Unité 1082 (A.L., A.H.), 86022 Poitiers, France; Paris-Sud-11 University (A.L.), 91405 Orsay, France; Department of Anatomy and Cytology (A.L., D.D.), Hôtel Dieu, 49033 Paris, France; Laboratory of Immunology (J.-M.G.), Poitiers, and Poitiers University (J.-M.G., A.H.), 86000 Poitiers, France; Centre Hospitalo-Universitaire de Poitiers (J.-M.G., A.H.), 86021 Poitiers, France; and Centre for Bone and Arthritis Research (C.O.), University of Gothenburg, S-405 30 Gothenburg, Sweden
| | - Diane Damotte
- INSERM Unité 1048 (P.G., E.R., F.B., J.-F.A.), Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; Toulouse University (P.G., J.-F.A.), 31059 Toulouse, France; Department of Diabetology (P.G.), Toulouse University Hospital, 31403 Toulouse, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 8147 (E.A.B., L.P., M.-L.A.), Necker Hospital, 75015 Paris, France; Paris Descartes University, Necker Hospital (E.A.B., L.P., M.-L.A., A.H.), 75014 Paris, France; INSERM Unité 1082 (A.L., A.H.), 86022 Poitiers, France; Paris-Sud-11 University (A.L.), 91405 Orsay, France; Department of Anatomy and Cytology (A.L., D.D.), Hôtel Dieu, 49033 Paris, France; Laboratory of Immunology (J.-M.G.), Poitiers, and Poitiers University (J.-M.G., A.H.), 86000 Poitiers, France; Centre Hospitalo-Universitaire de Poitiers (J.-M.G., A.H.), 86021 Poitiers, France; and Centre for Bone and Arthritis Research (C.O.), University of Gothenburg, S-405 30 Gothenburg, Sweden
| | - Jean-Marc Gombert
- INSERM Unité 1048 (P.G., E.R., F.B., J.-F.A.), Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; Toulouse University (P.G., J.-F.A.), 31059 Toulouse, France; Department of Diabetology (P.G.), Toulouse University Hospital, 31403 Toulouse, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 8147 (E.A.B., L.P., M.-L.A.), Necker Hospital, 75015 Paris, France; Paris Descartes University, Necker Hospital (E.A.B., L.P., M.-L.A., A.H.), 75014 Paris, France; INSERM Unité 1082 (A.L., A.H.), 86022 Poitiers, France; Paris-Sud-11 University (A.L.), 91405 Orsay, France; Department of Anatomy and Cytology (A.L., D.D.), Hôtel Dieu, 49033 Paris, France; Laboratory of Immunology (J.-M.G.), Poitiers, and Poitiers University (J.-M.G., A.H.), 86000 Poitiers, France; Centre Hospitalo-Universitaire de Poitiers (J.-M.G., A.H.), 86021 Poitiers, France; and Centre for Bone and Arthritis Research (C.O.), University of Gothenburg, S-405 30 Gothenburg, Sweden
| | - Francis Bayard
- INSERM Unité 1048 (P.G., E.R., F.B., J.-F.A.), Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; Toulouse University (P.G., J.-F.A.), 31059 Toulouse, France; Department of Diabetology (P.G.), Toulouse University Hospital, 31403 Toulouse, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 8147 (E.A.B., L.P., M.-L.A.), Necker Hospital, 75015 Paris, France; Paris Descartes University, Necker Hospital (E.A.B., L.P., M.-L.A., A.H.), 75014 Paris, France; INSERM Unité 1082 (A.L., A.H.), 86022 Poitiers, France; Paris-Sud-11 University (A.L.), 91405 Orsay, France; Department of Anatomy and Cytology (A.L., D.D.), Hôtel Dieu, 49033 Paris, France; Laboratory of Immunology (J.-M.G.), Poitiers, and Poitiers University (J.-M.G., A.H.), 86000 Poitiers, France; Centre Hospitalo-Universitaire de Poitiers (J.-M.G., A.H.), 86021 Poitiers, France; and Centre for Bone and Arthritis Research (C.O.), University of Gothenburg, S-405 30 Gothenburg, Sweden
| | - Claes Ohlsson
- INSERM Unité 1048 (P.G., E.R., F.B., J.-F.A.), Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; Toulouse University (P.G., J.-F.A.), 31059 Toulouse, France; Department of Diabetology (P.G.), Toulouse University Hospital, 31403 Toulouse, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 8147 (E.A.B., L.P., M.-L.A.), Necker Hospital, 75015 Paris, France; Paris Descartes University, Necker Hospital (E.A.B., L.P., M.-L.A., A.H.), 75014 Paris, France; INSERM Unité 1082 (A.L., A.H.), 86022 Poitiers, France; Paris-Sud-11 University (A.L.), 91405 Orsay, France; Department of Anatomy and Cytology (A.L., D.D.), Hôtel Dieu, 49033 Paris, France; Laboratory of Immunology (J.-M.G.), Poitiers, and Poitiers University (J.-M.G., A.H.), 86000 Poitiers, France; Centre Hospitalo-Universitaire de Poitiers (J.-M.G., A.H.), 86021 Poitiers, France; and Centre for Bone and Arthritis Research (C.O.), University of Gothenburg, S-405 30 Gothenburg, Sweden
