1
|
Huang W, Xie C, Jones KL, Horowitz M, Rayner CK, Wu T. Sex differences in the plasma glucagon responses to a high carbohydrate meal and a glucose drink in type 2 diabetes. Diabetes Res Clin Pract 2024; 214:111769. [PMID: 38971377 DOI: 10.1016/j.diabres.2024.111769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/20/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
Elevated fasting glucagon concentrations and/or attenuated postprandial glucagon suppression are characteristics of type 2 diabetes (T2D) and contribute to hyperglycaemia. This study shows that hyperglucagonaemia is more prominent in males than females after a nutrient load in T2D, adding insights into sex differences in relation to the pathophysiology of T2D.
Collapse
Affiliation(s)
- Weikun Huang
- Centre for Research Excellence in Translating Nutritional Sciences to Good Health, Adelaide Medical School, The University of Adelaide, Adelaide, Australia
| | - Cong Xie
- Centre for Research Excellence in Translating Nutritional Sciences to Good Health, Adelaide Medical School, The University of Adelaide, Adelaide, Australia; Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide, Australia
| | - Karen L Jones
- Centre for Research Excellence in Translating Nutritional Sciences to Good Health, Adelaide Medical School, The University of Adelaide, Adelaide, Australia; Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide, Australia
| | - Michael Horowitz
- Centre for Research Excellence in Translating Nutritional Sciences to Good Health, Adelaide Medical School, The University of Adelaide, Adelaide, Australia; Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide, Australia
| | - Christopher K Rayner
- Centre for Research Excellence in Translating Nutritional Sciences to Good Health, Adelaide Medical School, The University of Adelaide, Adelaide, Australia; Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Adelaide, Australia
| | - Tongzhi Wu
- Centre for Research Excellence in Translating Nutritional Sciences to Good Health, Adelaide Medical School, The University of Adelaide, Adelaide, Australia.
| |
Collapse
|
2
|
Liu H, Niu T, Qiu G, Cui S, Zhang D. Taurine promotes insulin synthesis by enhancing Isl-1 expression through miR-7a/RAF1/ERK1/2 pathway. In Vitro Cell Dev Biol Anim 2024; 60:23-35. [PMID: 38117455 DOI: 10.1007/s11626-023-00835-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 11/04/2023] [Indexed: 12/21/2023]
Abstract
It has been well established that the circulating taurine affects the insulin synthesis in pancreatic islet β-cells, whereas miR-7a and LIM-homeodomain transcription factor Isl-1 are important intracellular factors regulating insulin transcription and synthesis. However, it still remains unknown whether taurine regulates insulin synthesis by affecting miR-7a and/or Isl-1 expressions in mouse pancreatic islet β-cells. The present study was thus proposed to identify the effects of taurine on the expressions of miR-7a and/or Isl-1 and their relations to insulin synthesis in mouse pancreatic islet β-cells by using miR-7a2 knockout (KO) and taurine transporter (TauT) KO mouse models and the related in vitro experiments. The results demonstrated that taurine supplement significantly decreased the pancreas miR-7a expression, but sharply upregulated the pancreas Isl-1 and insulin expressions, and serum insulin levels. However, the enhanced effects of taurine on Isl-1 expression and insulin synthesis were mitigated in the TauT KO and miR-7a2 KO mice. In addition, our results confirmed that taurine markedly increased pancreas RAF1 and ERK1/2 expressions. Collectively, the present study firstly demonstrates that taurine regulates insulin synthesis through TauT/miR-7a/RAF1/ERK1/2/Isl-1 signaling pathway, which are crucial for our understanding the mechanisms of taurine affecting insulin synthesis, and also potential for establishing the therapeutic strategies for diabetes and the diseases related to metabolism.
Collapse
Affiliation(s)
- Hui Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China
- Institute of Reproduction and Metabolism, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China
| | - Tongjuan Niu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China
- Institute of Reproduction and Metabolism, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China
| | - Guobin Qiu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China
- Institute of Reproduction and Metabolism, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China
| | - Sheng Cui
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China
- Institute of Reproduction and Metabolism, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, Jiangsu, People's Republic of China
| | - Di Zhang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China.
- Institute of Reproduction and Metabolism, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China.
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, Jiangsu, People's Republic of China.
| |
Collapse
|
3
|
Wang F, Zhang Y, Zhang S, Han X, Wei Y, Guo H, Zhang X, Yang H, Wu T, He M. Combined effects of bisphenol A and diabetes genetic risk score on incident type 2 diabetes: A nested case-control study. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 307:119581. [PMID: 35680067 DOI: 10.1016/j.envpol.2022.119581] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 06/01/2022] [Accepted: 06/02/2022] [Indexed: 06/15/2023]
Abstract
Observational studies reported inconsistent results on the association between bisphenol A (BPA) and type 2 diabetes (T2D) risk. Whether genetic factors modified the association remains unclear. The present nested case-control study prospectively investigated the association of BPA with T2D risk, and the interaction and combined effects of diabetes genetic risk score (GRS) and serum BPA on T2D risk. Based on the Dongfeng-Tongji cohort study, 995 incident diabetes cases and 1:1 age- and gender-matched controls were included. T2D was diagnosed based on the American Diabetes Association criteria. Serum BPA concentration was measured at baseline. Diabetes GRS was constructed by 88 diabetes-related SNPs selected from large-scale GWASs. A U-shaped association was observed between serum BPA levels and T2D risk, with the lowest odds of T2D at the serum BPA levels of 1.00 ng/mL (P = 0.001 for nonlinearity). Compared with the middle group, the multivariate-adjusted ORs of T2D in the lowest group and the highest group of serum BPA were 1.52 (95% CI: 1.04, 2.22) and 1.40 (95% CI: 1.08, 1.81), respectively. Both serum BPA levels (β = 0.107, P = 0.001) and weighted-GRS (w-GRS) (β = 0.072, P = 0.02) were significantly associated with baseline FPG levels. Participants with both highest w-GRS and serum BPA levels had highest risk of T2D (OR = 2.53, 95%CI: 1.49, 4.31, P = 0.001) and higher baseline FPG levels (β = 0.218, P = 0.01), compared with those with both lowest w-GRS and serum BPA levels. Non modified effects of serum BPA levels and w-GRS on T2D, baseline FPG levels, and 5-y changes of FPG levels were detected (All Pinteraction > 0.05). Our results suggested a U-shaped association between serum BPA levels and T2D risk. Participants with higher serum BPA levels and diabetes genetic risk had higher FPG levels and higher risk of T2D.
Collapse
Affiliation(s)
- Fei Wang
- Department of Occupational and Environmental Health and Key Laboratory of Environmental and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, 530021, PR China
| | - Ying Zhang
- Department of Occupational and Environmental Health and Key Laboratory of Environmental and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Shiyang Zhang
- Department of Occupational and Environmental Health and Key Laboratory of Environmental and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Xu Han
- Department of Occupational and Environmental Health and Key Laboratory of Environmental and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Yue Wei
- Department of Occupational and Environmental Health and Key Laboratory of Environmental and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Huan Guo
- Department of Occupational and Environmental Health and Key Laboratory of Environmental and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Xiaomin Zhang
- Department of Occupational and Environmental Health and Key Laboratory of Environmental and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Handong Yang
- Dongfeng Central Hospital, Dongfeng Motor Corporation and Hubei University of Medicine, Shiyan, Hubei, 442008, PR China
| | - Tangchun Wu
- Department of Occupational and Environmental Health and Key Laboratory of Environmental and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Meian He
- Department of Occupational and Environmental Health and Key Laboratory of Environmental and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China.
| |
Collapse
|
4
|
Molecular dissection of cellular response of pancreatic islet cells to Bisphenol-A (BPA): a comprehensive review. Biochem Pharmacol 2022; 201:115068. [DOI: 10.1016/j.bcp.2022.115068] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 04/11/2022] [Accepted: 04/25/2022] [Indexed: 12/15/2022]
|
5
|
Hill M, Pařízek A, Šimják P, Koucký M, Anderlová K, Krejčí H, Vejražková D, Ondřejíková L, Černý A, Kancheva R. Steroids, steroid associated substances and gestational diabetes mellitus. Physiol Res 2021. [DOI: 10.33549//physiolres.934794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
As gestational diabetes mellitus (GDM) is both a frequent and serious complication, steroid levels in pregnancy are extremely elevated and their role in pregnancy is crucial, this review focuses on the role of steroids and related substances in the GDM pathophysiology. Low SHBG levels are associated with insulin resistance and hyperinsulinemia, while also predicting a predisposition to GDM. Other relevant agents are placental hormones such as kisspeptin and CRH, playing also an important role beyond pregnancy, but which are synthesized here in smaller amounts in the hypothalamus. These hormones affect both the course of pregnancy as well as the synthesis of pregnancy steroids and may also be involved in the GDM pathophysiology. Steroids, whose biosynthesis is mainly provided by the fetal adrenal glands, placenta, maternal adrenal glands, and both maternal and fetal livers, are also synthesized in limited amounts directly in the pancreas and may influence the development of GDM. These substances involve the sulfated Δ5 steroids primarily acting via modulating different ion channels and influencing the development of GDM in different directions, mostly diabetogenic progesterone and predominantly anti-diabetic estradiol acting both in genomic and non-genomic way, androgens associated with IR and hyperinsulinemia, neuroactive steroids affecting the pituitary functioning, and cortisol whose production is stimulated by CRH but which suppresses its pro-inflammatory effects. Due to the complex actions of steroids, studies assessing their predominant effect and studies assessing their predictive values for estimating predisposition to GDM are needed.
Collapse
Affiliation(s)
- M Hill
- Institute of Endocrinology, Prague, Czech Republic.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Hughson BN. The Glucagon-Like Adipokinetic Hormone in Drosophila melanogaster - Biosynthesis and Secretion. Front Physiol 2021; 12:710652. [PMID: 35002748 PMCID: PMC8733639 DOI: 10.3389/fphys.2021.710652] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 09/16/2021] [Indexed: 11/13/2022] Open
Abstract
Metabolic homeostasis requires the precise regulation of circulating sugar titers. In mammals, homeostatic control of circulating sugar titers requires the coordinated secretion and systemic activities of glucagon and insulin. Metabolic homeostasis is similarly regulated in Drosophila melanogaster through the glucagon-like adipokinetic hormone (AKH) and the Drosophila insulin-like peptides (DILPs). In flies and mammals, glucagon and AKH are biosynthesized in and secreted from specialized endocrine cells. KATP channels borne on these cells respond to fluctuations in circulating glucose titers and thereby regulate glucagon secretion. The influence of glucagon in the pathogenesis of type 2 diabetes mellitus is now recognized, and a crucial mechanism that regulates glucagon secretion was reported nearly a decade ago. Ongoing efforts to develop D. melanogaster models for metabolic syndrome must build upon this seminal work. These efforts make a critical review of AKH physiology timely. This review focuses on AKH biosynthesis and the regulation of glucose-responsive AKH secretion through changes in CC cell electrical activity. Future directions for AKH research in flies are discussed, including the development of models for hyperglucagonemia and epigenetic inheritance of acquired metabolic traits. Many avenues of AKH physiology remain to be explored and thus present great potential for improving the utility of D. melanogaster in metabolic research.
