1
|
Mariana SM, Brenda RP, Heriberto PG, Cristina L, David B, Guadalupe ÁL. GPER1 activation by estrogenic compounds in the inflammatory profile of breast cancer cells. J Steroid Biochem Mol Biol 2025; 245:106639. [PMID: 39571822 DOI: 10.1016/j.jsbmb.2024.106639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 10/28/2024] [Accepted: 11/19/2024] [Indexed: 11/24/2024]
Abstract
Breast cancer (BC) is the most frequent female neoplasm worldwide. Its establishment and development have been related to inflammatory cytokine expression. Steroid hormones such as estradiol (E2) can regulate proinflammatory cytokine secretion through interaction with its nuclear receptors. However, little is known regarding the activation of its membrane estrogen receptor (GPER1) and the inflammatory cytokine environment in BC. We have studied the synthesis and biological effects of molecules analogs to E2 for hormone replacement therapy (HRT), such as pentolame. Nevertheless, its interaction with GPER1 and the modulation of inflammatory cytokines in different BC types has been barely studied and deserves deeper investigation. In this research, the role of GPER1 in the proliferation and modulation of inflammatory cytokines involved in carcinogenesis and metastatic processes in different BC cell lines was assessed by binding to various compounds. To achieve this goal, the presence of GPER1 was identified in different BC cell lines. Subsequently, cell proliferation after exposure to E2, pentolame and GPER1 agonist, G1, was subsequently determined alone or in combination with the GPER1 antagonist, G15. Finally, the pro-inflammatory cytokine secretion derived from the supernatants of BC cells exposed to the previous treatments was also assessed. Interestingly, GPER1 activation or inhibition has significant effects on the cytokine regulation associated with invasion in BC. Notably, pentolame did not induce cell proliferation or increase the proinflammatory cytokine expression compared to E2 in BC cell lines. In addition, pentolame did not induce the presence of the cell adhesion molecule PECAM-1. In contrast, E2 treatment weakly induced the expression of PECAM-1 in MCF-7 and HCC1937 cells, and G1 treatment showed this effect only in MCF-7 cells. The results suggest that GPER1 might be a significant inflammatory modulator with angiogenic-related effects in BC cells. In addition, pentolame might represent an HRT alternative in patients with BC predisposition.
Collapse
Affiliation(s)
- Segovia-Mendoza Mariana
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico.
| | - Reyes-Plata Brenda
- Facultad de Estudios Superiores Zaragoza. Universidad Nacional Autónoma de México,Ciudad de México, Mexico
| | - Prado-Garcia Heriberto
- Laboratorio de Onco-Inmunobiología, Departamento de Enfermedades Crónico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias, "Ismael Cosio Villegas" Calzada de Tlalpan 4502, Col. Sección XVI, Ciudad de México 14080, Mexico
| | - Lemini Cristina
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Barrera David
- Departamento de Biología de la Reproducción "Dr. Carlos Gual Castro", Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Av. Vasco de Quiroga No. 15, Col. Belisario Domínguez, Sección XVI, Ciudad de México 14080, Mexico
| | - Ángeles-López Guadalupe
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico.
| |
Collapse
|
2
|
Gui Z, Shi W, Zhou F, Yan Y, Li Y, Xu Y. The role of estrogen receptors in intracellular estrogen signaling pathways, an overview. J Steroid Biochem Mol Biol 2025; 245:106632. [PMID: 39551163 DOI: 10.1016/j.jsbmb.2024.106632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 10/29/2024] [Accepted: 11/09/2024] [Indexed: 11/19/2024]
Abstract
To date five members of estrogen receptors (ESRs) have been reported. They are grouped into two classes, the nuclear estrogen receptors are members of the nuclear receptor family which found at nuclear, cytoplasm and plasma membrane, and the membrane estrogen receptors, such as G protein-coupled estrogen receptor 1, ESR-X and Gq-coupled membrane estrogen receptor. The structure and function of estrogen receptors, and interaction between ESR and coregulators were reviewed. In canonical pathway ESRs can translocate to the nucleus, bind to the target gene promotor with or without estrogen responsive element and regulate transcription, mediating the genomic effects of estrogen. Coactivators and corepressors are recruited to activate or inhibit transcription by activated ESRs. Many coactivators and corepressors are recruited to activate or inhibit ESR mediated gene transcription via different mechanisms. ESRs also indirectly bind to the promoter via interaction with other transcription factors, tethering the transcription factors. ESRs can be phosphorylated by several kinases such as p38, extracellular-signal-regulated kinase, and activated protein kinase B, and which activates transcription without ligand binding. Non-genomic estrogen action can be manifested by the increases of cytoplasmic NO and Ca2+ through the activation of membrane ESRs. In female, ESRs signaling is crucial for folliculogenesis, oocyte growth, ovulation, oviduct and uterus. In male, ESRs signaling modulates libido, erectile function, leydig cell steroidogenesis, sertoli cell's function, and epididymal fluid homeostatsis, supporting spermatogenesis and sperm maturation. The abnormal ESRs signaling is believed to be closely related to reproductive diseases and cancer.
Collapse
Affiliation(s)
- Zichang Gui
- College of Physical Education and Health, East China Normal University, Shanghai 200241, China.
| | - Wei Shi
- School of Chemistry, Biology and Environment, Yuxi Normal University, Yuxi 653100, China.
| | - Fangting Zhou
- School of Chemistry, Biology and Environment, Yuxi Normal University, Yuxi 653100, China.
| | - Yongqing Yan
- Yunnan Dasheng Biotechnology Co., LTD, Yuxi 653100, China.
| | - Yuntian Li
- School of Chemistry, Biology and Environment, Yuxi Normal University, Yuxi 653100, China.
| | - Yang Xu
- School of Chemistry, Biology and Environment, Yuxi Normal University, Yuxi 653100, China; Yunnan Dasheng Biotechnology Co., LTD, Yuxi 653100, China.
| |
Collapse
|
3
|
Ferriere F, Aasi N, Flouriot G, Pakdel F. Exploring the Complex Mechanisms of Isoflavones: From Cell Bioavailability, to Cell Dynamics and Breast Cancer. Phytother Res 2024. [PMID: 39707600 DOI: 10.1002/ptr.8417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 11/29/2024] [Accepted: 12/07/2024] [Indexed: 12/23/2024]
Abstract
In Western countries, the increase in the consumption of soy-derived products raises the population's exposure to isoflavones. These molecules, present in many foods, have numerous effects on the body's cells, including regulation of the transcription and epigenetics, cell signaling, cell cycle, cell growth, apoptosis, and oxidative stress. However, despite the multitude of studies conducted, on these compounds, it remains difficult to draw definitive conclusions regarding their safety or dangerousness in the diet. Indeed, some epidemiological studies highlight health benefits in consuming isoflavone-rich foods, notably by reducing the risk of certain cancers. However, several studies conducted on cell models show that these molecules can have negative effects on cell fate, particularly with regard to proliferation and survival of mammary tumor cells. Isoflavones are mainly genistein, daidzein, formononetin, and biochanin A. These molecules belong to the family of phytoestrogens, which are capable of interacting with both nuclear estrogen receptor, ERα and ERβ, to trigger agonistic and antagonistic effects. Due to their estrogenic properties, isoflavones are suspected to promote hormone-dependent cancers such as breast cancer. This suspicion is based primarily on their ability to bind to ERα in breast cells, thereby altering the signaling pathways that control cell growth. However, study results are sometimes contradictory. Some studies suggest that isoflavones may protect against breast cancer by acting as selective estrogen receptor modulators, while others highlight their potential role in stimulating tumor growth. This review explores the literature on the effects of isoflavones, focusing on their influence on ERα-dependent signaling in breast tumor cells.