| | - Jean-François Arnal
- INSERM Unité 1048 (P.G., E.R., F.B., J.-F.A.), Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; Toulouse University (P.G., J.-F.A.), 31059 Toulouse, France; Department of Diabetology (P.G.), Toulouse University Hospital, 31403 Toulouse, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 8147 (E.A.B., L.P., M.-L.A.), Necker Hospital, 75015 Paris, France; Paris Descartes University, Necker Hospital (E.A.B., L.P., M.-L.A., A.H.), 75014 Paris, France; INSERM Unité 1082 (A.L., A.H.), 86022 Poitiers, France; Paris-Sud-11 University (A.L.), 91405 Orsay, France; Department of Anatomy and Cytology (A.L., D.D.), Hôtel Dieu, 49033 Paris, France; Laboratory of Immunology (J.-M.G.), Poitiers, and Poitiers University (J.-M.G., A.H.), 86000 Poitiers, France; Centre Hospitalo-Universitaire de Poitiers (J.-M.G., A.H.), 86021 Poitiers, France; and Centre for Bone and Arthritis Research (C.O.), University of Gothenburg, S-405 30 Gothenburg, Sweden
| | - André Herbelin
- INSERM Unité 1048 (P.G., E.R., F.B., J.-F.A.), Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; Toulouse University (P.G., J.-F.A.), 31059 Toulouse, France; Department of Diabetology (P.G.), Toulouse University Hospital, 31403 Toulouse, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 8147 (E.A.B., L.P., M.-L.A.), Necker Hospital, 75015 Paris, France; Paris Descartes University, Necker Hospital (E.A.B., L.P., M.-L.A., A.H.), 75014 Paris, France; INSERM Unité 1082 (A.L., A.H.), 86022 Poitiers, France; Paris-Sud-11 University (A.L.), 91405 Orsay, France; Department of Anatomy and Cytology (A.L., D.D.), Hôtel Dieu, 49033 Paris, France; Laboratory of Immunology (J.-M.G.), Poitiers, and Poitiers University (J.-M.G., A.H.), 86000 Poitiers, France; Centre Hospitalo-Universitaire de Poitiers (J.-M.G., A.H.), 86021 Poitiers, France; and Centre for Bone and Arthritis Research (C.O.), University of Gothenburg, S-405 30 Gothenburg, Sweden
| |
Collapse
|
42
|
Yuchi Y, Cai Y, Legein B, De Groef S, Leuckx G, Coppens V, Van Overmeire E, Staels W, De Leu N, Martens G, Van Ginderachter JA, Heimberg H, Van de Casteele M. Estrogen Receptor α Regulates β-Cell Formation During Pancreas Development and Following Injury. Diabetes 2015; 64:3218-28. [PMID: 26015547 DOI: 10.2337/db14-1798] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 05/16/2015] [Indexed: 11/13/2022]
Abstract
Identifying pathways for β-cell generation is essential for cell therapy in diabetes. We investigated the potential of 17β-estradiol (E2) and estrogen receptor (ER) signaling for stimulating β-cell generation during embryonic development and in the severely injured adult pancreas. E2 concentration, ER activity, and number of ERα transcripts were enhanced in the pancreas injured by partial duct ligation (PDL) along with nuclear localization of ERα in β-cells. PDL-induced proliferation of β-cells depended on aromatase activity. The activation of Neurogenin3 (Ngn3) gene expression and β-cell growth in PDL pancreas were impaired when ERα was turned off chemically or genetically (ERα(-/-)), whereas in situ delivery of E2 promoted β-cell formation. In the embryonic pancreas, β-cell replication, number of Ngn3(+) progenitor cells, and expression of key transcription factors of the endocrine lineage were decreased by ERα inactivation. The current study reveals that E2 and ERα signaling can drive β-cell replication and formation in mouse pancreas.