Collapse
Affiliation(s)
- Bryon N. Hughson
- Department of Ecology and Evolutionary Biology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
7
|
Acconcia F, Fiocchetti M, Busonero C, Fernandez VS, Montalesi E, Cipolletti M, Pallottini V, Marino M. The extra-nuclear interactome of the estrogen receptors: implications for physiological functions. Mol Cell Endocrinol 2021; 538:111452. [PMID: 34500041 DOI: 10.1016/j.mce.2021.111452] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 08/19/2021] [Accepted: 09/02/2021] [Indexed: 02/07/2023]
Abstract
Over the last decades, a great body of evidence has defined a novel view of the cellular mechanism of action of the steroid hormone 17β-estradiol (E2) through its estrogen receptors (i.e., ERα and ERβ). It is now clear that the E2-activated ERs work both as transcription factors and extra-nuclear plasma membrane-localized receptors. The activation of a plethora of signal transduction cascades follows the E2-dependent engagement of plasma membrane-localized ERs and is required for the coordination of gene expression, which ultimately controls the occurrence of the pleiotropic effects of E2. The definition of the molecular mechanisms by which the ERs locate at the cell surface (i.e., palmitoylation and protein association) determined the quest for understanding the specificity of the extra-nuclear E2 signaling. The use of mice models lacking the plasma membrane ERα localization unveiled that the extra-nuclear E2 signaling is operational in vivo but tissue-specific. However, the underlying molecular details for such ERs signaling diversity in the perspective of the E2 physiological functions in the different cellular contexts are still not understood. Therefore, to gain insights into the tissue specificity of the extra-nuclear E2 signaling to physiological functions, here we reviewed the known ERs extra-nuclear interactors and tried to extrapolate from available databases the ERα and ERβ extra-nuclear interactomes. Based on literature data, it is possible to conclude that by specifically binding to extra-nuclear localized proteins in different sub-cellular compartments, the ERs fine-tune their molecular activities. Moreover, we report that the context-dependent diversity of the ERs-mediated extra-nuclear E2 actions can be ascribed to the great flexibility of the physical structures of ERs and the spatial-temporal organization of the logistics of the cells (i.e., the endocytic compartments). Finally, we provide lists of proteins belonging to the potential ERα and ERβ extra-nuclear interactomes and propose that the systematic experimental definition of the ERs extra-nuclear interactomes in different tissues represents the next step for the research in the ERs field. Such characterization will be fundamental for the identification of novel druggable targets for the innovative treatment of ERs-related diseases.
Collapse
Affiliation(s)
- Filippo Acconcia
- Department of Sciences, Section Biomedical Sciences, and Technology, University Roma Tre, Viale Guglielmo Marconi, 446, I-00146, Rome, Italy.
| | - Marco Fiocchetti
- Department of Sciences, Section Biomedical Sciences, and Technology, University Roma Tre, Viale Guglielmo Marconi, 446, I-00146, Rome, Italy
| | - Claudia Busonero
- Department of Sciences, Section Biomedical Sciences, and Technology, University Roma Tre, Viale Guglielmo Marconi, 446, I-00146, Rome, Italy
| | - Virginia Solar Fernandez
- Department of Sciences, Section Biomedical Sciences, and Technology, University Roma Tre, Viale Guglielmo Marconi, 446, I-00146, Rome, Italy
| | - Emiliano Montalesi
- Department of Sciences, Section Biomedical Sciences, and Technology, University Roma Tre, Viale Guglielmo Marconi, 446, I-00146, Rome, Italy
| | - Manuela Cipolletti
- Department of Sciences, Section Biomedical Sciences, and Technology, University Roma Tre, Viale Guglielmo Marconi, 446, I-00146, Rome, Italy
| | - Valentina Pallottini
- Department of Sciences, Section Biomedical Sciences, and Technology, University Roma Tre, Viale Guglielmo Marconi, 446, I-00146, Rome, Italy
| | - Maria Marino
- Department of Sciences, Section Biomedical Sciences, and Technology, University Roma Tre, Viale Guglielmo Marconi, 446, I-00146, Rome, Italy.
| |
Collapse
|
8
|
Sexual hormones and diabetes: The impact of estradiol in pancreatic β cell. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021. [PMID: 33832654 DOI: 10.1016/bs.ircmb.2021.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/13/2023]
Abstract
Diabetes is one of the most prevalent metabolic diseases and its incidence is increasing throughout the world. Data from World Health Organization (WHO) point-out that diabetes is a major cause of blindness, kidney failure, heart attacks, stroke and lower limb amputation and estimated 1.6 million deaths were directly caused by it in 2016. Population studies show that the incidence of this disease increases in women after menopause, when the production of estrogen is decreasing in them. Knowing the impact that estrogenic signaling has on insulin-secreting β cells is key to prevention and design of new therapeutic targets. This chapter explores the role of estrogen and their receptors in the regulation of insulin secretion and biosynthesis, proliferation, regeneration and survival in pancreatic β cells. In addition, delves into the genetic animal models developed and its application for the specific study of the different estrogen signaling pathways. Finally, discusses the impact of menopause and hormone replacement therapy on pancreatic β cell function.
Collapse
|
9
|
Begum M, Paul P, Das D, Chakraborty K, Bhattacharjee A, Ghosh S. Genes regulating development and behavior exhibited altered expression in Drosophila melanogaster exposed to bisphenol A: use of real-time quantitative PCR (qRT-PCR) and droplet digital PCR (ddPCR) in genotoxicity study. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:7090-7104. [PMID: 33025430 DOI: 10.1007/s11356-020-10805-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Accepted: 09/09/2020] [Indexed: 06/11/2023]
Abstract
Toxicity of bisphenol A on morphological and life-history traits of model insect Drosophila melanogaster was reported in our previous work. In the present study, we have analyzed the adversity of bisphenol A on the reproductive behavior of adult and on the expression of selected genes in the larva and adult stage of fruit fly exposed to bisphenol A (0.007 g/2 ml. or 3.5 mg/ml), in addition to determination of LC50 value of bisphenol A in larva and pupal stage. We employed both the quantitative reverse transcriptase PCR and droplet digital PCR for analyzing the expression profile of seven genes namely, decapentaplegic, vestigial, wingless, foraging, insulin-like receptor, doublesex, and fruitless. We found bisphenol A has more adverse effects on male sexual behavior than females. Moreover, we observed significant downregulation of all the selected genes in treated larvae except, fruitless in male where it showed significant upregulation. On contrary among the treated adult flies, significant downregulation of all target genes in both sexes is evident, except, doublesex and fruitless in males which showed significant upregulation. We did not observe any deviation of male: female sex ratio from 1:1 under bisphenol A exposure. All these results suggest bisphenol A adversely affects the optimum functioning of genes which are involved in the regulation of metabolic pathways, behavioral pattern, stress response, endocrine homeostasis, neural functioning, and the development of the specific organ in Drosophila melanogaster. Our result not only provides a foundation to study further the bisphenol A toxicity on different pivotal genes in Drosophila but also suggests the use of the droplet digital PCR technology in toxicity measurement at the molecular level in eukaryotic model systems.
Collapse
Affiliation(s)
- Morium Begum
- Department of Zoology, Cytogenetics & Genomics Research Unit, University of Calcutta, Taraknath-Palit-Siksha-Prangan (Ballygunge Science College Campus), 35, Ballygunge Circular Road.Kolkata, West Bengal, 700019, India
| | - Pallab Paul
- Department of Zoology, Cytogenetics & Genomics Research Unit, University of Calcutta, Taraknath-Palit-Siksha-Prangan (Ballygunge Science College Campus), 35, Ballygunge Circular Road.Kolkata, West Bengal, 700019, India
| | - Debasmita Das
- Department of Zoology, Cytogenetics & Genomics Research Unit, University of Calcutta, Taraknath-Palit-Siksha-Prangan (Ballygunge Science College Campus), 35, Ballygunge Circular Road.Kolkata, West Bengal, 700019, India
| | - Kaustav Chakraborty
- Amity Institute of Biotechnology, Amity-University Kolkata, Plot no 36, 37, and 38, Major Arterial Road (South-East), Action Area II, Newtown, Kolkata, West Bengal, 700135, India
| | - Ashima Bhattacharjee
- Amity Institute of Biotechnology, Amity-University Kolkata, Plot no 36, 37, and 38, Major Arterial Road (South-East), Action Area II, Newtown, Kolkata, West Bengal, 700135, India
| | - Sujay Ghosh
- Department of Zoology, Cytogenetics & Genomics Research Unit, University of Calcutta, Taraknath-Palit-Siksha-Prangan (Ballygunge Science College Campus), 35, Ballygunge Circular Road.Kolkata, West Bengal, 700019, India.
| |
Collapse
|
10
|
Maczewsky J, Kaiser J, Krippeit-Drews P, Drews G. Approved LXR agonists exert unspecific effects on pancreatic β-cell function. Endocrine 2020; 68:526-535. [PMID: 32146655 PMCID: PMC7308254 DOI: 10.1007/s12020-020-02241-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 02/24/2020] [Indexed: 12/20/2022]
Abstract
Novel agonists of the nuclear liver-X-receptor (LXR) are designed to treat metabolic disorders or cancer. The rationale to develop these new drugs is based on promising results with established LXR agonist like T0901317 and GW3965. LXRα and LXRβ are expressed in β-cells, and expression is increased by T0901317. The aim of the present study was to evaluate whether effects of these drugs on β-cell function are specific and reliably linked to LXR activation. T0901317 and GW3965, widely used as specific LXR agonists, show rapid, non-genomic effects on stimulus-secretion coupling of mouse pancreatic β-cells at low µM concentrations. T0901317 lowered the cytosolic Ca2+ concentration, reduced or completely inhibited action potentials, and decreased insulin secretion. GW3965 exerted similar effects on insulin secretion. T0901317 affected the production of reactive oxygen species and ATP. The involvement of the classical nuclear LXRs in T0901317- and GW3965-mediated effects in β-cells could be ruled out using LXRα, LXRβ and double knockout mice. Our results strongly suggest that LXR agonists, that are considered to be specific for this receptor, interfere with mitochondrial metabolism and metabolism-independent processes in β-cells. Thus, it is indispensable to test novel LXR agonists accompanying to ongoing clinical trials for acute and chronic effects on cell function in cellular systems and/or animal models lacking classical LXRs.
Collapse
Affiliation(s)
- Jonas Maczewsky
- Institute of Pharmacy, Department of Pharmacology, University of Tübingen, Auf der Morgenstelle 8, 72076, Tübingen, Germany
| | - Julia Kaiser
- Institute of Pharmacy, Department of Pharmacology, University of Tübingen, Auf der Morgenstelle 8, 72076, Tübingen, Germany
| | - Peter Krippeit-Drews
- Institute of Pharmacy, Department of Pharmacology, University of Tübingen, Auf der Morgenstelle 8, 72076, Tübingen, Germany
| | - Gisela Drews
- Institute of Pharmacy, Department of Pharmacology, University of Tübingen, Auf der Morgenstelle 8, 72076, Tübingen, Germany.
| |
Collapse
|
11
|
Manukyan L, Dunder L, Lind PM, Bergsten P, Lejonklou MH. Developmental exposure to a very low dose of bisphenol A induces persistent islet insulin hypersecretion in Fischer 344 rat offspring. ENVIRONMENTAL RESEARCH 2019; 172:127-136. [PMID: 30782532 DOI: 10.1016/j.envres.2019.02.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 01/22/2019] [Accepted: 02/07/2019] [Indexed: 06/09/2023]
Abstract
BACKGROUND In children with obesity, accentuated insulin secretion has been coupled with development of type 2 diabetes mellitus (T2DM). Bisphenol A (BPA) is a chemical with endocrine- and metabolism-disrupting properties which can be measured in a majority of the population. Exposure to BPA has been associated with the development of metabolic diseases including T2DM. OBJECTIVE The aim of this study was to investigate if exposure early in life to an environmentally relevant low dose of BPA causes insulin hypersecretion in rat offspring. METHODS Pregnant Fischer 344 rats were exposed to 0.5 (BPA0.5) or 50 (BPA50) µg BPA/kg BW/day via drinking water from gestational day 3.5 until postnatal day 22. Pancreata from dams and 5- and 52-week-old offspring were procured and islets were isolated by collagenase digestion. Glucose-stimulated insulin secretion and insulin content in the islets were determined by ELISA. RESULTS Basal (5.5 mM glucose) islet insulin secretion was not affected by BPA exposure. However, stimulated (11 mM glucose) insulin secretion was enhanced by about 50% in islets isolated from BPA0.5-exposed 5- and 52-week-old female and male offspring and by 80% in islets from dams, compared with control. In contrast, the higher dose, BPA50, reduced stimulated insulin secretion by 40% in both 5- and 52-week-old female and male offspring and dams, compared with control. CONCLUSION A BPA intake 8 times lower than the European Food Safety Authority's (EFSA's) current tolerable daily intake (TDI) of 4 µg/kg BW/day of BPA delivered via drinking water during gestation and early development causes islet insulin hypersecretion in rat offspring up to one year after exposure. The effects of BPA exposure on the endocrine pancreas may promote the development of metabolic disease including T2DM.