Collapse
Affiliation(s)
- François Ferriere
- Université de Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, Rennes, France
| | - Nagham Aasi
- Université de Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, Rennes, France
| | - Gilles Flouriot
- Université de Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, Rennes, France
| | - Farzad Pakdel
- Université de Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, Rennes, France
| |
Collapse
|
4
|
Rižner TL, Gjorgoska M. Steroid sulfatase and sulfotransferases in the estrogen and androgen action of gynecological cancers: current status and perspectives. Essays Biochem 2024; 68:411-422. [PMID: 38994718 PMCID: PMC11625860 DOI: 10.1042/ebc20230096] [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: 04/02/2024] [Revised: 06/07/2024] [Accepted: 06/26/2024] [Indexed: 07/13/2024]
Abstract
Sulfatase (STS) and sulfotransferases (SULT) have important role in the biosynthesis and action of steroid hormones. STS catalyzes the hydrolysis of estrone-sulfate (E1-S) and dehydroepiandrosterone-sulfate (DHEA-S), while sulfotransferases catalyze the reverse reaction and require 3-phosphoadenosine-5-phosphosulfate as a sulfate donor. These enzymes control the concentration of active estrogens and androgens in peripheral tissues. Aberant expression of STS and SULT genes has been found in both, benign hormone-dependent diseases and hormone-dependent cancers. The aim of this review is to present the current knowledge on the role of STS and SULT in gynecological cancers, endometrial (EC) and ovarian cancer (OC). EC is the most common and OC the most lethal gynecological cancer. These cancers primarily affect postmenopausal women and therefore rely on the local production of steroid hormones from inactive precursors, either DHEA-S or E1-S. Following cellular uptake by organic anion transporting polypeptides (OATP) or organic anion transporters (OAT), STS and SULT regulate the formation of active estrogens and androgens, thus disturbed balance between STS and SULT can contribute to the onset and progression of cancer. The importance of these enzymes in peripheral estrogen biosynthesis has long been recognized, and this review provides new data on the important role of STS and SULT in the formation and action of androgens, their regulation and inhibition, and their potential as prognostic biomarkers.
Collapse
Affiliation(s)
- Tea Lanišnik Rižner
- Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Marija Gjorgoska
- Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
5
|
Gu H, Zou H, Cheng J, Liu X, Jiang Z, Peng P, Li F, Li B. Mechanism of programmed cell death in the posterior silk gland of the silkworm, Bombyx mori, during pupation based on Ca 2+ homeostasis. INSECT MOLECULAR BIOLOGY 2024; 33:551-559. [PMID: 38545681 DOI: 10.1111/imb.12911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 03/18/2024] [Indexed: 11/06/2024]
Abstract
The silkworm, Bombyx mori, is a complete metamorphosed economic insect, and the silk gland is a significant organ for silk protein synthesis and secretion. The silk gland completely degenerates during pupation, but the regulatory mechanism of programmed cell death (PCD) has not yet been understood. In the present study, we investigated the non-genetic pathway of 20E-induced PCD in the posterior silk gland (PSG) based on intracellular Ca2+ levels. Silk gland morphology and silk gland index indicated rapid degeneration of silk gland during metamorphosis from mature silkworm (MS) to pupal day 1 (P1), and Ca2+ levels within the PSG were found to peak during the pre-pupal day 1 (PP1) stage. Moreover, the results of autophagy and apoptosis levels within the PSG showed that autophagy was significantly increased in MS-PP1 periods, and significantly decreased in PP2 and P1 periods. Apoptosis was almost absent in MS-PP1 periods and significantly increased in PP2 and P1 periods. Additionally, western blotting results showed that autophagy preceded apoptosis, and the autophagy-promoting ATG5 was cleaved by calpain to the autophagy-inhibiting and apoptosis-promoting NtATG5 since PP1 period, while decreased autophagy was accompanied by increased apoptosis. Collectively, these findings suggest that Ca2+ is a key factor in the shift from autophagy to apoptosis.
Collapse
Affiliation(s)
- Haoyi Gu
- School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, China
| | - Hongbin Zou
- School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, China
| | - Jialu Cheng
- School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, China
| | - Xiaohan Liu
- School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, China
| | - Zhe Jiang
- School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, China
| | - Peilin Peng
- School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, China
| | - Fanchi Li
- School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, China
- Sericulture Institute of Soochow University, Soochow University, Suzhou, China
| | - Bing Li
- School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, China
- Sericulture Institute of Soochow University, Soochow University, Suzhou, China
| |
Collapse
|
6
|
de Assis Ramos MM, Ricardo-da-Silva FY, Macedo LDO, Correia CJ, Moreira LFP, Löbenberg R, Breithaupt-Faloppa AC, Bou-Chacra N. A review on lipid and polymeric nano-based 17-β-estradiol delivery systems: advances and challenges. JOURNAL OF PHARMACY & PHARMACEUTICAL SCIENCES : A PUBLICATION OF THE CANADIAN SOCIETY FOR PHARMACEUTICAL SCIENCES, SOCIETE CANADIENNE DES SCIENCES PHARMACEUTIQUES 2024; 27:13633. [PMID: 39619127 PMCID: PMC11604423 DOI: 10.3389/jpps.2024.13633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 10/25/2024] [Indexed: 12/13/2024]
Abstract
17β-estradiol (E2) is an endogenous steroid hormone pivotal for the development of female secondary sexual characteristics and the maintenance of the female reproductive system. Its roles extend beyond these physiological functions, as E2 is employed in hormone replacement therapy to alleviate symptoms associated with menopause. Furthermore, E2 exhibits therapeutic potential in the management of osteoporosis, breast cancer, and various neurological and cardiovascular conditions, partly due to its anti-inflammatory effects via modulation of the MAPK/NFκB signaling pathway. Notwithstanding, the hydrophobic nature of E2 significantly hinders the formulation of efficacious delivery systems for its clinical deployment. Recent advances have highlighted nano-based delivery systems for E2 as a promising solution to this solubility challenge. This review critically examines contemporary nano-delivery strategies for E2, particularly emphasizing lipid and polymeric nanoparticle-based systems. These nanostructures are designed to enhance stability, biocompatibility, controlled release, and targeted delivery of E2, yet the selectivity of E2 delivery for therapeutic purposes remains an ongoing challenge. The novelty of this review lies in its focus on the advances in nano-based E2 delivery systems over the past decade, a topic not extensively covered in prior literature. We present a comprehensive analysis of the encapsulation of E2 within polymeric and lipid nanoparticles, underscoring the untapped potential of these strategies. This review identifies a significant research gap, advocating for intensified experimental investigations that could pave the way for the translation of nano-based E2 therapies from bench to bedside.
Collapse
Affiliation(s)
- Mayara Munhoz de Assis Ramos
- Laboratório de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), Instituto do Coração (INCOR), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Fernanda Yamamoto Ricardo-da-Silva
- Laboratório de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), Instituto do Coração (INCOR), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Luiza de Oliveira Macedo
- Departamento de Farmácia, Faculdade de Ciências Farmacêuticas da Universidade de São Paulo, São Paulo, Brazil
| | - Cristiano Jesus Correia
- Laboratório de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), Instituto do Coração (INCOR), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Luiz Felipe Pinho Moreira
- Laboratório de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), Instituto do Coração (INCOR), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Raimar Löbenberg
- Division of Pharmaceutical Sciences, Faculty of Pharmacy and Pharmaceutical Sciences, Katz Group-Rexall Centre for Pharmacy and Health Research, University of Alberta, Edmonton, AB, Canada
| | - Ana Cristina Breithaupt-Faloppa
- Laboratório de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), Instituto do Coração (INCOR), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Nadia Bou-Chacra
- Departamento de Farmácia, Faculdade de Ciências Farmacêuticas da Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
7
|
Nishimura K, Ueta Y, Yoshino K. Estrogen-dependent oxytocin expression in the hypothalamus and estrogen-dependent vasopressin in the median eminence. J Obstet Gynaecol Res 2024; 50:2009-2018. [PMID: 39340151 DOI: 10.1111/jog.16100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024]
Abstract
The posterior pituitary (PP) hormones oxytocin (OXT) and arginine vasopressin (AVP) are synthesized within the hypothalamic nucleus and released from the PP into systemic circulation. Hypothalamic AVP projects its axons into the external layer of median eminence (eME) and regulates anterior pituitary hormone secretion during stress responses. Although similar as PP hormones, we demonstrate distinct regulatory roles of estrogen in hypothalamic OXT and AVP dynamics. OXT dynamics in the hypothalamus exhibit sex-dependent variations and that estrogen may influence dynamic OXT level changes, as observed in OXT-mRFP1 transgenic rats. Estrogen was also observed to modulate dynamic changes in AVP levels in the axon terminals of eME in female AVP-eGFP transgenic rats. Although OXT and AVP are produced within the similar hypothalamic region, both exhibit distinct dynamics within the hypothalamus. Estrogen acts on the hypothalamus, and further effects of estrogen replacement therapy can be expected.