Collapse
Affiliation(s)
- Yixing Yuchi
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Ying Cai
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Bart Legein
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Sofie De Groef
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Gunter Leuckx
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Violette Coppens
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Eva Van Overmeire
- Myeloid Cell Immunology Laboratory, Vlaams Instituut voor Biotechnologie, Brussels, Belgium Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Willem Staels
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium Department of Pediatrics, Division of Pediatric Endocrinology, Ghent University Hospital, and Department of Pediatrics and Medical Genetics, Ghent University, Ghent, Belgium
| | - Nico De Leu
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium Department of Endocrinology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Geert Martens
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Jo A Van Ginderachter
- Myeloid Cell Immunology Laboratory, Vlaams Instituut voor Biotechnologie, Brussels, Belgium Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Harry Heimberg
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | | |
Collapse
|
43
|
Prossnitz ER, Hathaway HJ. What have we learned about GPER function in physiology and disease from knockout mice? J Steroid Biochem Mol Biol 2015; 153:114-26. [PMID: 26189910 PMCID: PMC4568147 DOI: 10.1016/j.jsbmb.2015.06.014] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 06/24/2015] [Accepted: 06/25/2015] [Indexed: 12/16/2022]
Abstract
Estrogens, predominantly 17β-estradiol, exert diverse effects throughout the body in both normal and pathophysiology, during development and in reproductive, metabolic, endocrine, cardiovascular, nervous, musculoskeletal and immune systems. Estrogen and its receptors also play important roles in carcinogenesis and therapy, particularly for breast cancer. In addition to the classical nuclear estrogen receptors (ERα and ERβ) that traditionally mediate predominantly genomic signaling, the G protein-coupled estrogen receptor GPER has become recognized as a critical mediator of rapid signaling in response to estrogen. Mouse models, and in particular knockout (KO) mice, represent an important approach to understand the functions of receptors in normal physiology and disease. Whereas ERα KO mice display multiple significant defects in reproduction and mammary gland development, ERβ KO phenotypes are more limited, and GPER KO exhibit no reproductive deficits. However, the study of GPER KO mice over the last six years has revealed that GPER deficiency results in multiple physiological alterations including obesity, cardiovascular dysfunction, insulin resistance and glucose intolerance. In addition, the lack of estrogen-mediated effects in numerous tissues of GPER KO mice, studied in vivo or ex vivo, including those of the cardiovascular, endocrine, nervous and immune systems, reveals GPER as a genuine mediator of estrogen action. Importantly, GPER KO mice have also demonstrated roles for GPER in breast carcinogenesis and metastasis. In combination with the supporting effects of GPER-selective ligands and GPER knockdown approaches, GPER KO mice demonstrate the therapeutic potential of targeting GPER activity in diseases as diverse as obesity, diabetes, multiple sclerosis, hypertension, atherosclerosis, myocardial infarction, stroke and cancer.