Collapse
Affiliation(s)
- Levon Manukyan
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden.
| | - Linda Dunder
- Department of Medical Sciences, Occupational and Environmental Medicine, Uppsala University, Uppsala, Sweden.
| | - P Monica Lind
- Department of Medical Sciences, Occupational and Environmental Medicine, Uppsala University, Uppsala, Sweden.
| | - Peter Bergsten
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden; Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden.
| | - Margareta H Lejonklou
- Department of Medical Sciences, Occupational and Environmental Medicine, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
12
|
Xiao X, Guo P, Shiota C, Zhang T, Coudriet GM, Fischbach S, Prasadan K, Fusco J, Ramachandran S, Witkowski P, Piganelli JD, Gittes GK. Endogenous Reprogramming of Alpha Cells into Beta Cells, Induced by Viral Gene Therapy, Reverses Autoimmune Diabetes. Cell Stem Cell 2019; 22:78-90.e4. [PMID: 29304344 DOI: 10.1016/j.stem.2017.11.020] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 09/14/2017] [Accepted: 11/26/2017] [Indexed: 12/25/2022]
Abstract
Successful strategies for treating type 1 diabetes need to restore the function of pancreatic beta cells that are destroyed by the immune system and overcome further destruction of insulin-producing cells. Here, we infused adeno-associated virus carrying Pdx1 and MafA expression cassettes through the pancreatic duct to reprogram alpha cells into functional beta cells and normalized blood glucose in both beta cell-toxin-induced diabetic mice and in autoimmune non-obese diabetic (NOD) mice. The euglycemia in toxin-induced diabetic mice and new insulin+ cells persisted in the autoimmune NOD mice for 4 months prior to reestablishment of autoimmune diabetes. This gene therapy strategy also induced alpha to beta cell conversion in toxin-treated human islets, which restored blood glucose levels in NOD/SCID mice upon transplantation. Hence, this strategy could represent a new therapeutic approach, perhaps complemented by immunosuppression, to bolster endogenous insulin production. Our study thus provides a potential basis for further investigation in human type 1 diabetes.
Collapse
Affiliation(s)
- Xiangwei Xiao
- Division of Pediatric Surgery, Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, 4401 Penn Avenue, Pittsburgh, PA 15224, USA.
| | - Ping Guo
- Division of Pediatric Surgery, Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, 4401 Penn Avenue, Pittsburgh, PA 15224, USA
| | - Chiyo Shiota
- Division of Pediatric Surgery, Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, 4401 Penn Avenue, Pittsburgh, PA 15224, USA
| | - Ting Zhang
- Division of Pediatric Surgery, Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, 4401 Penn Avenue, Pittsburgh, PA 15224, USA
| | - Gina M Coudriet
- Division of Pediatric Surgery, Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, 4401 Penn Avenue, Pittsburgh, PA 15224, USA
| | - Shane Fischbach
- Division of Pediatric Surgery, Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, 4401 Penn Avenue, Pittsburgh, PA 15224, USA
| | - Krishna Prasadan
- Division of Pediatric Surgery, Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, 4401 Penn Avenue, Pittsburgh, PA 15224, USA
| | - Joseph Fusco
- Division of Pediatric Surgery, Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, 4401 Penn Avenue, Pittsburgh, PA 15224, USA
| | | | - Piotr Witkowski
- Department of Surgery, University of Chicago, Chicago, IL 60637, USA
| | - Jon D Piganelli
- Division of Pediatric Surgery, Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, 4401 Penn Avenue, Pittsburgh, PA 15224, USA
| | - George K Gittes
- Division of Pediatric Surgery, Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, 4401 Penn Avenue, Pittsburgh, PA 15224, USA.
| |
Collapse
|
13
|
Sehl ME, Ganz PA. Potential Mechanisms of Age Acceleration Caused by Estrogen Deprivation: Do Endocrine Therapies Carry the Same Risks? JNCI Cancer Spectr 2018; 2:pky035. [PMID: 31360862 PMCID: PMC6649786 DOI: 10.1093/jncics/pky035] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 05/30/2018] [Accepted: 06/22/2018] [Indexed: 02/06/2023] Open
Abstract
Longer duration of endocrine therapy decreases breast cancer recurrence and mortality, but these benefits need to be weighed against potential risks to overall health. Notable side effects of endocrine therapy include cataracts, uterine cancer, thromboembolic events, osteoporosis and fracture risk, chronic musculoskeletal complaints, as well as vaginal dryness and discharge, and vasomotor symptoms. Estrogen deprivation in healthy women younger than 50 years undergoing bilateral oophorectomy has been shown to accelerate the development of diseases related to aging, including coronary artery disease, cardiac arrhythmias, stroke, dementia, and osteoporosis, raising concern that even less dramatic modulation of estrogen homeostasis may adversely affect health outcomes. Diminished available estrogen at the cellular and molecular level may facilitate mechanisms that underlie the aging process, often termed the hallmarks of aging. In this review, we describe estrogen's role in normal physiology across tissues, review the effects of estrogen deprivation on health outcomes in the setting of both surgical and natural menopause, and examine the hallmarks of aging with attention to the effects of estrogen and estrogen blockade on each molecular mechanism underlying the aging process.
Collapse
Affiliation(s)
- Mary E Sehl
- Medicine, Hematology-Oncology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA.,Biomathematics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| | - Patricia A Ganz
- Medicine, Hematology-Oncology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA.,Health Policy and Management, School of Public Health, University of California Los Angeles, Los Angeles, CA
| |
Collapse
|
14
|
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: 2.1] [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
|
15
|
Zhang L, Zhao Y, Guo L. 17β-estradiol protects INS-1 insulinoma cells from mitophagy via G protein-coupled estrogen receptors and the PI3K/Akt signaling pathway. Int J Mol Med 2018; 41:2839-2846. [PMID: 29436590 DOI: 10.3892/ijmm.2018.3470] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Accepted: 12/29/2017] [Indexed: 11/06/2022] Open
Abstract
17β-estradiol (17β-E2) is a steroid hormone that is known to exert effects on blood glucose homeostasis. The G protein‑coupled estrogen receptor (GPER) has been identified as a non-genomic estrogenic receptor, and is involved in numerous physiological processes, including cell survival, energy provision and metabolism. 17β-E2 may decrease apoptosis by binding to the GPER. The phosphoinositide 3-kinase (PI3K)/Akt signaling pathway is involved in physiological and pathological functions such as autophagy. The purpose of the present study was to investigate the role of the PI3K/Akt signaling pathway in the mediation of the effects of GPERs, and the effects of 17β-E2 on mitophagy in INS-1 cells, a rat insulin‑secreting β-cell line. In vitro, INS-1 cells were treated with different concentrations of 17β-E2 with and without pretreatment with a GPER antagonist (G15) or PI3K antagonist (LY294002) and compared with a negative control. An immunofluorescence assay demonstrated that GPERs are expressed in INS-1 cells. Western blot assays demonstrated that 17β-E2 increased GPER levels and the phosphorylation of Akt. Transmission electronic microscopy revealed that 17β-E2 reduced the formation of mitophagosomes and autophagosomes in INS-1 cells. An immunofluorescence staining assay indicated that the co-localization of translocase of mitochondrial outer membrane complex 20 (TOM20) with lysosomal-associated membrane protein 2 (LAMP2) was decreased in INS-1 cells treated with 17β-E2 alone. Western blotting demonstrated that 17β-E2 reduced the protein levels of activated microtubule-associated protein-1 light chain 3, and increased those of TOM20 and mitochondrial heat-shock protein 60. Notably, the protective effects of 17β-E2 were significantly diminished by G15 or LY294002. In conclusion, the present study suggests that 17β-E2 activates the PI3K/Akt pathway via the GPER in INS-1 cells. Furthermore, 17β-E2 may be involved in mitophagy by the regulating the GPER/PI3K/Akt pathway.
Collapse
Affiliation(s)
- Liang Zhang
- Department of Endocrinology, The First Affiliated Hospital, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Yuyan Zhao
- Department of Endocrinology, The First Affiliated Hospital, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Lei Guo
- Department of Orthopedic Surgery, The First Affiliated Hospital, China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
16
|
Rorsman P, Ashcroft FM. Pancreatic β-Cell Electrical Activity and Insulin Secretion: Of Mice and Men. Physiol Rev 2018; 98:117-214. [PMID: 29212789 PMCID: PMC5866358 DOI: 10.1152/physrev.00008.2017] [Citation(s) in RCA: 475] [Impact Index Per Article: 67.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 05/30/2017] [Accepted: 06/18/2017] [Indexed: 12/14/2022] Open
Abstract
The pancreatic β-cell plays a key role in glucose homeostasis by secreting insulin, the only hormone capable of lowering the blood glucose concentration. Impaired insulin secretion results in the chronic hyperglycemia that characterizes type 2 diabetes (T2DM), which currently afflicts >450 million people worldwide. The healthy β-cell acts as a glucose sensor matching its output to the circulating glucose concentration. It does so via metabolically induced changes in electrical activity, which culminate in an increase in the cytoplasmic Ca2+ concentration and initiation of Ca2+-dependent exocytosis of insulin-containing secretory granules. Here, we review recent advances in our understanding of the β-cell transcriptome, electrical activity, and insulin exocytosis. We highlight salient differences between mouse and human β-cells, provide models of how the different ion channels contribute to their electrical activity and insulin secretion, and conclude by discussing how these processes become perturbed in T2DM.
Collapse
Affiliation(s)
- Patrik Rorsman
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, United Kingdom; Department of Neuroscience and Physiology, Metabolic Research Unit, Göteborg, Sweden; and Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Frances M Ashcroft
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, United Kingdom; Department of Neuroscience and Physiology, Metabolic Research Unit, Göteborg, Sweden; and Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
17
|
Effect of Bisphenol-A (BPA) on insulin signal transduction and GLUT4 translocation in gastrocnemius muscle of adult male albino rat. Int J Biochem Cell Biol 2017; 90:38-47. [PMID: 28739533 DOI: 10.1016/j.biocel.2017.07.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 07/05/2017] [Accepted: 07/13/2017] [Indexed: 01/22/2023]
Abstract
Environmental estrogens bind to estrogen receptors, mimic estrogenic actions, and have adverse effects on human health like Bisphenol - A (BPA) which is used as a monomer in the production of polycarbonate plastics (PC) and epoxy resins which are used in variety of canned foods. Skeletal muscle plays an essential role in maintaining systemic glucose metabolism. In the present study, we investigated the possible effects of BPA on insulin signalling molecules and GLUT4 translocation in the gastrocnemius muscle of adult male rat. Rats were divided into four groups - Group I: Control (vehicle-corn oil treated), Group II, III and IV were administered with BPA (10, 100 and 400mg/kg b.wt/day, respectively) through oral gavage. Fasting blood glucose level of BPA treated groups showed a significant increase, oral glucose tolerance and insulin tolerance were also impaired in these animals. BPA significantly decreased the protein levels of insulin signalling molecules like IR, IRS-1, Akt, AS160 and its phosphorylated forms and blunts GLUT4 translocation by altering the levels of v- and t- SNARE proteins that assist the translocation process, thereby decreasing glucose uptake and oxidation in the gastrocnemius muscle. These results suggest that BPA has detrimental effects on insulin signalling molecules and GLUT4 translocation in the gastrocnemius muscle and thus impairs glucose homeostasis.