Collapse
Affiliation(s)
- Kazuaki Nishimura
- Department of Obstetrics and Gynecology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Yoichi Ueta
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Kiyoshi Yoshino
- Department of Obstetrics and Gynecology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| |
Collapse
|
8
|
Takaya K, Kishi K. Ligustilide, A Novel Senolytic Compound Isolated from the Roots of Angelica Acutiloba. Adv Biol (Weinh) 2024; 8:e2300434. [PMID: 38183407 DOI: 10.1002/adbi.202300434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 12/14/2023] [Indexed: 01/08/2024]
Abstract
Senescent cells accumulate with age and contribute to age-related diseases and organ dysfunctions. Early evidence suggests that removal of senescent cells using senolytic drugs improves the aging phenotype in mice and may improve the health of individuals with chronic diseases. Signs of skin aging, including wrinkles, and sagging, occur largely due to the accumulation of senescent fibroblasts within the dermis; However, there is currently no skin treatment that eliminates senescent cells. In this study, human fibroblasts subjected to replicative aging and ionizing radiation exposure are used to screen plant extracts for potential senescent cell-destructive and/or senescent cell-forming activities. Angelica acutiloba-a traditional Chinese herbal medicine-selectively kills senescent cells without affecting the proliferating cells. Among the major components of this herb, ligustilide shows promising senescent cell-destructive properties, and selectively eliminates senescent cells by inducing an apoptosis. Moreover, ligustilide markedly inhibits senescence-associated secretory phenotypes. Administration of ligustilide to mouse skin eliminates senescent cells and increases dermal collagen density and subcutaneous adipose tissue content; it selectively promotes death of senescent cells without affecting non-senescent cells. These results provide evidence that a natural compound-ligustilide-may exhibit therapeutic effects on the skin aging phenotype by specifically inducing apoptosis in senescent cells.
Collapse
Affiliation(s)
- Kento Takaya
- Department of Plastic and Reconstructive Surgery, Keio University School of Medicine, Shinjuku, Tokyo, 160-8582, Japan
| | - Kazuo Kishi
- Department of Plastic and Reconstructive Surgery, Keio University School of Medicine, Shinjuku, Tokyo, 160-8582, Japan
| |
Collapse
|
9
|
Vilaça M, Lopes C, Seabra R, Rocha E. 17α-Ethynylestradiol and Levonorgestrel Exposure of Rainbow Trout RTL-W1 Cells at 18 °C and 21 °C Mainly Reveals Thermal Tolerance, Absence of Estrogenic Effects, and Progestin-Induced Upregulation of Detoxification Genes. Genes (Basel) 2024; 15:1189. [PMID: 39336780 PMCID: PMC11431550 DOI: 10.3390/genes15091189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/04/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
Fish are exposed to increased water temperatures and aquatic pollutants, including endocrine-disrupting compounds (EDCs). Although each stressor can disturb fish liver metabolism independently, combined effects may exist. To unveil the molecular mechanisms behind the effects of EDCs and temperature, fish liver cell lines are potential models needing better characterisation. Accordingly, we exposed the rainbow trout RTL-W1 cells (72 h), at 18 °C and 21 °C, to ethynylestradiol (EE2), levonorgestrel (LNG), and a mixture of both hormones (MIX) at 10 µM. The gene expression of a selection of targets related to detoxification (CYP1A, CYP3A27, GST, UGT, CAT, and MRP2), estrogen exposure (ERα, VtgA), lipid metabolism (FAS, FABP1, FATP1), and temperature stress (HSP70b) was analysed by RT-qPCR. GST expression was higher after LNG exposure at 21 °C than at 18 °C. LNG further enhanced the expression of CAT, while both LNG and MIX increased the expressions of CYP3A27 and MRP2. In contrast, FAS expression only increased in MIX, compared to the control. ERα, VtgA, UGT, CYP1A, HSP70b, FABP1, and FATP1 expressions were not influenced by the temperature or the tested EDCs. The RTL-W1 model was unresponsive to EE2 alone, sensitive to LNG (in detoxification pathway genes), and mainly insensitive to the temperature range but had the potential to unveil specific interactions.
Collapse
Affiliation(s)
- Margarida Vilaça
- Laboratory of Histology and Embryology, Department of Microscopy, ICBAS-School of Medicine and Biomedical Sciences, University of Porto (U.Porto), Rua Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
- Team of Animal Morphology and Toxicology, CIIMAR/CIMAR-Interdisciplinary Centre of Marine and Environmental Research, University of Porto (U.Porto), Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos s/n, 4450-208 Matosinhos, Portugal
| | - Célia Lopes
- Laboratory of Histology and Embryology, Department of Microscopy, ICBAS-School of Medicine and Biomedical Sciences, University of Porto (U.Porto), Rua Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
- Team of Animal Morphology and Toxicology, CIIMAR/CIMAR-Interdisciplinary Centre of Marine and Environmental Research, University of Porto (U.Porto), Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos s/n, 4450-208 Matosinhos, Portugal
| | - Rosária Seabra
- Laboratory of Histology and Embryology, Department of Microscopy, ICBAS-School of Medicine and Biomedical Sciences, University of Porto (U.Porto), Rua Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
- Team of Animal Morphology and Toxicology, CIIMAR/CIMAR-Interdisciplinary Centre of Marine and Environmental Research, University of Porto (U.Porto), Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos s/n, 4450-208 Matosinhos, Portugal
| | - Eduardo Rocha
- Laboratory of Histology and Embryology, Department of Microscopy, ICBAS-School of Medicine and Biomedical Sciences, University of Porto (U.Porto), Rua Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
- Team of Animal Morphology and Toxicology, CIIMAR/CIMAR-Interdisciplinary Centre of Marine and Environmental Research, University of Porto (U.Porto), Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos s/n, 4450-208 Matosinhos, Portugal
| |
Collapse
|
10
|
Fan C, Cowgill J, Howard RJ, Lindahl E. Divergent mechanisms of steroid inhibition in the human ρ1 GABA A receptor. Nat Commun 2024; 15:7795. [PMID: 39242530 PMCID: PMC11379708 DOI: 10.1038/s41467-024-51904-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 08/19/2024] [Indexed: 09/09/2024] Open
Abstract
ρ-type γ-aminobutyric acid-A (GABAA) receptors are widely distributed in the retina and brain, and are potential drug targets for the treatment of visual, sleep and cognitive disorders. Endogenous neuroactive steroids including β-estradiol and pregnenolone sulfate negatively modulate the function of ρ1 GABAA receptors, but their inhibitory mechanisms are not clear. By combining five cryo-EM structures with electrophysiology and molecular dynamics simulations, we characterize binding sites and negative modulation mechanisms of β-estradiol and pregnenolone sulfate at the human ρ1 GABAA receptor. β-estradiol binds in a pocket at the interface between extracellular and transmembrane domains, apparently specific to the ρ subfamily, and disturbs allosteric conformational transitions linking GABA binding to pore opening. In contrast, pregnenolone sulfate binds inside the pore to block ion permeation, with a preference for activated structures. These results illuminate contrasting mechanisms of ρ1 inhibition by two different neuroactive steroids, with potential implications for subtype-specific gating and pharmacological design.
Collapse
Affiliation(s)
- Chen Fan
- Dept. of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Solna, Sweden
- Dept. of Biochemistry and Biophysics, Science for Life Laboratory, Stockholm University, Solna, Sweden
| | - John Cowgill
- Dept. of Biochemistry and Biophysics, Science for Life Laboratory, Stockholm University, Solna, Sweden
| | - Rebecca J Howard
- Dept. of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Solna, Sweden.
- Dept. of Biochemistry and Biophysics, Science for Life Laboratory, Stockholm University, Solna, Sweden.
| | - Erik Lindahl
- Dept. of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Solna, Sweden.
- Dept. of Biochemistry and Biophysics, Science for Life Laboratory, Stockholm University, Solna, Sweden.