Collapse
Affiliation(s)
- Eric R Prossnitz
- Department of Internal Medicine, University of New Mexico, Albuquerque, NM 87131, United States; University of New Mexico Cancer Center, Albuquerque, NM 87131, United States.
| | - Helen J Hathaway
- Department of Cell Biology & Physiology, University of New Mexico, Albuquerque, NM 87131, United States; University of New Mexico Cancer Center, Albuquerque, NM 87131, United States.
| |
Collapse
|
44
|
Prossnitz ER, Arterburn JB. International Union of Basic and Clinical Pharmacology. XCVII. G Protein-Coupled Estrogen Receptor and Its Pharmacologic Modulators. Pharmacol Rev 2015; 67:505-40. [PMID: 26023144 PMCID: PMC4485017 DOI: 10.1124/pr.114.009712] [Citation(s) in RCA: 193] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Estrogens are critical mediators of multiple and diverse physiologic effects throughout the body in both sexes, including the reproductive, cardiovascular, endocrine, nervous, and immune systems. As such, alterations in estrogen function play important roles in many diseases and pathophysiological conditions (including cancer), exemplified by the lower prevalence of many diseases in premenopausal women. Estrogens mediate their effects through multiple cellular receptors, including the nuclear receptor family (ERα and ERβ) and the G protein-coupled receptor (GPCR) family (GPR30/G protein-coupled estrogen receptor [GPER]). Although both receptor families can initiate rapid cell signaling and transcriptional regulation, the nuclear receptors are traditionally associated with regulating gene expression, whereas GPCRs are recognized as mediating rapid cellular signaling. Estrogen-activated pathways are not only the target of multiple therapeutic agents (e.g., tamoxifen, fulvestrant, raloxifene, and aromatase inhibitors) but are also affected by a plethora of phyto- and xeno-estrogens (e.g., genistein, coumestrol, bisphenol A, dichlorodiphenyltrichloroethane). Because of the existence of multiple estrogen receptors with overlapping ligand specificities, expression patterns, and signaling pathways, the roles of the individual receptors with respect to the diverse array of endogenous and exogenous ligands have been challenging to ascertain. The identification of GPER-selective ligands however has led to a much greater understanding of the roles of this receptor in normal physiology and disease as well as its interactions with the classic estrogen receptors ERα and ERβ and their signaling pathways. In this review, we describe the history and characterization of GPER over the past 15 years focusing on the pharmacology of steroidal and nonsteroidal compounds that have been employed to unravel the biology of this most recently recognized estrogen receptor.
Collapse
Affiliation(s)
- Eric R Prossnitz
- Department of Internal Medicine (E.R.P.) and University of New Mexico Cancer Center (E.R.P., J.B.A.), The University of New Mexico Health Sciences Center, Albuquerque, New Mexico; and Department of Chemistry and Biochemistry, New Mexico State University, Las Cruces, New Mexico (J.B.A.)
| | - Jeffrey B Arterburn
- Department of Internal Medicine (E.R.P.) and University of New Mexico Cancer Center (E.R.P., J.B.A.), The University of New Mexico Health Sciences Center, Albuquerque, New Mexico; and Department of Chemistry and Biochemistry, New Mexico State University, Las Cruces, New Mexico (J.B.A.)
| |
Collapse
|
45
|
Tiano JP, Tate CR, Yang BS, DiMarchi R, Mauvais-Jarvis F. Effect of targeted estrogen delivery using glucagon-like peptide-1 on insulin secretion, insulin sensitivity and glucose homeostasis. Sci Rep 2015; 5:10211. [PMID: 25970118 PMCID: PMC4429560 DOI: 10.1038/srep10211] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 04/07/2015] [Indexed: 12/21/2022] Open
Abstract
The female estrogen 17β-estradiol (E2) enhances pancreatic β-cell function via estrogen receptors (ERs). However, the risk of hormone dependent cancer precludes the use of general estrogen therapy as a chronic treatment for diabetes. To target E2 to β-cells without the undesirable effects of general estrogen therapy, we created fusion peptides combining active or inactive glucagon-like peptide-1 (GLP-1) and E2 in a single molecule (aGLP1-E2 and iGLP1-E2 respectively). By combining the activities of GLP-1 and E2, we envisioned synergistic insulinotropic activities of these molecules on β-cells. In cultured human islets and in C57BL/6 mice, both aGLP1 and aGLP1-E2 enhanced glucose-stimulated insulin secretion (GSIS) compared to vehicle and iGLP1-E2 without superior efficacy of aGLP1-E2 compared to GLP-1 alone. However, aGLP1-E2 decreased fasting and fed blood glucose to a greater extent than aGLP1 and iGLP1-E2 alone. Further, aGLP1-E2 exhibited improved insulin sensitivity compared to aGLP1 and iGLP1-E2 alone (HOMA-IR and insulin tolerance test). In conclusion, targeted estrogen delivery to non-diabetic islets in the presence of GLP-1 does not enhance GSIS. However, combining GLP-1 to estrogen delivers additional efficacy relative to GLP-1 alone on insulin sensitivity and glucose homeostasis in non-diabetic mice.