Collapse
|
18
|
Dose-dependent effect of Bisphenol-A on insulin signaling molecules in cardiac muscle of adult male rat. Chem Biol Interact 2017; 266:10-16. [DOI: 10.1016/j.cbi.2017.01.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 01/20/2017] [Accepted: 01/26/2017] [Indexed: 01/16/2023]
|
19
|
Lillo MA, Nichols C, Seagroves TN, Miranda-Carboni GA, Krum SA. Bisphenol A Induces Sox2 in ER + Breast Cancer Stem-Like Cells. Discov Oncol 2017; 8:90-99. [PMID: 28244015 DOI: 10.1007/s12672-017-0286-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 02/06/2017] [Indexed: 12/11/2022] Open
Abstract
Bisphenol A (BPA) is an endocrine disrupting compound used in food and beverage plastic containers and has been shown to increase breast cancer cellular proliferation. However, the concentrations of BPA used in these experiments are far higher than the physiological levels of BPA detected in the human body. We observed in vitro that exposure of MCF-7 cells to physiological concentrations of BPA failed to increase cell proliferation or to induce canonical estrogen-responsive genes (pS2 and progesterone receptor (PR)), in contrast to 17β-estradiol (E2) treatment. However, MCF-7 cells treated with 10 nM BPA induced ALDH1 expression, a marker of human mammary stem cells. When treated with 10 nM BPA, mammospheres derived either from MCF-7 cells, a patient-derived xenograft, or the normal mouse mammary gland exhibited increased size; however, these effects were not observed in MDA-MB-231 mammospheres. Mechanistically, BPA induced SOX2 mRNA and protein in MCF-7 mammospheres, resulting from enhanced CREB phosphorylation, and subsequent binding of pCREB to a SOX2 downstream enhancer. These findings suggest that physiological levels of BPA increase estrogen receptor-positive breast cancer tumor maintenance through enhanced cancer stem-like cell activity via direct regulation of SOX2 transcription.
Collapse
Affiliation(s)
- M Angeles Lillo
- Department of Orthopaedic Surgery and Biomedical Engineering, University of Tennessee Health Science Center, Memphis, TN, USA.,Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Cydney Nichols
- Department of Microbiology, Immunology and Molecular Genetics, UCLA, Los Angeles, CA, USA
| | - Tiffany N Seagroves
- Department of Pathology, University of Tennessee Health Science Center, Memphis, TN, USA.,Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Gustavo A Miranda-Carboni
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA.,Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Susan A Krum
- Department of Orthopaedic Surgery and Biomedical Engineering, University of Tennessee Health Science Center, Memphis, TN, USA. .,Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
20
|
Santos RS, Batista TM, Camargo RL, Morato PN, Borck PC, Leite NC, Kurauti MA, Wanschel ACBA, Nadal Á, Clegg DJ, Carneiro EM. Lacking of estradiol reduces insulin exocytosis from pancreatic β-cells and increases hepatic insulin degradation. Steroids 2016; 114:16-24. [PMID: 27192429 DOI: 10.1016/j.steroids.2016.05.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 04/18/2016] [Accepted: 05/11/2016] [Indexed: 01/08/2023]
Abstract
Low levels of plasma estrogens are associated with weight-gain, android fat distribution, and a high prevalence of obesity-related comorbidities such as glucose intolerance and type II diabetes. The mechanisms underlying the association between low levels of estrogens and impaired glucose homeostasis are not completely understood. To begin to test this, we used three-month-old female C57BL/6J mice that either underwent ovariectomy (OVX) or received a sham surgery (Sham), and we characterized glucose homeostasis. In a subsequent series of experiments, OVX mice received estradiol treatment (OVX+E2) or vehicle (OVX) for 6 consecutive days. As has been previously reported, lack of ovarian hormones resulted in dysregulated glucose homeostasis. To begin to explore the mechanisms by which this occurs, we characterized the impact of estrogens on insulin secretion and degradation in these mice. Insulin secretion and plasma insulin levels were lower in OVX mice. OVX mice had lower levels of pancreatic Syntaxin 1-A (Synt-1A) protein, which is involved in insulin extrusion from the pancreas. In the liver, OVX mice had higher levels of insulin-degrading enzyme (IDE) and this was associated with higher insulin clearance. Estradiol treatment improved glucose intolerance in OVX mice and restored insulin secretion, as well as normalized the protein content of pancreatic Synt-1A. The addition of estrogens to OVX mice reduced IDE protein to that of Sham mice. Our data suggest loss of ovarian estradiol following OVX led to impaired glucose homeostasis due to pancreatic β-cell dysfunction in the exocytosis of insulin, and upregulation of hepatic IDE protein content resulting in lower insulinemia, which was normalized by estradiol replacement.
Collapse
Affiliation(s)
- Roberta S Santos
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, e Centro de Pesquisa em Obesidade e Comorbidades, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil; Biomedical Research Department, Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Beverly Hills, CA, United States.
| | - Thiago M Batista
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, e Centro de Pesquisa em Obesidade e Comorbidades, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil
| | - Rafael L Camargo
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, e Centro de Pesquisa em Obesidade e Comorbidades, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil
| | - Priscila N Morato
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, e Centro de Pesquisa em Obesidade e Comorbidades, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil
| | - Patrícia C Borck
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, e Centro de Pesquisa em Obesidade e Comorbidades, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil
| | - Nayara C Leite
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, e Centro de Pesquisa em Obesidade e Comorbidades, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil
| | - Mirian A Kurauti
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, e Centro de Pesquisa em Obesidade e Comorbidades, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil
| | - Amarylis C B A Wanschel
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, e Centro de Pesquisa em Obesidade e Comorbidades, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil
| | - Ángel Nadal
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Universidad Miguel Hernández de Elche, Elche, Alicante, Spain
| | - Deborah J Clegg
- Biomedical Research Department, Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Beverly Hills, CA, United States
| | - Everardo M Carneiro
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, e Centro de Pesquisa em Obesidade e Comorbidades, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil
| |
Collapse
|
21
|
Abstract
Bisphenol A (BPA) is used extensively in the world and is present in a diverse range of manufactured articles including dental resins, polycarbonate plastics, and the inner coating of food cans. It is a high volume chemical, with global production at 3.6 × 10(9) kg per year. BPA was identified as a high priority for assessment of human health risk because it was considered to present greatest potential for human exposure. Most studies of the health effects of BPA have focused on endocrine disruption leading to reproductive toxicity, but it displays additional side effects, including liver damage, disrupted pancreatic β-cell function, thyroid hormone disruption, and obesity-promoting effects. In this article, we reviewed specifically on the effects of BPA in energy balance.
Collapse
Affiliation(s)
- L Le Corre
- a Laboratoire de Toxicologie Alimentaire, INSERM U866, NUTOX, Derttech Packtox , University of Burgundy , Dijon , France
| | | | | |
Collapse
|
22
|
Zhang X, Mu L. Association between macrophage migration inhibitory factor in the endometrium and estrogen in endometriosis. Exp Ther Med 2015; 10:787-791. [PMID: 26622394 PMCID: PMC4509047 DOI: 10.3892/etm.2015.2516] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 04/01/2015] [Indexed: 11/05/2022] Open
Abstract
Recent studies have shown that macrophage migration inhibitory factor (MIF) has a possible role in endometriosis-related pain and infertility, yet it has not been explored whether the mRNA level of MIF is altered in endometrial tissues from patients with endometriosis. The aim of the present study was to compare the expression of MIF in endometrial tissues from women with and without endometriosis, and to analyze the association between endometrial MIF expression and 17β-estradiol (E2). The protein and mRNA expression of MIF in the human endometrial tissue was assessed by western blotting and reverse transcription-polymerase chain reaction analysis, respectively. The MIF expression of women with endometriosis was found to be significantly higher than that of the controls. A positive correlation was noted between the serum E2 level and MIF expression. In endometrial cells from women with endometriosis, the level of E2-induced MIF upregulation was significantly higher than that in cells from women without endometriosis. In conclusion, this study demonstrated a significant increase in MIF expression in the endometrial tissues of women with endometriosis and an association between MIF expression and E2 level. MIF expression in endometrial cells from patients with endometriosis showed an increased sensitivity to stimulation by E2.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310000, P.R. China
| | - Lin Mu
- Department of Gynecology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310000, P.R. China
| |
Collapse
|
23
|
Role of G protein-coupled orphan receptors in intestinal inflammation: novel targets in inflammatory bowel diseases. Inflamm Bowel Dis 2015; 21:666-73. [PMID: 25461621 DOI: 10.1097/mib.0000000000000258] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
A large number of proteins were classified into the family of G protein-coupled receptors (GPCRs). Based on their characteristic serpentine domain, they are called 7 TM receptors. Presently, their ligands and physiological functions remain unknown. In this review, we summarize what is known on these receptors and discuss the potential use of these orphan GPCRs (GPRs) in the induction or maintenance of remission in inflammatory bowel diseases. We focus on GPRs 30, 41, 43, 55, 119, and 120, where scientific evidence supports a potential role in intestinal inflammation.
Collapse
|
24
|
Yoon K, Kwack SJ, Kim HS, Lee BM. Estrogenic endocrine-disrupting chemicals: molecular mechanisms of actions on putative human diseases. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2014; 17:127-74. [PMID: 24749480 DOI: 10.1080/10937404.2014.882194] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Endocrine-disrupting chemicals (EDC), including phthalates, bisphenol A (BPA), phytoestrogens such as genistein and daidzein, dichlorodiphenyltrichloroethane (DDT), and 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), are associated with a variety of adverse health effects in organisms or progeny by altering the endocrine system. Environmental estrogens, including BPA, phthalates, and phytoestrogens, are the most extensively studied and are considered to mimic the actions of endogenous estrogen, 17β-estradiol (E2). Diverse modes of action of estrogen and estrogen receptors (ERα and ERβ) have been described, but the mode of action of estrogenic EDC is postulated to be more complex and needs to be more clearly elucidated. This review examines the adverse effects of estrogenic EDC on male or female reproductive systems and molecular mechanisms underlying EDC effects that modulate ER-mediated signaling. Mechanisms of action for estrogenic EDC may involve both ER-dependent and ER-independent pathways. Recent findings from systems toxicology of examining estrogenic EDC are also discussed.
Collapse
Affiliation(s)
- Kyungsil Yoon
- a Lung Cancer Branch , Research Institute, National Cancer Center , Goyang , Gyeonggi-do , South Korea
| | | | | | | |
Collapse
|
25
|
Evans PD, Bayliss A, Reale V. GPCR-mediated rapid, non-genomic actions of steroids: comparisons between DmDopEcR and GPER1 (GPR30). Gen Comp Endocrinol 2014; 195:157-63. [PMID: 24188886 DOI: 10.1016/j.ygcen.2013.10.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Revised: 10/16/2013] [Accepted: 10/21/2013] [Indexed: 10/26/2022]
Abstract
Steroid hormones classically mediate their actions by binding to intracellular receptor proteins that migrate to the nucleus and act as transcription factors to change gene expression. However, evidence is now accumulating for rapid, non-genomic effects of steroids. There is considerable controversy over the mechanisms underlying such effects. In a number of cases evidence has been presented for the direct activation of G-protein coupled receptors (GPCRs) by steroids, either at the plasma membrane, or at intracellular locations. Here, we will focus on the non-genomic actions of ecdysteroids on a Drosophila GPCR, DopEcR (CG18314), which can be activated by both ecdysone and the catecholamine, dopamine. We will also point out parallels between this system and the activation of the vertebrate GPCR, GPER1 (GPR30), which is thought to be activated by 17β-estradiol. We propose that the cellular localization and signalling properties of both DopEcR and GPER1 may be cell specific and depend upon their interactions with both accessory molecules and signalling pathways.