| |
Collapse
|
11
|
Niebora J, Woźniak S, Domagała D, Data K, Farzaneh M, Zehtabi M, Dari MAG, Pour FK, Bryja A, Kulus M, Mozdziak P, Dzięgiel P, Kempisty B. The role of ncRNAs and exosomes in the development and progression of endometrial cancer. Front Oncol 2024; 14:1418005. [PMID: 39188680 PMCID: PMC11345653 DOI: 10.3389/fonc.2024.1418005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 06/26/2024] [Indexed: 08/28/2024] Open
Abstract
Endometrial cancer (EC) is one of the most common gynecologic cancers. In recent years, research has focused on the genetic characteristics of the tumors to detail their prognosis and tailor therapy. In the case of EC, genetic mutations have been shown to underlie their formation. It is very important to know the mechanisms of EC formation related to mutations induced by estrogen, among other things. Noncoding RNAs (ncRNAs), composed of nucleotide transcripts with very low protein-coding capacity, are proving to be important. Their expression patterns in many malignancies can inhibit tumor formation and progression. They also regulate protein coding at the epigenetic, transcriptional, and posttranscriptional levels. MicroRNAs (miRNAs), several varieties of which are associated with normal endometrium as well as its tumor, also play a particularly important role in gene expression. MiRNAs and long noncoding RNAs (lncRNAs) affect many pathways in EC tissues and play important roles in cancer development, invasion, and metastasis, as well as resistance to anticancer drugs through mechanisms such as suppression of apoptosis and progression of cancer stem cells. It is also worth noting that miRNAs are highly precise, sensitive, and robust, making them potential markers for diagnosing gynecologic cancers and their progression. Unfortunately, as the incidence of EC increases, treatment becomes challenging and is limited to invasive tools. The prospect of using microRNAs as potential candidates for diagnostic and therapeutic use in EC seems promising. Exosomes are extracellular vesicles that are released from many types of cells, including cancer cells. They contain proteins, DNA, and various types of RNA, such as miRNAs. The noncoding RNA components of exosomes vary widely, depending on the physiology of the tumor tissue and the cells from which they originate. Exosomes contain both DNA and RNA and have communication functions between cells. Exosomal miRNAs mediate communication between EC cells, tumor-associated fibroblasts (CAFs), and tumor-associated macrophages (TAMs) and play a key role in tumor cell proliferation and tumor microenvironment formation. Oncogenes carried by tumor exosomes induce malignant transformation of target cells. During the synthesis of exosomes, various factors, such as genetic and proteomic data are upregulated. Thus, they are considered an interesting therapeutic target for the diagnosis and prognosis of endometrial cancer by analyzing biomarkers contained in exosomes. Expression of miRNAs, particularly miR-15a-5p, was elevated in exosomes derived from the plasma of EC patients. This may suggest the important utility of this biomarker in the diagnosis of EC. In recent years, researchers have become interested in the topic of prognostic markers for EC, as there are still too few identified markers to support the limited treatment of endometrial cancer. Further research into the effects of ncRNAs and exosomes on EC may allow for cancer treatment breakthroughs.
Collapse
Affiliation(s)
- Julia Niebora
- Division of Anatomy, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Sławomir Woźniak
- Division of Anatomy, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Dominika Domagała
- Division of Anatomy, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Krzysztof Data
- Division of Anatomy, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Maryam Farzaneh
- Fertility, Infertility and Perinatology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Clinical Research Development Unit, Imam Khomeini Hospital, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mojtaba Zehtabi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahrokh Abouali Gale Dari
- Department of Obstetrics and Gynecology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Fatemeh Khojasteh Pour
- Department of Obstetrics and Gynecology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Artur Bryja
- Division of Anatomy, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Magdalena Kulus
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, Torun, Poland
| | - Paul Mozdziak
- Physiology Graduate Program, North Carolina State University, Raleigh, NC, United States
| | - Piotr Dzięgiel
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Bartosz Kempisty
- Division of Anatomy, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, Torun, Poland
- Physiology Graduate Program, North Carolina State University, Raleigh, NC, United States
- Department of Obstetrics and Gynecology, University Hospital and Masaryk University, Brno, Czechia
| |
Collapse
|
12
|
Marolt N, Pavlič R, Kreft T, Gjogorska M, Rižner TL. Targeting estrogen metabolism in high-grade serous ovarian cancer shows promise to overcome platinum resistance. Biomed Pharmacother 2024; 177:117069. [PMID: 38968802 DOI: 10.1016/j.biopha.2024.117069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 06/26/2024] [Accepted: 06/27/2024] [Indexed: 07/07/2024] Open
Abstract
The high mortality rate due to chemoresistance in patients with high-grade ovarian cancer (HGSOC) emphasizes the urgent need to determine optimal treatment strategies for advanced and recurrent cases. Our study investigates the interplay between estrogens and chemoresistance in HGSOC and shows clear differences between platinum-sensitive and -resistant tumors. Through comprehensive transcriptome analyzes, we uncover differences in the expression of genes of estrogen biosynthesis, metabolism, transport and action underlying platinum resistance in different tissues of HGSOC subtypes and in six HGSOC cell lines. Furthermore, we identify genes involved in estrogen biosynthesis and metabolism as prognostic biomarkers for HGSOC. Additionally, our study elucidates different patterns of estrogen formation/metabolism and their effects on cell proliferation between six HGSOC cell lines with different platinum sensitivity. These results emphasize the dynamic interplay between estrogens and HGSOC chemoresistance. In particular, targeting the activity of steroid sulfatase (STS) proves to be a promising therapeutic approach with potential efficacy in limiting estrogen-driven cell proliferation. Our study reveals potential prognostic markers as well as identifies novel therapeutic targets that show promise for overcoming resistance and improving treatment outcomes in HGSOC.
Collapse
Affiliation(s)
- Nika Marolt
- Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana 1000, Slovenia
| | - Renata Pavlič
- Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana 1000, Slovenia
| | - Tinkara Kreft
- Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana 1000, Slovenia
| | - Marija Gjogorska
- Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana 1000, Slovenia
| | - Tea Lanišnik Rižner
- Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana 1000, Slovenia.
| |
Collapse
|
13
|
Dong H, Zeng X, Xu J, He C, Sun Z, Liu L, Huang Y, Sun Z, Cao Y, Peng Z, Qiu YA, Yu T. Advances in immune regulation of the G protein-coupled estrogen receptor. Int Immunopharmacol 2024; 136:112369. [PMID: 38824903 DOI: 10.1016/j.intimp.2024.112369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 04/12/2024] [Accepted: 05/27/2024] [Indexed: 06/04/2024]
Abstract
Estrogen and related receptors have been shown to have a significant impact on human development, reproduction, metabolism and immune regulation and to play a critical role in tumor development and treatment. Traditionally, the nuclear estrogen receptors (nERs) ERα and ERβ have been thought to be involved in mediating the estrogenic effects. However, our group and others have previously demonstrated that the G protein-coupled estrogen receptor (GPER) is the third independent ER, and estrogen signaling mediated by GPER is known to play an important role in normal physiology and a variety of abnormal diseases. Interestingly, recent studies have progressively revealed GPER involvement in the maintenance of the normal immune system, abnormal immune diseases, and inflammatory lesions, which may be of significant clinical value primarily in the immunotherapy of tumors. In this article, we review current advances in GPER-related immunomodulators and provide a theoretical basis and potential clinical targets to ameliorate immune-related diseases and immunotherapy for tumors.
Collapse
Affiliation(s)
- Hanzhi Dong
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330000, China
| | - Xiaoqiang Zeng
- Jiangxi Medical College, Nanchang University, Nanchang 330000, China
| | - Jiawei Xu
- Department of Breast Surgery, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Clinical Research Center for Cancer, Nanchang 330029, China
| | - Chongwu He
- Department of Breast Surgery, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Clinical Research Center for Cancer, Nanchang 330029, China
| | - Zhengkui Sun
- Department of Breast Surgery, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Clinical Research Center for Cancer, Nanchang 330029, China
| | - Liyan Liu
- Department of Pharmacy, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Clinical Research Center for Cancer, Nanchang 330029, China
| | - Yanxiao Huang
- Jiangxi Medical College, Nanchang University, Nanchang 330000, China
| | - Zhe Sun
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330000, China
| | - Yuan Cao
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330000, China
| | - Zhiqiang Peng
- Department of Lymphohematology, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Clinical Research Center for Cancer, Nanchang 330029, China.
| | - Yu-An Qiu
- Department of Critical Care Medicine, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Clinical Research Center for Cancer, Nanchang 330029, China.
| | - Tenghua Yu
- Department of Breast Surgery, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Clinical Research Center for Cancer, Nanchang 330029, China.