Collapse
Affiliation(s)
- Joseph P Tiano
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University School of Medicine, Chicago, IL 60611
| | - Chandra R Tate
- Division of Endocrinology &Metabolism, Department of Medicine, Tulane University Health Sciences Center, School of Medicine, New Orleans, LA 70112
| | - Bin S Yang
- Department of Chemistry, Indiana University, Bloomington, IN, 47405
| | - Richard DiMarchi
- Department of Chemistry, Indiana University, Bloomington, IN, 47405
| | - Franck Mauvais-Jarvis
- 1] Division of Endocrinology &Metabolism, Department of Medicine, Tulane University Health Sciences Center, School of Medicine, New Orleans, LA 70112 [2] Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University School of Medicine, Chicago, IL 60611
| |
Collapse
|
46
|
Barton M, Prossnitz ER. Emerging roles of GPER in diabetes and atherosclerosis. Trends Endocrinol Metab 2015; 26:185-92. [PMID: 25767029 PMCID: PMC4731095 DOI: 10.1016/j.tem.2015.02.003] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 01/31/2015] [Accepted: 02/04/2015] [Indexed: 01/13/2023]
Abstract
The G protein-coupled estrogen receptor (GPER) is a 7-transmembrane receptor implicated in rapid estrogen signaling. Originally cloned from vascular endothelial cells, GPER plays a central role in the regulation of vascular tone and cell growth as well as lipid and glucose homeostasis. This review highlights our knowledge of the physiological and pathophysiological functions of GPER in the pancreas, peripheral and immune tissues, and the arterial vasculature. Recent findings on its roles in obesity, diabetes, and atherosclerosis, including GPER-dependent regulation of lipid metabolism and inflammation, are presented. The therapeutic potential of targeting GPER-dependent pathways in chronic diseases such as coronary artery disease and diabetes and in the context of menopause is also discussed.
Collapse
Affiliation(s)
- Matthias Barton
- Molecular Internal Medicine, University of Zurich, Switzerland.
| | - Eric R Prossnitz
- Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87120, USA; UNM Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87120, USA.
| |
Collapse
|
47
|
Hwang HJ, Lee M, Park JH, Jung HS, Kang JG, Kim CS, Lee SJ, Ihm SH. Improved islet transplantation outcome by the co-delivery of siRNAs for iNOS and 17β-estradiol using an R3V6 peptide carrier. Biomaterials 2015; 38:36-42. [DOI: 10.1016/j.biomaterials.2014.10.060] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2014] [Accepted: 10/19/2014] [Indexed: 01/06/2023]
|
48
|
De Marco P, Cirillo F, Vivacqua A, Malaguarnera R, Belfiore A, Maggiolini M. Novel Aspects Concerning the Functional Cross-Talk between the Insulin/IGF-I System and Estrogen Signaling in Cancer Cells. Front Endocrinol (Lausanne) 2015; 6:30. [PMID: 25798130 PMCID: PMC4351617 DOI: 10.3389/fendo.2015.00030] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 02/19/2015] [Indexed: 12/13/2022] Open
Abstract
The insulin/IGF system plays an important role in cancer progression. Accordingly, elevated levels of circulating insulin have been associated with an increased cancer risk as well as with aggressive and metastatic cancer phenotypes. Numerous studies have documented that estrogens cooperate with the insulin/IGF system in multiple pathophysiological conditions. The biological responses to estrogens are mainly mediated by the estrogen receptors (ER)α and ERβ, which act as transcription factors; however, several studies have recently demonstrated that a member of the G protein-coupled receptors, named GPR30/G-protein estrogen receptor (GPER), is also involved in the estrogen signaling in normal and malignant cells as well as in cancer-associated fibroblasts (CAFs). In this regard, novel mechanisms linking the action of estrogens through GPER with the insulin/IGF system have been recently demonstrated. This review recapitulates the relevant aspects of this functional cross-talk between the insulin/IGF and the estrogenic GPER transduction pathways, which occurs in various cell types and may account for cancer progression.