Collapse
Affiliation(s)
- Peter D Evans
- The Inositide Laboratory, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK.
| | - Asha Bayliss
- The Inositide Laboratory, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK.
| | - Vincenzina Reale
- The Inositide Laboratory, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK.
| |
Collapse
|
26
|
Srivastava DP, Evans PD. G-protein oestrogen receptor 1: trials and tribulations of a membrane oestrogen receptor. J Neuroendocrinol 2013; 25:1219-30. [PMID: 23822769 DOI: 10.1111/jne.12071] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Revised: 06/20/2013] [Accepted: 06/29/2013] [Indexed: 11/29/2022]
Abstract
Oestrogens are now recognised to be able to initiate rapid, fast responses, in addition to their classical, longer-term actions. There is a growing appreciation of the potential implications of this mode of action for oestrogenic signalling in both neuronal and non-neuronal systems. As such, much effort has been made to determine the mechanisms that are critical for transducing these rapid effects into cellular responses. Recently, an orphan G-protein-coupled receptor (GPCR), termed GPR30, was identified as an oestrogen-sensitive receptor in cancer cells. This receptor, now term G-protein oestrogen receptor 1 (GPER1) has been the subject of many investigations, and a role for this receptor in the nervous system is now emerging. In this review, we highlight some of the more recent advances in our understanding of the distribution and subcellular localisation of this receptor in the brain, as well as some of the evidence for the potential role that this receptor may play in the brain. We then discuss some of the controversies surrounding the pharmacology of this receptor, and attempt to reconcile these by suggesting that the 'agonist-specific coupling' model of GPCR function may provide a potential explanation for some of the divergent reports of GPER1 pharmacology.
Collapse
Affiliation(s)
- D P Srivastava
- Department of Neuroscience & Centre for the Cellular Basis of Behaviour, The James Black Centre, Institute of Psychiatry, King's College London, London, UK
| | | |
Collapse
|
27
|
Dimitrijević M, Stanojević S, Kuštrimović N, Mitić K, Vujić V, Aleksić I, Radojević K, Leposavić G. The influence of aging and estradiol to progesterone ratio on rat macrophage phenotypic profile and NO and TNF-α production. Exp Gerontol 2013; 48:1243-54. [PMID: 23850866 DOI: 10.1016/j.exger.2013.07.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Revised: 06/17/2013] [Accepted: 07/02/2013] [Indexed: 11/17/2022]
Abstract
The phenotype and function of tissue macrophages substantially depend on the cellular milieu and biological effector molecules, such as steroid hormones, to which they are exposed. Furthermore, in female rats, aging is associated with the altered macrophage functioning and the increased estrogen level is followed by a decrease in that of progesterone. Therefore, the present study aimed to investigate the influence of estradiol/progesterone balance on rat macrophage function and phenotype throughout whole adult lifespan. We ovariectomized rats at the late prepubertal age or at the very end of reproductive lifespan, and examined the expression of ED2 (CD163, a marker of mature resident macrophages related to secretion of inflammatory mediators) on peritoneal macrophages and their ability to produce TNF-α and NO upon LPS-stimulation at different age points. In addition, to delineate direct and indirect effects of estrogen, we assessed the in vitro influence of different concentrations of 17β-estradiol on LPS-induced macrophage TNF-α and NO production. Results showed that: (a) the low frequency of ED2(high) cells amongst peritoneal macrophages of aged rats was accompanied with the reduced TNF-α, but not NO production; (b) estradiol level gradually increased following ovariectomy; (c) macrophage ED2 expression and TNF-α production were dependent on estradiol/progesterone balance and they changed in the same direction; (d) changes in estradiol/progesterone balance differentially affected macrophages TNF-α and NO production; and (e) estradiol exerted pro-inflammatory and anti-inflammatory effects on macrophages in vivo and in vitro, respectively. Overall, our study discloses that estradiol/progesterone balance contributes to the fine-tuning of rat macrophage secretory capacity, and adds to a better understanding of the ovarian steroid hormone role in the regulation of macrophage function, and its significance for the age-associated changes in innate immunity.
Collapse
Affiliation(s)
- Mirjana Dimitrijević
- Institute of Virology, Vaccines and Sera, "Torlak", Immunology Research Center "Branislav Janković", Vojvode Stepe 458, 11152 Belgrade, Serbia.
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Indumathi D, Jayashree S, Selvaraj J, Sathish S, Mayilvanan C, Akilavalli N, Balasubramanian K. Effect of bisphenol-A on insulin signal transduction and glucose oxidation in skeletal muscle of adult male albino rat. Hum Exp Toxicol 2013; 32:960-71. [PMID: 23424204 DOI: 10.1177/0960327112470273] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The estrogenic monomer bisphenol-A (BPA) is an endocrine-disrupting chemical used in the production of epoxy resins, plastic food and beverage containers, leading to ubiquitous human exposure. Environmentally relevant doses of BPA have profound effects on mice endocrine pancreas. It increases pancreatic insulin content and favors postprandial hyperinsulinemia and insulin resistance in male mice. Skeletal muscle plays a crucial role in maintaining systemic glucose metabolism. In the present study, we investigated the possible effects of BPA on insulin-signaling molecules and glucose oxidation in skeletal muscle of male rat. Adult male Wistar albino rats were divided into three groups. Group I: control (vehicle treated) and groups II and III were administered with BPA orally (20 and 200 mg/kg bw/day, respectively). Although there was no change in the levels of insulin receptor (IR), Akt (protein kinase B) and glucose transporter-4 (GLUT4) messenger RNA, BPA significantly decreased the IR, Akt and GLUT4 protein levels (both plasma membrane and cytosolic fraction) of the gastrocnemius muscle. There was an increase in serum insulin and decrease in serum testosterone levels but fasting blood glucose level remained unaltered. In conclusion, BPA has adverse effects on phosphorylation of Akt, GLUT4 translocation and (14)C-glucose oxidation.
Collapse
Affiliation(s)
- D Indumathi
- Department of Endocrinology, Dr. ALM Post Graduate Institute of Basic Medical Sciences, University of Madras, Sekkizhar Campus, Taramani, Chennai, India
| | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
Protecting the functional mass of insulin-producing β cells of the pancreas is a major therapeutic challenge in patients with type 1 (T1DM) or type 2 diabetes mellitus (T2DM). The gonadal hormone 17β-oestradiol (E2) is involved in reproductive, bone, cardiovascular and neuronal physiology. In rodent models of T1DM and T2DM, treatment with E2 protects pancreatic β cells against oxidative stress, amyloid polypeptide toxicity, lipotoxicity and apoptosis. Three oestrogen receptors (ERs)--ERα, ERβ and the G protein-coupled ER (GPER)--have been identified in rodent and human β cells. Whereas activation of ERα enhances glucose-stimulated insulin biosynthesis, reduces islet toxic lipid accumulation and promotes β-cell survival from proapoptotic stimuli, activation of ERβ increases glucose-stimulated insulin secretion. However, activation of GPER protects β cells from apoptosis, raises glucose-stimulated insulin secretion and lipid homeostasis without affecting insulin biosynthesis. Oestrogens are also improving islet engraftment in rodent models of pancreatic islet transplantation. This Review describes developments in the role of ERs in islet insulin biosynthesis and secretion, lipid homeostasis and survival. Moreover, we discuss why and how enhancing ER action in β cells without the undesirable effect of general oestrogen therapy is a therapeutic avenue to preserve functional β-cell mass in patients with diabetes mellitus.
Collapse
Affiliation(s)
- Joseph P Tiano
- Feinberg School of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine and Comprehensive Center on Obesity, Northwestern University, Chicago, IL 60611, USA
| | | |
Collapse
|
30
|
Nilsson BO, Olde B, Leeb-Lundberg LMF. G protein-coupled oestrogen receptor 1 (GPER1)/GPR30: a new player in cardiovascular and metabolic oestrogenic signalling. Br J Pharmacol 2011; 163:1131-9. [PMID: 21250980 DOI: 10.1111/j.1476-5381.2011.01235.x] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Oestrogens are important sex hormones central to health and disease in both genders that have protective effects on the cardiovascular and metabolic systems. These hormones act in complex ways via both genomic and non-genomic mechanisms. The genomic mechanisms are relatively well characterized, whereas the non-genomic ones are only beginning to be explored. Two oestrogen receptors (ER), ERα and ERβ, have been described that act as nuclear transcription factors but can also associate with the plasma membrane and influence cytosolic signalling. ERα has been shown to mediate both anti-atherogenic effects and pro-survival effects in pancreatic β-cells. In recent years, a third membrane-bound ER has emerged, G protein-coupled receptor 30 or G protein-coupled oestrogen receptor 1 (GPER1), which mediates oestrogenic responses in cardiovascular and metabolic regulation. Both GPER1 knock-out models and pharmacological agents are now available to study GPER1 function. These tools have revealed that GPER1 activation may have several beneficial effects in the cardiovascular system including vasorelaxation, inhibition of smooth muscle cell proliferation, and protection of the myocardium against ischaemia/reperfusion injury, and in the metabolic system including stimulation of insulin release and protection against pancreatic β-cell apoptosis. Thus, GPER1 is emerging as a candidate therapeutic target in both cardiovascular and metabolic disease.
Collapse
Affiliation(s)
- Bengt-Olof Nilsson
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | | | |
Collapse
|
31
|
Abstract
Estrogens mediate profound effects throughout the body and regulate physiological and pathological processes in both women and men. The low prevalence of many diseases in premenopausal women is attributed to the presence of 17β-estradiol, the predominant and most potent endogenous estrogen. In addition to endogenous estrogens, several man-made and plant-derived molecules, such as bisphenol A and genistein, also exhibit estrogenic activity. Traditionally, the actions of 17β-estradiol are ascribed to two nuclear estrogen receptors (ERs), ERα and ERβ, which function as ligand-activated transcription factors. However, 17β-estradiol also mediates rapid signaling events via pathways that involve transmembrane ERs, such as G-protein-coupled ER 1 (GPER; formerly known as GPR30). In the past 10 years, GPER has been implicated in both rapid signaling and transcriptional regulation. With the discovery of GPER-selective ligands that can selectively modulate GPER function in vitro and in preclinical studies and with the use of Gper knockout mice, many more potential roles for GPER are being elucidated. This Review highlights the physiological roles of GPER in the reproductive, nervous, endocrine, immune and cardiovascular systems, as well as its pathological roles in a diverse array of disorders including cancer, for which GPER is emerging as a novel therapeutic target and prognostic indicator.
Collapse
Affiliation(s)
- Eric R Prossnitz
- Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
| | | |
Collapse
|
32
|
Abstract
OBJECTIVE The physiologic significance of the nitric oxide (NO)/cGMP signaling pathway in islets is unclear. We hypothesized that cGMP-dependent protein kinase type I (cGKI) is directly involved in the secretion of islet hormones and glucose homeostasis. RESEARCH DESIGN AND METHODS Gene-targeted mice that lack cGKI in islets (conventional cGKI mutants and cGKIα and Iβ rescue mice [α/βRM] that express cGKI only in smooth muscle) were studied in comparison to control (CTR) mice. cGKI expression was mapped in the endocrine pancreas by Western blot, immuno-histochemistry, and islet-specific recombination analysis. Insulin, glucagon secretion, and cytosolic Ca²(+) ([Ca²(+)](i)) were assayed by radioimmunoassay and FURA-2 measurements, respectively. Serum levels of islet hormones were analyzed at fasting and upon glucose challenge (2 g/kg) in vivo. RESULTS Immunohistochemistry showed that cGKI is present in α- but not in β-cells in islets of Langerhans. Mice that lack α-cell cGKI had significantly elevated fasting glucose and glucagon levels, whereas serum insulin levels were unchanged. High glucose concentrations strongly suppressed the glucagon release in CTR mice, but had only a moderate effect on islets that lacked cGKI. 8-Br-cGMP reduced stimulated [Ca²(+)](i) levels and glucagon release rates of CTR islets at 0.5 mmol/l glucose, but was without effect on [Ca²(+)](i) or hormone release in cGKI-deficient islets. CONCLUSIONS We propose that cGKI modulates glucagon release by suppression of [Ca²(+)](i) in α-cells.