| |
Collapse
|
14
|
Zhang D, Chen H, Wang J, Ji J, Imam M, Zhang Z, Yan S. Current progress and prospects for G protein-coupled estrogen receptor in triple-negative breast cancer. Front Cell Dev Biol 2024; 12:1338448. [PMID: 38476263 PMCID: PMC10928007 DOI: 10.3389/fcell.2024.1338448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 02/08/2024] [Indexed: 03/14/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is a biologically and clinically heterogeneous disease. The G protein-coupled estrogen receptor (GPER) plays a crucial role in mediating the effect of estrogen and estrogen-like compounds in TNBC cells. Compared with other subtypes, GPER has a higher expression in TNBC. The GPER mechanisms have been thoroughly characterized and analyzed in estrogen receptor α (ERα) positive breast cancer, but not in TNBC. Our previous work revealed that a higher expression of GPER mRNA indicates a better prognosis for ERα-positive breast cancer; however, its effects in TNBC differ. Whether GPER could serve as a predictive prognostic marker or therapeutic target for TNBC remains unclear. In this review, we provide a detailed introduction to the subcellular localization of GPER, the different effects of various ligands, and the interactions between GPER and closely associated factors in TNBC. We focused on the internal molecular mechanisms specific to TNBC and thoroughly explored the role of GPER in promoting tumor development. We also discussed the interaction of GPER with specific cytokines and chemokines, and the relationship between GPER and immune evasion. Additionally, we discussed the feasibility of using GPER as a therapeutic target in the context of existing studies. This comprehensive review highlights the effects of GPER on TNBC, providing a framework and directions for future research.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Shunchao Yan
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
15
|
Sommer B, González-Ávila G, Flores-Soto E, Montaño LM, Solís-Chagoyán H, Romero-Martínez BS. Phytoestrogen-Based Hormonal Replacement Therapy Could Benefit Women Suffering Late-Onset Asthma. Int J Mol Sci 2023; 24:15335. [PMID: 37895016 PMCID: PMC10607548 DOI: 10.3390/ijms242015335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/10/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
It has been observed that plasmatic concentrations of estrogens, progesterone, or both correlate with symptoms in asthmatic women. Fluctuations in female sex steroid concentrations during menstrual periods are closely related to asthma symptoms, while menopause induces severe physiological changes that might require hormonal replacement therapy (HRT), that could influence asthma symptoms in these women. Late-onset asthma (LOA) has been categorized as a specific asthmatic phenotype that includes menopausal women and novel research regarding therapeutic alternatives that might provide relief to asthmatic women suffering LOA warrants more thorough and comprehensive analysis. Therefore, the present review proposes phytoestrogens as a promising HRT that might provide these females with relief for both their menopause and asthma symptoms. Besides their well-recognized anti-inflammatory and antioxidant capacities, phytoestrogens activate estrogen receptors and promote mild hormone-like responses that benefit postmenopausal women, particularly asthmatics, constituting therefore a very attractive potential therapy largely due to their low toxicity and scarce side effects.
Collapse
Affiliation(s)
- Bettina Sommer
- Departamento de Investigación en Hiperreactividad Bronquial, Instituto Nacional de Enfermedades Respiratorias ‘Ismael Cosio Villegas’, Calzada de Tlalpan 4502, Colonia Sección XVI, Mexico City CP 14080, Mexico
| | - Georgina González-Ávila
- Laboratorio de Oncología Biomédica, Departamento de Enfermedades Crónico Degenerativas, Instituto Nacional de Enfermedades Respiratorias ‘Ismael Cosio Villegas’, Mexico City CP 14080, Mexico;
| | - Edgar Flores-Soto
- Departmento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City CP 04510, Mexico; (E.F.-S.); (L.M.M.); (B.S.R.-M.)
| | - Luis M. Montaño
- Departmento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City CP 04510, Mexico; (E.F.-S.); (L.M.M.); (B.S.R.-M.)
| | - Héctor Solís-Chagoyán
- Neurociencia Cognitiva Evolutiva, Centro de Investigación en Ciencias Cognitivas, Universidad Autónoma del Estado de Morelos, Cuernavaca CP 62209, Morelos, Mexico;
| | - Bianca S. Romero-Martínez
- Departmento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City CP 04510, Mexico; (E.F.-S.); (L.M.M.); (B.S.R.-M.)
| |
Collapse
|
16
|
Anderson AP, Renn SCP. The Ancestral Modulation Hypothesis: Predicting Mechanistic Control of Sexually Heteromorphic Traits Using Evolutionary History. Am Nat 2023; 202:241-259. [PMID: 37606950 DOI: 10.1086/725438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2023]
Abstract
AbstractAcross the animal kingdom there are myriad forms within a sex across, and even within, species, rendering concepts of universal sex traits moot. The mechanisms that regulate the development of these trait differences are varied, although in vertebrates, common pathways involve gonadal steroid hormones. Gonadal steroids are often associated with heteromorphic trait development, where the steroid found at higher circulating levels is the one involved in trait development for that sex. Occasionally, there are situations in which a gonadal steroid associated with heteromorphic trait development in one sex is involved in heteromorphic or monomorphic trait development in another sex. We propose a verbal hypothesis, the ancestral modulation hypothesis (AMH), that uses the evolutionary history of the trait-particularly which sex ancestrally possessed higher trait values-to predict the regulatory pathway that governs trait expression. The AMH predicts that the genomic architecture appears first to resolve sexual conflict in an initially monomorphic trait. This architecture takes advantage of existing sex-biased signals, the gonadal steroid pathway, to generate trait heteromorphism. In cases where the other sex experiences evolutionary pressure for the new phenotype, that sex will co-opt the existing architecture by altering its signal to match that of the original high-trait-value sex. We describe the integrated levels needed to produce this pattern and what the expected outcomes will be given the evolutionary history of the trait. We present this framework as a testable hypothesis for the scientific community to investigate and to create further engagement and analysis of both ultimate and proximate approaches to sexual heteromorphism.
Collapse
|
17
|
Erdem C, Gross SM, Heiser LM, Birtwistle MR. MOBILE pipeline enables identification of context-specific networks and regulatory mechanisms. Nat Commun 2023; 14:3991. [PMID: 37414767 PMCID: PMC10326020 DOI: 10.1038/s41467-023-39729-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 06/27/2023] [Indexed: 07/08/2023] Open
Abstract
Robust identification of context-specific network features that control cellular phenotypes remains a challenge. We here introduce MOBILE (Multi-Omics Binary Integration via Lasso Ensembles) to nominate molecular features associated with cellular phenotypes and pathways. First, we use MOBILE to nominate mechanisms of interferon-γ (IFNγ) regulated PD-L1 expression. Our analyses suggest that IFNγ-controlled PD-L1 expression involves BST2, CLIC2, FAM83D, ACSL5, and HIST2H2AA3 genes, which were supported by prior literature. We also compare networks activated by related family members transforming growth factor-beta 1 (TGFβ1) and bone morphogenetic protein 2 (BMP2) and find that differences in ligand-induced changes in cell size and clustering properties are related to differences in laminin/collagen pathway activity. Finally, we demonstrate the broad applicability and adaptability of MOBILE by analyzing publicly available molecular datasets to investigate breast cancer subtype specific networks. Given the ever-growing availability of multi-omics datasets, we envision that MOBILE will be broadly useful for identification of context-specific molecular features and pathways.
Collapse
Affiliation(s)
- Cemal Erdem
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, SC, USA
| | - Sean M Gross
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
| | - Laura M Heiser
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA.
| | - Marc R Birtwistle
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, SC, USA.
- Department of Bioengineering, Clemson University, Clemson, SC, USA.
| |
Collapse
|
18
|
Alencar AKN, Swan KF, Pridjian G, Lindsey SH, Bayer CL. Connecting G protein-coupled estrogen receptor biomolecular mechanisms with the pathophysiology of preeclampsia: a review. Reprod Biol Endocrinol 2023; 21:60. [PMID: 37393260 DOI: 10.1186/s12958-023-01112-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 06/20/2023] [Indexed: 07/03/2023] Open
Abstract
BACKGROUND Throughout the course of pregnancy, small maternal spiral arteries that are in contact with fetal tissue undergo structural remodeling, lose smooth muscle cells, and become less responsive to vasoconstrictors. Additionally, placental extravillous trophoblasts invade the maternal decidua to establish an interaction between the fetal placental villi with the maternal blood supply. When successful, this process enables the transport of oxygen, nutrients, and signaling molecules but an insufficiency leads to placental ischemia. In response, the placenta releases vasoactive factors that enter the maternal circulation and promote maternal cardiorenal dysfunction, a hallmark of preeclampsia (PE), the leading cause of maternal and fetal death. An underexplored mechanism in the development of PE is the impact of membrane-initiated estrogen signaling via the G protein-coupled estrogen receptor (GPER). Recent evidence indicates that GPER activation is associated with normal trophoblast invasion, placental angiogenesis/hypoxia, and regulation of uteroplacental vasodilation, and these mechanisms could explain part of the estrogen-induced control of uterine remodeling and placental development in pregnancy. CONCLUSION Although the relevance of GPER in PE remains speculative, this review provides a summary of our current understanding on how GPER stimulation regulates some of the features of normal pregnancy and a potential link between its signaling network and uteroplacental dysfunction in PE. Synthesis of this information will facilitate the development of innovative treatment options.