Collapse
Affiliation(s)
- Paola De Marco
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Francesca Cirillo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Adele Vivacqua
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Roberta Malaguarnera
- Endocrinology, Department of Health, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Antonino Belfiore
- Endocrinology, Department of Health, University Magna Graecia of Catanzaro, Catanzaro, Italy
- *Correspondence: Antonino Belfiore, Università degli Studi Magna Graecia di Catanzaro, Viale Europa, Loc. Germaneto, Catanzaro 88100, Italy e-mail: ; Marcello Maggiolini, Università della Calabria, via P. Bucci, Rende 87036, Italy e-mail:
| | - Marcello Maggiolini
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
- *Correspondence: Antonino Belfiore, Università degli Studi Magna Graecia di Catanzaro, Viale Europa, Loc. Germaneto, Catanzaro 88100, Italy e-mail: ; Marcello Maggiolini, Università della Calabria, via P. Bucci, Rende 87036, Italy e-mail:
| |
Collapse
|
49
|
Mirzamohammadi S, Aali E, Najafi R, Kamarul T, Mehrabani M, Aminzadeh A, Sharifi AM. Effect of 17β-estradiol on mediators involved in mesenchymal stromal cell trafficking in cell therapy of diabetes. Cytotherapy 2014; 17:46-57. [PMID: 25457279 DOI: 10.1016/j.jcyt.2014.06.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2014] [Revised: 06/17/2014] [Accepted: 06/23/2014] [Indexed: 12/13/2022]
Abstract
BACKGROUND AIMS Mesenchymal stromal cells (MSCs) have shown great promise for cell therapy of a wide range of diseases such as diabetes. However, insufficient viability of transplanted cells reaching to damaged tissues has limited their potential therapeutic effects. Expression of estrogen receptors on stem cells may suggest a role for 17β-estradiol (E2) in regulating some functions in these cells. There is evidence that E2 enhances homing of stem cells. Induction of hypoxia-inducible factor-1α (HIF-1α) by E2 and the profound effect of HIF-1α on migration of cells have previously been demonstrated. We investigated the effect of E2 on major mediators involved in trafficking and subsequent homing of MSCs both in vitro and in vivo in diabetic rats. METHODS E2 has been selected to improve the poor migration capacity of MSCs toward sites of injury. MSCs were incubated with different concentrations of E2 for varying periods of time to investigate whether estradiol treatment could be effective to enhance the efficiency of MSC transplantation. RESULTS E2 significantly enhanced the viability of the cells that were blocked by ICI 182,780 (estrogen receptor antagonist). E2 also increased HIF-1α, CXC chemokine receptor 4 and C-C chemokine receptor 2 protein and messenger RNA levels measured by Western blot and reverse transcription-polymerase chain reaction. The enzymatic activity of matrix metalloproteinase 2 and metalloproteinase 9 was elevated in E2-treated cells through the use of gelatin zymography. Finally, the improved migration capacity of E2-treated MSCs was evaluated with the use of a Boyden chamber and in vivo migration assays. CONCLUSIONS Our data support that conditioning of MSCs with E2 promotes migration of cells in cultured MSCs in vitro and in a diabetic rat model in vivo through regulation of major mediators of cell trafficking.