Collapse
Affiliation(s)
- Veronika Leiss
- FOR 923, Technische Universität München, München, Germany, and Center for Integrated Protein Science, Ludwig-Maximilians-Universität München, München, Germany
- Institut für Pharmakologie und Toxikologie, Abteilung Pharmakologie und Experimentelle Therapie, Universitätsklinikum Tübingen, Tübingen, Germany
| | - Andreas Friebe
- Lehrstuhl für Physiologie I, Julius-Maximilians Universität Würzburg, Würzburg, Germany
| | - Andrea Welling
- FOR 923, Technische Universität München, München, Germany, and Center for Integrated Protein Science, Ludwig-Maximilians-Universität München, München, Germany
- Institut für Pharmakologie und Toxikologie, Technische Universität München, München, Germany
| | - Franz Hofmann
- FOR 923, Technische Universität München, München, Germany, and Center for Integrated Protein Science, Ludwig-Maximilians-Universität München, München, Germany
| | - Robert Lukowski
- FOR 923, Technische Universität München, München, Germany, and Center for Integrated Protein Science, Ludwig-Maximilians-Universität München, München, Germany
- Institut für Pharmazie, Abteilung Pharmakologie, Toxikologie und Klinische Pharmazie, Universität Tübingen, Tübingen, Germany
- Corresponding author: Robert Lukowski,
| |
Collapse
|
33
|
Stratton RC, Squires PE, Green AK. 17Beta-estradiol elevates cGMP and, via plasma membrane recruitment of protein kinase GIalpha, stimulates Ca2+ efflux from rat hepatocytes. J Biol Chem 2010; 285:27201-27212. [PMID: 20566641 DOI: 10.1074/jbc.m110.103630] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Rapid non-genomic effects of 17beta-estradiol, the principal circulating estrogen, have been observed in a wide variety of cell types. Here we investigate rapid signaling effects of 17beta-estradiol in rat hepatocytes. We show that, above a threshold concentration of 1 nm, 17beta-estradiol, but not 17alpha-estradiol, stimulates particulate guanylyl cyclase to elevate cGMP, which through activation and plasma membrane recruitment of protein kinase G isoform Ialpha, stimulates plasma membrane Ca(2+)-ATPase-mediated Ca(2+) efflux from rat hepatocytes. These effects are extremely rapid in onset and are mimicked by a membrane-impermeant 17beta-estradiol-BSA conjugate, suggesting that 17beta-estradiol acts at the extracellular face of the plasma membrane. We also show that 17beta-estradiol binds specifically to the intact hepatocyte plasma membrane through an interaction that is competed by an excess of atrial natriuretic peptide but also shows many similarities to the pharmacological characteristics of the putative gamma-adrenergic receptor. We, therefore, propose that the observed rapid signaling effects of 17beta-estradiol are mediated either through the guanylyl cyclase A receptor for atrial natriuretic peptide or through the gamma-adrenergic receptor, which is either itself a transmembrane guanylyl cyclase or activates a transmembrane guanylyl cyclase through cross-talk signaling.
Collapse
Affiliation(s)
- Rebecca C Stratton
- Department of Biological Sciences, The University of Warwick, Gibbet Hill Road, Coventry CV4 7AL, United Kingdom
| | - Paul E Squires
- Department of Biological Sciences, The University of Warwick, Gibbet Hill Road, Coventry CV4 7AL, United Kingdom
| | - Anne K Green
- Department of Biological Sciences, The University of Warwick, Gibbet Hill Road, Coventry CV4 7AL, United Kingdom.
| |
Collapse
|
34
|
Balhuizen A, Kumar R, Amisten S, Lundquist I, Salehi A. Activation of G protein-coupled receptor 30 modulates hormone secretion and counteracts cytokine-induced apoptosis in pancreatic islets of female mice. Mol Cell Endocrinol 2010; 320:16-24. [PMID: 20122988 DOI: 10.1016/j.mce.2010.01.030] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2009] [Revised: 01/21/2010] [Accepted: 01/21/2010] [Indexed: 12/22/2022]
Abstract
The role of the newly discovered estrogen receptor GPR30 in islet physiology and pathophysiology is unclear. We examined GPR30 expression in relation to hormone secretion and possible anti-apoptotic effects in isolated mouse islets using the synthetic GPR30 ligand G-1. The mRNA and protein expression of GPR30 was analyzed by qPCR, Western blot and confocal microscopy. Hormone secretion and cAMP content were determined with RIA and apoptosis in islet cells with the Annexin-V method. GPR30 mRNA and protein expression was markedly higher in islets from females compared to male. This gender difference was not found for the genomic estrogen receptors ER alpha and ER beta, the ER alpha expression being 10-fold higher than ER beta in both genders. Confocal microscopy revealed abounden GPR30 expression in insulin, glucagon and somatostatin cells. Dose-response studies of G-1 vs 17beta-estradiol in isolated islets at 1 or 12 mM glucose showed an almost identical pattern in that both compounds increased insulin and inhibited glucagon and somatostatin secretion. ICI-182,780 and EM-652, potent antagonists of the 17beta-estradiol receptors (ER alpha and ER beta) did not influence the amplifying effect of G-1 or 17beta-estradiol on cAMP content or insulin secretion from isolated islets. Cytokine-induced (IL-1 beta+TNFalpha+INF gamma) apoptosis in islets, cultured for 24h at 5mM glucose, was almost abolished by G-1 or 17beta-estradiol treatment. Addition of ICI-182,780 or EM-652 did not affect this beneficial effect of G-1 or 17beta-estradiol. Taken together, our findings show that GPR30 is expressed in most islet endocrine cells. The synthetic GPR30 ligand G-1 mimics the non-genomic effects of 17beta-estradiol on islet hormone secretion, cAMP content in islets and its anti-apoptotic effects. G-1 or analogs thereof might be new potential candidates in the therapeutic strategy for type 2 diabetes in women.
Collapse
Affiliation(s)
- Alexander Balhuizen
- Department of Clinical Science, Islet Cell Physiology, University of Lund, Sweden
| | | | | | | | | |
Collapse
|
35
|
Soriano S, Ropero AB, Alonso-Magdalena P, Ripoll C, Quesada I, Gassner B, Kuhn M, Gustafsson JA, Nadal A. Rapid regulation of K(ATP) channel activity by 17{beta}-estradiol in pancreatic {beta}-cells involves the estrogen receptor {beta} and the atrial natriuretic peptide receptor. Mol Endocrinol 2009; 23:1973-82. [PMID: 19855088 DOI: 10.1210/me.2009-0287] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The ATP-sensitive potassium (K(ATP)) channel is a key molecule involved in glucose-stimulated insulin secretion. The activity of this channel regulates beta-cell membrane potential, glucose- induced [Ca(2+)](i) signals, and insulin release. In this study, the rapid effect of physiological concentrations of 17beta-estradiol (E2) on K(ATP) channel activity was studied in intact beta-cells by use of the patch-clamp technique. When cells from wild-type (WT) mice were used, 1 nm E2 rapidly reduced K(ATP) channel activity by 60%. The action of E2 on K(ATP) channel was not modified in beta-cells from ERalpha-/- mice, yet it was significantly reduced in cells from ERbeta-/- mice. The effect of E2 was mimicked by the ERbeta agonist 2,3-bis(4-hydroxyphenyl)-propionitrile (DPN). Activation of ERbeta by DPN enhanced glucose-induced Ca(2+) signals and insulin release. Previous evidence indicated that the acute inhibitory effects of E2 on K(ATP) channel activity involve cyclic GMP and cyclic GMP-dependent protein kinase. In this study, we used beta-cells from mice with genetic ablation of the membrane guanylate cyclase A receptor for atrial natriuretic peptide (also called the atrial natriuretic peptide receptor) (GC-A KO mice) to demonstrate the involvement of this membrane receptor in the rapid E2 actions triggered in beta-cells. E2 rapidly inhibited K(ATP) channel activity and enhanced insulin release in islets from WT mice but not in islets from GC-A KO mice. In addition, DPN reduced K(ATP) channel activity in beta-cells from WT mice, but not in beta-cells from GC-A KO mice. This work unveils a new role for ERbeta as an insulinotropic molecule that may have important physiological and pharmacological implications.
Collapse
Affiliation(s)
- Sergi Soriano
- Institute of Bioengineering and CIBERDEM, Universidad Miguel Hernández de Elche, Alicante, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Olde B, Leeb-Lundberg LMF. GPR30/GPER1: searching for a role in estrogen physiology. Trends Endocrinol Metab 2009; 20:409-16. [PMID: 19734054 DOI: 10.1016/j.tem.2009.04.006] [Citation(s) in RCA: 160] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2009] [Revised: 04/17/2009] [Accepted: 04/21/2009] [Indexed: 12/24/2022]
Abstract
Estrogens are sex hormones that are central to health and disease in both genders. These hormones have long been recognized to act in complex ways, both through relatively slow genomic mechanisms and via fast non-genomic mechanisms. Several recent in vitro studies suggest that GPR30, or G protein-coupled estrogen receptor 1 (GPER1), is a functional membrane estrogen receptor involved in non-genomic estrogen signaling. However, this function is not universally accepted. Studies concerning the role of GPER1 in vivo are now beginning to appear but with divergent results. In this review we discuss current knowledge on the physiological role of GPER1 in the nervous system as well as in reproduction, metabolism, bone, and in the cardiovascular and immune systems.
Collapse
Affiliation(s)
- Björn Olde
- Unit of Drug Target Discovery, Department of Experimental Medical Science, Lund University, SE-22184 Lund, Sweden
| | | |
Collapse
|
37
|
Kim KH, Bender JR. Membrane-initiated actions of estrogen on the endothelium. Mol Cell Endocrinol 2009; 308:3-8. [PMID: 19549586 PMCID: PMC2701909 DOI: 10.1016/j.mce.2009.03.025] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2008] [Revised: 03/27/2009] [Accepted: 03/30/2009] [Indexed: 12/31/2022]
Abstract
Estrogen-induced rapid, membrane-initiated activation of numerous signal transduction cascades has been shown in animal, cellular and molecular vascular studies, which support the favorable effects of estrogen on vascular structure and function. These effects are mediated by distinct forms of estrogen receptor (ER) alpha. This includes estrogen-stimulated, rapid activation of endothelial nitric oxide synthase (eNOS), resulting in elaboration of the athero-protective, angiogenesis-promoting product nitric oxide (NO). An N-terminus truncated short isoform of ERalpha, ER46, plays a critical role in membrane-initiated, rapid responses to 17beta-estradiol (E2) in human endothelial cells (ECs). We have proposed a ER46-centered, eNOS-activating molecular complex in human EC caveolar membranes, containing c-Src, phosphatidylinositol 3-kinase (PI3K), Akt and eNOS. In this review, we describe estrogen-induced, rapid, non-genomic actions in the endothelium.