Collapse
Affiliation(s)
| | - Kenneth F Swan
- Department of Obstetrics & Gynecology, Tulane University, New Orleans, LA, 70112, USA
| | - Gabriella Pridjian
- Department of Obstetrics & Gynecology, Tulane University, New Orleans, LA, 70112, USA
| | - Sarah H Lindsey
- Department of Pharmacology, Tulane University, New Orleans, LA, 70112, USA
| | - Carolyn L Bayer
- Department of Biomedical Engineering, Tulane University, 500 Lindy Boggs Center, New Orleans, LA, 70118, USA.
| |
Collapse
|
19
|
Yuan M, Chen S, Zeng C, Fan Y, Ge W, Chen W. Estrogenic and non-estrogenic effects of bisphenol A and its action mechanism in the zebrafish model: An overview of the past two decades of work. ENVIRONMENT INTERNATIONAL 2023; 176:107976. [PMID: 37236126 DOI: 10.1016/j.envint.2023.107976] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 04/11/2023] [Accepted: 05/10/2023] [Indexed: 05/28/2023]
Abstract
Bisphenol A (BPA) is the most simple and predominant component of the Bisphenol family. BPA is widely present in the environment and the human body as a result of its extensive usage in the plastic and epoxy resins of consumer goods like water bottles, food containers, and tableware. Since the 1930s, when BPA's estrogenic activity was first observed, and it was labeled as a "mimic hormone of E2", studies on the endocrine-disrupting effects of BPA then have been widely conducted. As a top vertebrate model for genetic and developmental studies, the zebrafish has caught tremendous attention in the past two decades. By using the zebrafish, the negative effects of BPA either through estrogenic signaling pathways or non-estrogenic signaling pathways were largely found. In this review, we tried to draw a full picture of the current state of knowledge on the estrogenic and non-estrogenic effects of BPA with their mechanisms of action through the zebrafish model of the past two decades, which may help to fully understand the endocrine-disrupting effects of BPA and its action mechanism, and give a direction for the future studies.
Collapse
Affiliation(s)
- Mingzhe Yuan
- Department of Biomedical Sciences and Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Shan Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Marine Sciences, Ningbo University, Ningbo 315211, China
| | - Chu Zeng
- Department of Biomedical Sciences and Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Yuqin Fan
- Guangdong Provincial Key Laboratory of Conservation and Precision Utilization of Characteristic Agricultural Resources in Mountainous Area, School of Life Sciences, Jiaying University, Meizhou 514015, China
| | - Wei Ge
- Department of Biomedical Sciences and Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau, China.
| | - Weiting Chen
- Guangdong Provincial Key Laboratory of Conservation and Precision Utilization of Characteristic Agricultural Resources in Mountainous Area, School of Life Sciences, Jiaying University, Meizhou 514015, China.
| |
Collapse
|
20
|
Non-genomic Effect of Estradiol on the Neurovascular Unit and Possible Involvement in the Cerebral Vascular Accident. Mol Neurobiol 2023; 60:1964-1985. [PMID: 36596967 DOI: 10.1007/s12035-022-03178-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 12/16/2022] [Indexed: 01/05/2023]
Abstract
Cerebrovascular diseases, such as ischemic cerebral vascular accident (CVA), are responsible for causing high rates of morbidity, mortality, and disability in the population. The neurovascular unit (NVU) during and after ischemic CVA plays crucial roles in cell regulation and preservation, the immune and inflammatory response, and cell and/or tissue survival and repair. Cellular responses to 17β-estradiol (E2) can be triggered by two mechanisms: one called classical or genomic, which is due to the activation of the "classical" nuclear estrogen receptors α (ERα) and β (ERβ), and the non-genomic or rapid mechanism, which is due to the activation of the G protein-coupled estrogen receptor 1 (GPER) that is located in the plasma membrane and some in intracellular membranes, such as in the Golgi apparatus and endoplasmic reticulum. Nuclear receptors can regulate gene expression and cellular functions. On the contrary, activating the GPER by E2 and/or its G-1 agonist triggers several rapid cell signaling pathways. Therefore, E2 or its G-1 agonist, by mediating GPER activation and/or expression, can influence several NVU cell types. Most studies argue that the activation of the GPER may be used as a potential therapeutic target in various pathologies, such as CVA. Thus, with this review, we aimed to summarize the existing literature on the role of GPER mediated by E2 and/or its agonist G-1 in the physiology and pathophysiology of NVU.
Collapse
|
21
|
Yang X, Jiang H, Ning J, Zhang S, Cai Y, Wang L, Yang J, Xu G, Chen W, Wang J. Inhibition of GPR30 sensitized gefitinib to NSCLC cells via regulation of epithelial-mesenchymal transition. Int J Immunopathol Pharmacol 2023; 37:3946320231210737. [PMID: 37890097 PMCID: PMC10612443 DOI: 10.1177/03946320231210737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
Introduction: G-protein coupled receptor 30 (GPR30) is associated with cell metastasis and drug resistance in many different cancer cells. The present study aimed to reveal the sensitivity of GPR30 to gefitinib in non-small cell lung cancer (NSCLC) cells.Methods: Cell viability and proliferation were detected using cell counting kit 8 and 5-ethynyl-2'-deoxyuridine assays, respectively. Western blotting and quantitative real-time reverse transcription PCR were used to detect GPR30 or epithelial-mesenchyme transition (EMT)-related mRNA and protein expression.Results: The results showed that GPR30 expression is associated with gefitinib sensitivity. G15, as a GPR30 antagonist, reduced GPR30 expression. We chose the maximum concentration of G15 with minimal cytotoxicity to detect cell viability after combined treatment with gefitinib in NSCLC cells, which indicated that G15 could increase sensitivity to gefitinib. However, the effect of G15 on gefitinib sensitivity disappeared after treatment with a small interfering RNA targeting GPR30. Further research showed that G15 or GPR30 siRNA treatment could upregulate E-cadherin and downregulate vimentin levels.Conclusion: Taken together, these data suggested that G15 could enhance NSCLC sensitivity to gefitinib by inhibition of GPR30 and EMT.
Collapse
Affiliation(s)
- Xiaomin Yang
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Hongyan Jiang
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jiang Ning
- Department of Pharmacy, Zi Yang Street Community Health Service Center, Hangzhou, Zhejiang, China
| | - Shufen Zhang
- Cancer Institute of Integrated Traditional Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Ying Cai
- Cancer Institute of Integrated Traditional Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Liang Wang
- The Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Jinsong Yang
- Department of Radiation Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Guodong Xu
- Department of Cardiothoracic Surgery, The Affiliated Lihuili Hospital, Ningbo University, Ningbo, China
| | - Wei Chen
- Cancer Institute of Integrated Traditional Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Jianfei Wang
- Department of Pharmacy, Tongxiang Third People's Hospital, Tongxiang, Zhejiang, China
| |
Collapse
|
22
|
Luo W, Yan Y, Cao Y, Zhang Y, Zhang Z. The effects of GPER on age-associated memory impairment induced by decreased estrogen levels. Front Mol Biosci 2023; 10:1097018. [PMID: 37021109 PMCID: PMC10069632 DOI: 10.3389/fmolb.2023.1097018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 02/08/2023] [Indexed: 04/07/2023] Open
Abstract
Estrogen, as a pleiotropic endocrine hormone, not only regulates the physiological functions of peripheral tissues but also exerts vital neuroregulatory effects in the central nervous system (CNS), such as the development of neurons and the formation of neural network connections, wherein rapid estrogen-mediated reactions positively stimulate spinogenesis and regulate synaptic plasticity and synaptic transmission to facilitate cognitive and memory performance. These fast non-genomic effects can be initiated by membrane-bound estrogen receptors (ERs), three best known of which are ERα, ERβ, and G protein-coupled estrogen receptor (GPER). To date, the effects of ERα and ERβ have been well studied in age-associated memory impairment, whereas there is still a lack of attention to the role of GPER in age-associated memory impairment, and there are still disputes about whether GPER indeed functions as an ER to enhance learning and memory. In this review, we provide a systematic overview of the role of GPER in age-associated memory impairment based on its expression, distribution, and signaling pathways, which might bring some inspiration for translational drugs targeting GPER for age-related diseases and update knowledge on the role of estrogen and its receptor system in the brain.