Collapse
Affiliation(s)
- Solmaz Mirzamohammadi
- Razi Drug Research Center and Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ehsan Aali
- Razi Drug Research Center and Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Rezvan Najafi
- Department of Molecular Medicine, Hamedan University of Medical Sciences, Hamedan, Iran
| | - Tunku Kamarul
- Tissue Engineering Group (TEG) and Research, National Orthopedic Centre of Excellence in Research and Learning (NOCERAL), Department of Orthopedics, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Mehrnaz Mehrabani
- Razi Drug Research Center and Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Azadeh Aminzadeh
- Razi Drug Research Center and Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Mohammad Sharifi
- Razi Drug Research Center and Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Department of Tissue Engineering and Cell Therapy, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
50
|
De Marco P, Romeo E, Vivacqua A, Malaguarnera R, Abonante S, Romeo F, Pezzi V, Belfiore A, Maggiolini M. GPER1 is regulated by insulin in cancer cells and cancer-associated fibroblasts. Endocr Relat Cancer 2014; 21:739-53. [PMID: 25012984 DOI: 10.1530/erc-14-0245] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Elevated insulin levels have been associated with an increased cancer risk as well as with aggressive and metastatic cancer phenotypes characterized by a poor prognosis. Insulin stimulates the proliferation, migration, and invasiveness of cancer cells through diverse transduction pathways, including estrogen signaling. As G protein estrogen receptor 1 (GPER1) mediates rapid cell responses to estrogens, we evaluated the potential of insulin to regulate GPER1 expression and function in leiomyosarcoma cancer cells (SKUT-1) and breast cancer-associated fibroblasts (CAFs), which were used as a model system. We found that insulin transactivates the GPER1 promoter sequence and increases the mRNA and protein expression of GPER1 through the activation of the PRKCD/MAPK1/c-Fos/AP1 transduction pathway, as ascertained by means of specific pharmacological inhibitors and gene-silencing experiments. Moreover, cell migration triggered by insulin occurred through GPER1 and its main target gene CTGF, whereas the insulin-induced expression of GPER1 boosted cell-cycle progression and the glucose uptake stimulated by estrogens. Notably, a positive correlation between insulin serum levels and GPER1 expression was found in cancer fibroblasts obtained from breast cancer patients. Altogether, our data indicate that GPER1 may be included among the complex network of transduction signaling triggered by insulin that drives cells toward cancer progression.
Collapse
Affiliation(s)
- Paola De Marco
- Department of PharmacyHealth and Nutritional Sciences, University of Calabria, 87036 Rende (CS), ItalyRegional HospitalCosenza, ItalyEndocrinologyDepartment of Health, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Enrica Romeo
- Department of PharmacyHealth and Nutritional Sciences, University of Calabria, 87036 Rende (CS), ItalyRegional HospitalCosenza, ItalyEndocrinologyDepartment of Health, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Adele Vivacqua
- Department of PharmacyHealth and Nutritional Sciences, University of Calabria, 87036 Rende (CS), ItalyRegional HospitalCosenza, ItalyEndocrinologyDepartment of Health, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Roberta Malaguarnera
- Department of PharmacyHealth and Nutritional Sciences, University of Calabria, 87036 Rende (CS), ItalyRegional HospitalCosenza, ItalyEndocrinologyDepartment of Health, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Sergio Abonante
- Department of PharmacyHealth and Nutritional Sciences, University of Calabria, 87036 Rende (CS), ItalyRegional HospitalCosenza, ItalyEndocrinologyDepartment of Health, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Francesco Romeo
- Department of PharmacyHealth and Nutritional Sciences, University of Calabria, 87036 Rende (CS), ItalyRegional HospitalCosenza, ItalyEndocrinologyDepartment of Health, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Vincenzo Pezzi
- Department of PharmacyHealth and Nutritional Sciences, University of Calabria, 87036 Rende (CS), ItalyRegional HospitalCosenza, ItalyEndocrinologyDepartment of Health, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Antonino Belfiore
- Department of PharmacyHealth and Nutritional Sciences, University of Calabria, 87036 Rende (CS), ItalyRegional HospitalCosenza, ItalyEndocrinologyDepartment of Health, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Marcello Maggiolini
- Department of PharmacyHealth and Nutritional Sciences, University of Calabria, 87036 Rende (CS), ItalyRegional HospitalCosenza, ItalyEndocrinologyDepartment of Health, University Magna Graecia of Catanzaro, Catanzaro, Italy
| |
Collapse
|