Collapse
Affiliation(s)
| | - Jeffrey R. Bender
- Corresponding author: Jeffrey R. Bender, Division of Cardiovascular Medicine and Departments of Internal Medicine and Immunobiology, Yale University School of Medicine, 300 Cedar Street, New Haven, Connecticut 06520, USA. Tel. 203-737-2223; Fax. 203-785-7567; E-Mail:
| |
Collapse
|
38
|
Nadal A, Alonso-Magdalena P, Soriano S, Ropero AB, Quesada I. The role of oestrogens in the adaptation of islets to insulin resistance. J Physiol 2009; 587:5031-7. [PMID: 19687125 DOI: 10.1113/jphysiol.2009.177188] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Pregnancy is characterized by peripheral insulin resistance, which is developed in parallel with a plasma increase of maternal hormones; these include prolactin, placental lactogens, progesterone and oestradiol among others. Maternal insulin resistance is counteracted by the adaptation of the islets of Langerhans to the higher insulin demand. If this adjustment is not produced, gestational diabetes may be developed. The adaptation process of islets is characterized by an increase of insulin biosynthesis, an enhanced glucose-stimulated insulin secretion (GSIS) and an increase of beta-cell mass. It is not completely understood why, in some individuals, beta-cell mass and function fail to adapt to the metabolic demands of pregnancy, yet a disruption of the beta-cell response to maternal hormones may play a key part. The role of the maternal hormone 17beta-oestradiol (E2) in this adaptation process has been largely unknown. However, in recent years, it has been demonstrated that E2 acts directly on beta-cells to increase insulin biosynthesis and to enhance GSIS through different molecular mechanisms. E2 does not increase beta-cell proliferation but it is involved in beta-cell survival. Classical oestrogen receptors ERalpha and ERbeta, as well as the G protein-coupled oestrogen receptor (GPER) seem to be involved in these adaptation changes. In addition, as the main production of E2 in post-menopausal women comes from the adipose tissue, E2 may act as a messenger between adipocytes and islets in obesity.
Collapse
Affiliation(s)
- Angel Nadal
- Instituto de Bioingeniería and CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Universidad Miguel Hernández de Elche, 03202 Elche, Alicante, Spain.
| | | | | | | | | |
Collapse
|
39
|
Ghanaat-Pour H, Sjöholm A. Gene expression regulated by pioglitazone and exenatide in normal and diabetic rat islets exposed to lipotoxicity. Diabetes Metab Res Rev 2009; 25:163-84. [PMID: 19065603 DOI: 10.1002/dmrr.896] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND Hyperlipidaemia has been suggested to contribute by pro-apoptotic actions to the loss of beta-cell mass, its secretory defects, and thereby impaired beta-cell function in type 2 diabetes. Treatment of genetically diabetic rats and also type 2 diabetic patients with pioglitazone, a PPAR-gamma agonist, lowers fasting levels of plasma glucose and triglycerides, and has been suggested to protect beta-cells against diabetic lipotoxicity in vitro and in vivo. Another recently launched anti-diabetic drug, exenatide, an incretin mimetic, has been shown to stimulate insulin secretion, growth, and proliferation of pancreatic beta-cells and to protect them against apoptosis. We aimed to investigate global alterations in beta-cell gene expression under lipotoxic conditions and the influence of in vitro treatment with pioglitazone and exenatide. METHODS Global gene expression profiling was thus performed to characterize genes differently regulated by palmitate, pioglitazone, and exenatide in isolated islets from non-diabetic Wistar rats and type 2 diabetic Goto-Kakizaki (GK) rats. RESULTS Gene expression profiling revealed significant changes in islet mRNAs involved in control of several aspects of beta-cell function, e.g. epigenetic regulation of gene expression, cell differentiation and morphogenesis, also metabolism, response to stimulus, transport, and signal transduction. Pioglitazone and exenatide appear to significantly impact epigenetic processes, e.g. stable alterations in gene expression potential, which arise during development and cell proliferation. Bcl2-like 1 (Bcl2l1), an anti-apoptotic protein, and Bcl2 modifying factor (Bmf), a pro-apoptotic protein, were both down-regulated by pioglitazone and exenatide in the presence of palmitate in diabetic GK islets. In contrast, Bmf was downregulated by pioglitazone in the presence of palmitate in non-diabetic Wistar islets. Exposure of non-diabetic Wistar islets to palmitate led to a reduction in the expression of PPAR beta/delta. This suggests that palmitate may increase the accumulation of triglycerides by reducing PPAR signalling. Moreover, treatment with either pioglitazone or exenatide restored and increased the expression of PPAR beta/delta in non-diabetic Wistar islets. CONCLUSIONS Taking into account that these drugs target different components of the epigenetic machinery, our findings suggest that they might participate in restoring normal gene activity in dysfunctional islets and that additive benefits may occur. Whether such events contribute to the beta-cell sparing, proliferative, and anti-apoptotic effects of these drugs in diabetes remains to be elucidated.
Collapse
Affiliation(s)
- Hamedeh Ghanaat-Pour
- Karolinska Institutet, Department of Internal Medicine, Stockholm South Hospital, Stockholm, Sweden
| | | |
Collapse
|
40
|
Mårtensson UEA, Salehi SA, Windahl S, Gomez MF, Swärd K, Daszkiewicz-Nilsson J, Wendt A, Andersson N, Hellstrand P, Grände PO, Owman C, Rosen CJ, Adamo ML, Lundquist I, Rorsman P, Nilsson BO, Ohlsson C, Olde B, Leeb-Lundberg LMF. Deletion of the G protein-coupled receptor 30 impairs glucose tolerance, reduces bone growth, increases blood pressure, and eliminates estradiol-stimulated insulin release in female mice. Endocrinology 2009; 150:687-98. [PMID: 18845638 DOI: 10.1210/en.2008-0623] [Citation(s) in RCA: 308] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
In vitro studies suggest that the G protein-coupled receptor (GPR) 30 is a functional estrogen receptor. However, the physiological role of GPR30 in vivo is unknown, and it remains to be determined whether GPR30 is an estrogen receptor also in vivo. To this end, we studied the effects of disrupting the GPR30 gene in female and male mice. Female GPR30((-/-)) mice had hyperglycemia and impaired glucose tolerance, reduced body growth, increased blood pressure, and reduced serum IGF-I levels. The reduced growth correlated with a proportional decrease in skeletal development. The elevated blood pressure was associated with an increased vascular resistance manifested as an increased media to lumen ratio of the resistance arteries. The hyperglycemia and impaired glucose tolerance in vivo were associated with decreased insulin expression and release in vivo and in vitro in isolated pancreatic islets. GPR30 is expressed in islets, and GPR30 deletion abolished estradiol-stimulated insulin release both in vivo in ovariectomized adult mice and in vitro in isolated islets. Our findings show that GPR30 is important for several metabolic functions in female mice, including estradiol-stimulated insulin release.
Collapse
Affiliation(s)
- Ulrika E A Mårtensson
- Units of Drug Target Discovery, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Ropero AB, Alonso-Magdalena P, Quesada I, Nadal A. The role of estrogen receptors in the control of energy and glucose homeostasis. Steroids 2008; 73:874-9. [PMID: 18249429 DOI: 10.1016/j.steroids.2007.12.018] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2007] [Accepted: 12/18/2007] [Indexed: 10/22/2022]
Abstract
Estrogens have been related to energy balance and glucose metabolism for a long time; however, the mechanisms involved in their actions are now being unveiled. The development of ERalpha and ERbeta knockout mice has demonstrated the participation of these receptors in the regulation of many processes related to the control of energy homeostasis. These include food intake and energy expenditure, insulin sensitivity in the liver and muscle, adipocyte growth and its body distribution as well as the pancreatic beta-cell function. In addition, other membrane receptors unrelated to ERalpha and ERbeta function in key tissues involved in energy balance and glucose homeostasis, i.e. the islet of Langerhans and the hypothalamus. Along with naturally occurring estrogens, there are endocrine disrupters that act as environmental estrogens and can impair the physiological action of ERalpha, ERbeta and other membrane ERs. New research is revealing a link between environmental estrogenic pollutants and the metabolic syndrome.
Collapse
Affiliation(s)
- Ana B Ropero
- Instituto de Bioingeniería, Universidad Miguel Hernández de Elche, Elche 03202, Alicante, Spain
| | | | | | | |
Collapse
|
42
|
Abstract
The definition of estrogen's actions has expanded from transcriptional regulation to the rapid, membrane-initiated activation of numerous signal transduction cascades. Multiple biological effects of estrogen have been shown in numerous animals, cellular and molecular studies, which support the favorable effects of estrogen on vascular structure, function, and cell signaling. Work from several laboratories has shown that these effects are mediated by distinct forms of estrogen receptor (ER) alpha. This includes estrogen-stimulated rapid activation of endothelial nitric oxide synthase (eNOS), resulting in the elaboration of the athero-protective, angiogenesis-promoting product nitric oxide (NO). We have described the expression of ER46, an N-terminus truncated isoform of the ERalpha, in human endothelial cells (EC), and its critical role in membrane-initiated, rapid responses to 17beta-estradiol (E2). We have proposed an ER46-centered, eNOS activating molecular complex in human EC caveolar membranes, containing c-Src, phosphatidylinositol 3-kinase (PI3K), Akt and eNOS. Our previous studies support estrogen-induced rapid eNOS activation via a sequential c-Src/PI3K/Akt cascade in EC. In this review, we describe estrogen-induced, rapid, non-genomic actions in endothelium, driven by c-Src-ER46-caveolin-1 interactions, with consequent activation of eNOS. Amidst ongoing controversies in hormone replacement therapy, these molecular and cellular data, defining favorable estrogenic effects on the endothelium, provide a strong impetus to resolve these clinical questions.
Collapse
Affiliation(s)
| | | | - Jeffrey R. Bender
- Corresponding author : Jeffrey R. Bender, Division of Cardiovascular Medicine and Departments of Internal Medicine and Immunobiology, Yale University School of Medicine, 300 Cedar Street, New Haven, Connecticut 06520, USA. Tel. 203-737-2223; Fax. 203-785-7567; E-Mail:
| |
Collapse
|
43
|
Milanesi L, de Boland AR, Boland R. Expression and localization of estrogen receptor α in the C2C12 murine skeletal muscle cell line. J Cell Biochem 2008; 104:1254-73. [DOI: 10.1002/jcb.21706] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
44
|
Thomas P. Characteristics of membrane progestin receptor alpha (mPRalpha) and progesterone membrane receptor component 1 (PGMRC1) and their roles in mediating rapid progestin actions. Front Neuroendocrinol 2008; 29:292-312. [PMID: 18343488 PMCID: PMC2600886 DOI: 10.1016/j.yfrne.2008.01.001] [Citation(s) in RCA: 270] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2008] [Accepted: 01/22/2008] [Indexed: 10/22/2022]
Abstract
Rapid, progestin actions initiated at the cell surface that are often nongenomic have been described in a variety of reproductive tissues, but until recently the identities of the membrane receptors mediating these nonclassical progestins actions remained unclear. Evidence has been obtained in the last 4-5 years for the involvement of two types of novel membrane proteins unrelated to nuclear steroid receptors, progesterone membrane receptors (mPRs) and progesterone receptor membrane component 1 (PGMRC1), in progestin signaling in several vertebrate reproductive tissues and in the brain. The mPRs, (M(W) approximately 40 kDa) initially discovered in fish ovaries, comprise at least three subtypes, alpha, beta and gamma and belong to the seven-transmembrane progesterone adiponectin Q receptor (PAQR) family. Both recombinant and wildtype mPRs display high affinity (K(d) approximately 5 nM), limited capacity, displaceable and specific progesterone binding. The mPRs are directly coupled to G proteins and typically activate pertussis-sensitive inhibitory G proteins (G(i)), to down-regulate adenylyl cyclase activity. Recent studies suggest the alpha subtype (mPRalpha) has important physiological functions in variety of reproductive tissues. The mPRalpha is an intermediary in progestin induction of oocyte maturation and stimulation of sperm hypermotility in fish. In mammals, the mPRalphas have been implicated in progesterone regulation of uterine function in humans and GnRH secretion in rodents. The single-transmembrane protein PGMRC1 (M(W) 26-28 kDa) was first purified from porcine livers and its cDNA was subsequently cloned from porcine smooth muscle cells and a variety of other tissues by different investigators. PGMRC1 and the closely-related PGMRC2 belong to the membrane-associated progesterone receptor (MAPR) family. The PGMRC1 protein displays moderately high binding affinity for progesterone which is 2- to 10-fold greater than that for testosterone and glucocorticoids, and also can bind to other molecules such as heme, cholesterol metabolites and proteins. The signal transduction pathways induced by binding of progesterone to PGMRC1 have not been described to date, although motifs for tyrosine kinase, kinase binding, SH2 and SH3 have been predicted from the amino acid sequence. Evidence has been obtained that PGMRC1 mediates the antiapoptotic affects of progesterone in rat granulosa cells. The PGMRC1 protein may also be an intermediary in the progesterone induction of the acrosome reaction in mammalian sperm. Despite these recent advances, many aspects of progestin signaling through these two families of novel membrane proteins remain unresolved. Biochemical characterization of the receptors has been hampered by rapid degradation of the partially purified proteins. A major technical challenge has been to express sufficient amounts of the recombinant receptors on the plasma membranes in eukaryotic systems to permit investigations of their progestin binding and signal transduction characteristics. Additional basic information on the molecular and cellular mechanisms by which mPRs and PGMRC1 interact with progestins, signal transductions pathways and other proteins will be required to establish a comprehensive model of nontraditional progestin actions mediated through these novel proteins.