Collapse
Affiliation(s)
- Wenyu Luo
- Department of Ultrasound, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yudie Yan
- Department of Ultrasound, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yunpeng Cao
- Department of Neurology, The First Hospital of China Medical University, Shenyang, Liaoning, China
- *Correspondence: Zhen Zhang, ; Yunpeng Cao, ; Yanbo Zhang,
| | - Yanbo Zhang
- Department of Psychiatry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- *Correspondence: Zhen Zhang, ; Yunpeng Cao, ; Yanbo Zhang,
| | - Zhen Zhang
- Department of Ultrasound, The First Hospital of China Medical University, Shenyang, Liaoning, China
- *Correspondence: Zhen Zhang, ; Yunpeng Cao, ; Yanbo Zhang,
| |
Collapse
|
23
|
Ardevines S, Marqués-López E, Herrera RP. Heterocycles in Breast Cancer Treatment: The Use of Pyrazole Derivatives. Curr Med Chem 2023; 30:1145-1174. [PMID: 36043746 PMCID: PMC11475274 DOI: 10.2174/0929867329666220829091830] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 04/21/2022] [Accepted: 05/05/2022] [Indexed: 11/22/2022]
Abstract
Among the aromatic heterocycle rings, pyrazole -a five-membered ring with two adjacent nitrogen atoms in its structure has been postulated as a potent candidate in the pharmacological context. This moiety is an interesting therapeutic target covering a broad spectrum of biological activities due to its presence in many natural substances. Hence, the potential of the pyrazole derivatives as antitumor agents has been explored in many investigations, showing promising results in some cases. In this sense, breast cancer, which is already the leading cause of cancer mortality in women in some countries, has been the topic selected for this review, which covers a range of different research from the earliest studies published in 2003 to the most recent ones in 2021.
Collapse
Affiliation(s)
- Sandra Ardevines
- Laboratorio de Organocatálisis Asimétrica, Departamento de Química Orgánica. Instituto de Síntesis Química y Catálisis Homogénea (ISQCH), CSIC-Universidad de Zaragoza. C/Pedro Cerbuna 12, E-50009 Zaragoza, Spain
| | - Eugenia Marqués-López
- Laboratorio de Organocatálisis Asimétrica, Departamento de Química Orgánica. Instituto de Síntesis Química y Catálisis Homogénea (ISQCH), CSIC-Universidad de Zaragoza. C/Pedro Cerbuna 12, E-50009 Zaragoza, Spain
| | - Raquel P. Herrera
- Laboratorio de Organocatálisis Asimétrica, Departamento de Química Orgánica. Instituto de Síntesis Química y Catálisis Homogénea (ISQCH), CSIC-Universidad de Zaragoza. C/Pedro Cerbuna 12, E-50009 Zaragoza, Spain
| |
Collapse
|
24
|
Gravitte A, Kintner J, Brown S, Cobble A, Kennard B, Hall JV. The hormonal environment and estrogen receptor signaling alters Chlamydia muridarum infection in vivo. Front Cell Infect Microbiol 2022; 12:939944. [PMID: 36636722 PMCID: PMC9831676 DOI: 10.3389/fcimb.2022.939944] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 11/29/2022] [Indexed: 12/28/2022] Open
Abstract
Genital Chlamydia is the most common bacterial sexually transmitted infection in the United States and worldwide. Previous studies indicate that the progression of chlamydial infection is influenced by various factors, including the female sex hormones estrogen and progesterone. Sex hormone levels naturally fluctuate in women throughout their menstrual cycle. Varying concentrations of estrogen and progesterone may impact the progression of chlamydial infection and the host's immune response to Chlamydia. Estrogen signals through estrogen receptors (ERs), ERα and ERβ. These receptors are similar in structure and function, but are differentially expressed in tissues throughout the body, including the genital tract and on cells of the immune system. In this study, we used ovariectomized (OVT) BALB/c mice to investigate the impact of long-term administration of physiologically relevant concentrations of estrogen (E2), progesterone (P4), or a combination of E2/P4 on the progression of and immune response to C. muridarum infection. Additionally, we used ERα and ERβ knockout C57/BL6 mice to determine the how ERs affect chlamydial infection and the resulting immune response. Estrogen exposure prevented C. muridarum infection in vaginally infected OVT mice exposed to E2 alone or in combination with P4, while OVT or Sham mice exposed to hormone free, P4 or depo-medroxyprogesterone acetate shed similar amounts of chlamydiae. The hormonal environment also altered T cell recruitment and IFNϵ production the genital tracts of infected OVT and Sham mice on day 10 post infection. The absence of ERα, but not ERβ, in ER knockout mouse strains significantly changed the timing of C. muridarum infection. ERαKO mice shed significantly more chlamydiae at day 3 post infection and resolved the infection faster than WT or ERβKO animals. At day 9 post infection, flow cytometry showed that ERαKO mice had more T cells present and targeted RNA sequencing revealed increased expression of CD4 and FOXP3, suggesting that ERαKO mice had increased numbers of regulatory T cells compared to ERβKO and WT mice. Mock and chlamydia-infected ERαKO mice also expressed more IFNϵ early during infection. Overall, the data from these studies indicate that sex hormones and their receptors, particularly ERα and ERβ, differentially affect C. muridarum infection in murine models of infection.
Collapse
Affiliation(s)
- Amy Gravitte
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States,Center of Excellence for Inflammation, Infection Disease, and Immunity, East Tennessee State University, Johnson City, TN, United States
| | - Jennifer Kintner
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Stacy Brown
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN, United States
| | - Allison Cobble
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN, United States
| | - Benjamin Kennard
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN, United States
| | - Jennifer V. Hall
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States,Center of Excellence for Inflammation, Infection Disease, and Immunity, East Tennessee State University, Johnson City, TN, United States,*Correspondence: Jennifer V. Hall,
| |
Collapse
|
25
|
Khan MZI, Uzair M, Nazli A, Chen JZ. An overview on Estrogen receptors signaling and its ligands in breast cancer. Eur J Med Chem 2022; 241:114658. [PMID: 35964426 DOI: 10.1016/j.ejmech.2022.114658] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 07/31/2022] [Accepted: 08/01/2022] [Indexed: 02/07/2023]
Abstract
Estrogen governs the regulations of various pathological and physiological actions throughout the body in both males and females. Generally, 17β-estradiol an endogenous estrogen is responsible for different health problems in pre and postmenopausal women. The major activities of endogenous estrogen are executed by nuclear estrogen receptors (ERs) ERα and ERβ while non-genomic cytoplasmic pathways also govern cell growth and apoptosis. Estrogen accomplished a fundamental role in the formation and progression of breast cancer. In this review, we have hyphenated different studies regarding ERs and a thorough and detailed study of estrogen receptors is presented. This review highlights different aspects of estrogens ranging from receptor types, their isoforms, structures, signaling pathways of ERα, ERβ and GPER along with their crystal structures, pathological roles of ER, ER ligands, and therapeutic strategies to overcome the resistance.
Collapse
Affiliation(s)
| | - Muhammad Uzair
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450001, China
| | - Adila Nazli
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Jian-Zhong Chen
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China.
| |
Collapse
|
26
|
Berridge CW, Martin AJ, Hupalo S, Nicol SE. Estrus cycle-dependent working memory effects of prefrontal cortex corticotropin-releasing factor neurotransmission. Neuropsychopharmacology 2022; 47:2016-2023. [PMID: 35618840 PMCID: PMC9556710 DOI: 10.1038/s41386-022-01349-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/05/2022] [Accepted: 05/18/2022] [Indexed: 11/08/2022]
Abstract
The prefrontal cortex (PFC) supports a diversity of cognitive processes. Impairment in PFC-dependent cognition is associated with multiple psychiatric disorders, including those known to display sex differences. Our ability to treat this impairment is limited, in part due to an incomplete understanding of the neural mechanisms that support PFC-dependent cognition. In previous studies in male rats, we demonstrated that corticotropin-releasing factor (CRF) receptors and neurons in caudal dorsomedial PFC (dmPFC) regulate PFC-dependent working memory. Subcortically, CRF can exert sex-specific actions, a subset of which are ovarian steroid dependent. To date, the cognitive actions of dmPFC CRF neurotransmission in females are unknown. To address this gap, the current studies examined the effects of chemogenetic and pharmacological manipulations of CRF receptors and neurons within the dmPFC of female rats tested in a spatial working memory task. Outside of proestrus, activation of both CRF receptors and neurons in the caudal, but not rostral, dmPFC impaired working memory. Meanwhile, blockade of CRF receptors in the caudal dmPFC or globally in the brain, improved working memory performance, similar to that seen in males. In contrast, these effects were not observed during proestrus. These observations demonstrate that while CRF neurotransmission in the PFC regulates working memory similarly in males and females, these actions are not observed in females when ovarian steroids are at peak levels.