Collapse
Affiliation(s)
- Peter Thomas
- University of Texas at Austin, Marine Science, 750 Channel View Drive, Port Aransas, TX 78373, USA.
| |
Collapse
|
45
|
Tudurí E, Filiputti E, Carneiro EM, Quesada I. Inhibition of Ca2+ signaling and glucagon secretion in mouse pancreatic alpha-cells by extracellular ATP and purinergic receptors. Am J Physiol Endocrinol Metab 2008; 294:E952-60. [PMID: 18349114 DOI: 10.1152/ajpendo.00641.2007] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Glucagon secreted from pancreatic alpha-cells plays a critical role in glycemia, mainly by hepatic glucose mobilization. In diabetic patients, an impaired control of glucagon release can worsen glucose homeostasis. Despite its importance, the mechanisms that regulate its secretion are still poorly understood. Since alpha-cells are particularly sensitive to neural and paracrine factors, in this report we studied the role of purinergic receptors and extracellular ATP, which can be released from nerve terminals and beta-cell secretory granules. Using immunocytochemistry, we identified in alpha-cells the P2 receptor subtype P2Y1, as well as the P1 receptors A1 and A2A. In contrast, only P2Y1 and A1 receptors were localized in beta-cells. To analyze the role of purinergic receptors in alpha-cell function, we studied their participation in Ca2+ signaling. At low glucose concentrations, mouse alpha-cells exhibited the characteristic oscillatory Ca2+ signals that lead to secretion. Application of ATP (1-10 microM) abolished these oscillations or reduced their frequency in alpha-cells within intact islets and isolated in culture. ATPgammaS, a nonhydrolyzable ATP derivative, indicated that the ATP effect was mainly direct rather than through ATP-hydrolytic products. Additionally, adenosine (1-10 microM) was also found to reduce Ca2+ signals. ATP-mediated inhibition of Ca2+ signaling was accompanied by a decrease in glucagon release from intact islets in contrast to the adenosine effect. Using pharmacological agonists, we found that only P2Y1 and A2A were likely involved in the inhibitory effect on Ca2+ signaling. All these findings indicate that extracellular ATP and purinergic stimulation are effective regulators of the alpha-cell function.
Collapse
Affiliation(s)
- Eva Tudurí
- Institute of Bioengineering, Miguel Hernandez University, Elche, Spain
| | | | | | | |
Collapse
|
46
|
Alonso-Magdalena P, Ropero AB, Carrera MP, Cederroth CR, Baquié M, Gauthier BR, Nef S, Stefani E, Nadal A. Pancreatic insulin content regulation by the estrogen receptor ER alpha. PLoS One 2008; 3:e2069. [PMID: 18446233 PMCID: PMC2323613 DOI: 10.1371/journal.pone.0002069] [Citation(s) in RCA: 287] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2008] [Accepted: 03/20/2008] [Indexed: 01/11/2023] Open
Abstract
The function of pancreatic beta-cells is the synthesis and release of insulin, the main hormone involved in blood glucose homeostasis. Estrogen receptors, ER alpha and ER beta, are important molecules involved in glucose metabolism, yet their role in pancreatic beta-cell physiology is still greatly unknown. In this report we show that both ER alpha and ER beta are present in pancreatic beta-cells. Long term exposure to physiological concentrations of 17beta-estradiol (E2) increased beta-cell insulin content, insulin gene expression and insulin release, yet pancreatic beta-cell mass was unaltered. The up-regulation of pancreatic beta-cell insulin content was imitated by environmentally relevant doses of the widespread endocrine disruptor Bisphenol-A (BPA). The use of ER alpha and ER beta agonists as well as ER alphaKO and ER betaKO mice suggests that the estrogen receptor involved is ER alpha. The up-regulation of pancreatic insulin content by ER alpha activation involves ERK1/2. These data may be important to explain the actions of E2 and environmental estrogens in endocrine pancreatic function and blood glucose homeostasis.
Collapse
Affiliation(s)
| | - Ana B. Ropero
- Instituto de Bioingeniería, Universidad Miguel Hernández de Elche, Alicante, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Alicante, Spain
| | - M. Pilar Carrera
- Instituto de Bioingeniería, Universidad Miguel Hernández de Elche, Alicante, Spain
| | - Christopher R. Cederroth
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
| | - Mathurin Baquié
- Department of Cell Physiology and Metabolism, University of Geneva Medical School, Geneva, Switzerland
| | - Benoit R. Gauthier
- Department of Cell Physiology and Metabolism, University of Geneva Medical School, Geneva, Switzerland
| | - Serge Nef
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
| | - Enrico Stefani
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, United States of America
- Department of Physiology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, United States of America
| | - Angel Nadal
- Instituto de Bioingeniería, Universidad Miguel Hernández de Elche, Alicante, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Alicante, Spain
- * E-mail:
| |
Collapse
|
47
|
Ho KJ, Liao JK. Non-nuclear actions of estrogen: new targets for prevention and treatment of cardiovascular disease. Mol Interv 2008; 2:219-28. [PMID: 14993393 PMCID: PMC2633129 DOI: 10.1124/mi.2.4.219] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Gender-based differences in the incidence of hypertensive and coronary artery disease, the development of atherosclerosis, and myocardial remodeling after infarction are attributable to the indirect effect of estrogen on risk factor profiles, such as cholesterol levels, glucose metabolism, and insulin levels. More recent evidence, however, suggests that activated estrogen receptor (ER) mediates signaling cascades that culminate in direct protective effects such as vasodilation, inhibition of response to vessel injury, limiting myocardial injury after infarction, and attenuating cardiac hypertrophy. Although the ER is usually thought of as a ligand-dependent transcription factor, it can also rapidly mobilize signals at the plasma membrane and in the cytoplasm. Thus, a greater understanding of ER function and regulation may lead to the development of highly specific therapeutics that mediate the prevention and treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Karen J Ho
- The Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, and Harvard Medical School, Cambridge, MA 02139, USA
| | | |
Collapse
|
48
|
Sandoval DA, Gong B, Davis SN. Antecedent short-term central nervous system administration of estrogen and progesterone alters counterregulatory responses to hypoglycemia in conscious male rats. Am J Physiol Endocrinol Metab 2007; 293:E1511-6. [PMID: 17940215 DOI: 10.1152/ajpendo.00340.2007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The aim of this study was to test the hypothesis that antecedent short-term administration of estradiol or progesterone into the central nervous system (CNS) reduces levels of neuroendocrine counterregulatory hormones during subsequent hypoglycemia. Conscious unrestrained male Sprague-Dawley rats were studied during randomized 2-day experiments. Day 1 consisted of an 8-h lateral ventricle infusion of estradiol (1 mug/mul; n = 9), progesterone (1 mug/mul; n = 9), or saline (0.2 mul/min; n = 10). On day 2, a 2-h hyperinsulinemic (30 pmol.kg(-1).min(-1)) hypoglycemic (2.9 +/- 0.2 mM) clamp was performed on all rats. Central administration of estradiol on day 1 resulted in significantly lower plasma epinephrine levels during hypoglycemia compared with saline, whereas central administration of progesterone resulted in increased levels of plasma norepinephrine and decreased levels of corticosterone both at baseline and during hypoglycemia. Glucagon responses during hypoglycemia were unaffected by prior administration of estradiol or progesterone. Endogenous glucose production following day 1 estradiol was significantly lower during day 2 hypoglycemia, and consequently, the glucose infusion rate to maintain the glycemia was significantly greater after estradiol administration compared with saline. These data suggest that 1) CNS administration of both female reproductive hormones can have rapid effects in modulating levels of counterregulatory hormones during subsequent hypoglycemia in conscious male rats, 2) forebrain administration of reproductive hormones can significantly reduce pituitary adrenal and sympathetic nervous system drive during hypoglycemia, 3) reproductive steroid hormones produce differential effects on sympathetic nervous system activity during hypoglycemia, and 4) reduction of epinephrine resulted in significantly blunted metabolic counterregulatory responses during hypoglycemia.
Collapse
Affiliation(s)
- Darleen A Sandoval
- Univ. of Cincinnati, 2170 E. Galbraith Rd., Bldg. E., Rm. 317, Cincinnati, OH 45237, USA.
| | | | | |
Collapse
|
49
|
Affiliation(s)
- David W Good
- Department of Medicine, University of Texas Medical Branch, Galveston 77555-0562, USA.
| |
Collapse
|
50
|
Quesada I, Todorova MG, Alonso-Magdalena P, Beltrá M, Carneiro EM, Martin F, Nadal A, Soria B. Glucose induces opposite intracellular Ca2+ concentration oscillatory patterns in identified alpha- and beta-cells within intact human islets of Langerhans. Diabetes 2006; 55:2463-9. [PMID: 16936194 DOI: 10.2337/db06-0272] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Homeostasis of blood glucose is mainly regulated by the coordinated secretion of glucagon and insulin from alpha- and beta-cells within the islets of Langerhans. The release of both hormones is Ca(2+) dependent. In the current study, we used confocal microscopy and immunocytochemistry to unequivocally characterize the glucose-induced Ca(2+) signals in alpha- and beta-cells within intact human islets. Extracellular glucose stimulation induced an opposite response in these two cell types. Although the intracellular Ca(2+) concentration ([Ca(2+)](i)) in beta-cells remained stable at low glucose concentrations, alpha-cells exhibited an oscillatory [Ca(2+)](i) response. Conversely, the elevation of extracellular glucose elicited an oscillatory [Ca(2+)](i) pattern in beta-cells but inhibited low-glucose-induced [Ca(2+)](i) signals in alpha-cells. These Ca(2+) signals were synchronic among beta-cells grouped in clusters within the islet, although they were not coordinated among the whole beta-cell population. The response of alpha-cells was totally asynchronic. Therefore, both the alpha- and beta-cell populations within human islets did not work as a syncitium in response to glucose. A deeper knowledge of alpha- and beta-cell behavior within intact human islets is important to better understand the physiology of the human endocrine pancreas and may be useful to select high-quality islets for transplantation.
Collapse
Affiliation(s)
- Ivan Quesada
- Institute of Bioengineering, Miguel Hernandez University, Ctra. N-332, Km. 87, 03550 Sant Joan d'Alacant, Spain.
| | | | | | | | | | | | | | | |
Collapse
|