Collapse
Affiliation(s)
- Craig W Berridge
- Department of Psychology, University of Wisconsin-Madison, Madison, WI, 53706, USA.
| | - Andrea J Martin
- Department of Psychology, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Sofiya Hupalo
- Department of Psychology, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Division of Neuroscience and Basic Behavioral Science, National Institute of Mental Health, Bethesda, MD, 20892, USA
| | - Shannon E Nicol
- Department of Psychology, University of Wisconsin-Madison, Madison, WI, 53706, USA
| |
Collapse
|
27
|
Oestrogen-dependent hypothalamic oxytocin expression with changes in feeding and body weight in female rats. Commun Biol 2022; 5:912. [PMID: 36064966 PMCID: PMC9445083 DOI: 10.1038/s42003-022-03889-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 08/25/2022] [Indexed: 11/08/2022] Open
Abstract
Oxytocin (OXT) is produced in the hypothalamic nuclei and secreted into systemic circulation from the posterior pituitary gland. In the central nervous system, OXT regulates behaviours including maternal and feeding behaviours. Our aim is to evaluate whether oestrogen regulates hypothalamic OXT dynamics. Herein, we provide the first evidence that OXT dynamics in the hypothalamus vary with sex and that oestrogen may modulate dynamic changes in OXT levels, using OXT-mRFP1 transgenic rats. The fluorescence intensity of OXT-mRFP1 and expression of the OXT and mRFP1 genes in the hypothalamic nuclei is highest during the oestrus stage in female rats and decreased significantly in ovariectomised rats. Oestrogen replacement caused significant increases in fluorescence intensity and gene expression in a dose-related manner. This is also demonstrated in the rats' feeding behaviour and hypothalamic Fos neurons using cholecystokinin-8 and immunohistochemistry. Hypothalamic OXT expression is oestrogen-dependent and can be enhanced centrally by the administration of oestrogen.
Collapse
|
28
|
Rahman MS, Pang WK, Amjad S, Ryu DY, Adegoke EO, Park YJ, Pang MG. Hepatic consequences of a mixture of endocrine-disrupting chemicals in male mice. JOURNAL OF HAZARDOUS MATERIALS 2022; 436:129236. [PMID: 35739755 DOI: 10.1016/j.jhazmat.2022.129236] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/22/2022] [Accepted: 05/23/2022] [Indexed: 06/15/2023]
Abstract
The global epidemic of metabolic syndrome has been partially linked to ubiquitous exposure to endocrine-disrupting chemicals (EDCs). Although the impacts of exposure to single EDCs have been thoroughly studied, the consequences of simultaneous uncontrolled exposure to multiple EDCs require further investigations. Therefore, in this study, we evaluated how exposure to mixtures containing bisphenol A and seven phthalates impacts liver functions and metabolic homeostasis. Male mice were gavaged with either EDCs at four different dose combinations or corn oil (control) for six weeks. The results showed that exposure to EDCs at the human daily exposure limit had a negligible impact on liver function. However, EDC at ≥ 25 orders of magnitude of human-relevant doses had detrimental impacts on overall liver function, leading to metabolic abnormalities, steatohepatitis, and hepatic fibrosis via the activation of both genomic and non-genomic pathways. The metabolic phenotype was linked to alterations in key genes involved in hepatic lipid and glucose metabolism. In contrast, alterations in cytokine expression, oxidative stress, and apoptosis impacted steatohepatitis and fibrosis. Because EDC exposure does not occur independently, the findings of the combined effects of exposure to multiple EDCs have significant relevance for public health.
Collapse
Affiliation(s)
- Md Saidur Rahman
- Department of Animal Science & Technology and BET Research Institute, Chung-Ang University, Anseong, Gyeonggi-do 17546, Republic of Korea
| | - Won-Ki Pang
- Department of Animal Science & Technology and BET Research Institute, Chung-Ang University, Anseong, Gyeonggi-do 17546, Republic of Korea
| | - Shehreen Amjad
- Department of Animal Science & Technology and BET Research Institute, Chung-Ang University, Anseong, Gyeonggi-do 17546, Republic of Korea
| | - Do-Yeal Ryu
- Department of Animal Science & Technology and BET Research Institute, Chung-Ang University, Anseong, Gyeonggi-do 17546, Republic of Korea
| | - Elikanah Olusayo Adegoke
- Department of Animal Science & Technology and BET Research Institute, Chung-Ang University, Anseong, Gyeonggi-do 17546, Republic of Korea
| | - Yoo-Jin Park
- Department of Animal Science & Technology and BET Research Institute, Chung-Ang University, Anseong, Gyeonggi-do 17546, Republic of Korea
| | - Myung-Geol Pang
- Department of Animal Science & Technology and BET Research Institute, Chung-Ang University, Anseong, Gyeonggi-do 17546, Republic of Korea.
| |
Collapse
|
29
|
Yan Y, Bao G, Pei J, Cao Y, Zhang C, Zhao P, Zhang Y, Damirin A. NF-κB and EGFR participate in S1PR3-mediated human renal cell carcinomas progression. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166401. [PMID: 35346818 DOI: 10.1016/j.bbadis.2022.166401] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 03/13/2022] [Accepted: 03/20/2022] [Indexed: 11/24/2022]
Abstract
The bioactive lipid sphingosine 1-phosphate (S1P) is implicated in many pivotal processes for the physiological and pathological actions via activating five types of G-protein-coupled S1P receptors (S1PR1-5). The role of S1P in renal cell carcinoma (RCC) and its receptor subtype specific mediating mechanism are poorly studied. So we focus on the regulatory role of S1P in RCC progression and the receptor subtypes involved in S1P-induced actions, intending to further clarify a novel therapeutic target for RCC. Analysis of The Cancer Genome Atlas (TCGA) databases showed that the patients with high expression of S1PR3 had significantly worse overall than with low expression. We further demonstrated that S1P could promote proliferation, migration, and epithelial-mesenchymal transition (EMT) of renal cancer cells in vitro, and the actions were enhanced with the increase of S1PR3 expression. Meanwhile, the results in animal experiments also showed that S1PR3 could accelerate tumorigenesis and metastasis of RCC. Our study also clarified the mechanism for S1P induced cell proliferation is mediated by S1PR3/Gi/p38/Akt/p65/cyclin D1-CDK4 pathway and the main pathway for migration is S1PR3/Gi/q/ERK/p38/p65. In addition, S1PR3 was involved in epidermal growth factor (EGF)-induced actions by enhancing protein expression, not by transactivation of epidermal growth factor receptor (EGFR). These results also further supported our conclusion that the carcinogenic role of S1P/S1PR3 axis. Thus, our findings provide that S1PR3 may be a promising small molecular therapeutic target for S1PR3 expressed cancers.
Collapse
Affiliation(s)
- Yali Yan
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Gegentuya Bao
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Jingyuan Pei
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Ying Cao
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Chenyu Zhang
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Pengfei Zhao
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Yantao Zhang
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Alatangaole Damirin
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, China.
| |
Collapse
|
30
|
Estrogenic Action in Stress-Induced Neuroendocrine Regulation of Energy Homeostasis. Cells 2022; 11:cells11050879. [PMID: 35269500 PMCID: PMC8909319 DOI: 10.3390/cells11050879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/16/2022] [Accepted: 02/28/2022] [Indexed: 01/27/2023] Open
Abstract
Estrogens are among important contributing factors to many sex differences in neuroendocrine regulation of energy homeostasis induced by stress. Research in this field is warranted since chronic stress-related psychiatric and metabolic disturbances continue to be top health concerns, and sex differences are witnessed in these aspects. For example, chronic stress disrupts energy homeostasis, leading to negative consequences in the regulation of emotion and metabolism. Females are known to be more vulnerable to the psychological consequences of stress, such as depression and anxiety, whereas males are more vulnerable to the metabolic consequences of stress. Sex differences that exist in the susceptibility to various stress-induced disorders have led researchers to hypothesize that gonadal hormones are regulatory factors that should be considered in stress studies. Further, estrogens are heavily recognized for their protective effects on metabolic dysregulation, such as anti-obesogenic and glucose-sensing effects. Perturbations to energy homeostasis using laboratory rodents, such as physiological stress or over-/under- feeding dietary regimen prevalent in today’s society, offer hints to the underlying mechanisms of estrogenic actions. Metabolic effects of estrogens primarily work through estrogen receptor α (ERα), which is differentially expressed between the sexes in hypothalamic nuclei regulating energy metabolism and in extrahypothalamic limbic regions that are not typically associated with energy homeostasis. In this review, we discuss estrogenic actions implicated in stress-induced sex-distinct metabolic disorders.
Collapse
|
31
|
Estrogens—Origin of Centrosome Defects in Human Cancer? Cells 2022; 11:cells11030432. [PMID: 35159242 PMCID: PMC8833882 DOI: 10.3390/cells11030432] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/24/2022] [Accepted: 01/25/2022] [Indexed: 12/22/2022] Open
Abstract
Estrogens are associated with a variety of diseases and play important roles in tumor development and progression. Centrosome defects are hallmarks of human cancers and contribute to ongoing chromosome missegragation and aneuploidy that manifest in genomic instability and tumor progression. Although several mechanisms underlie the etiology of centrosome aberrations in human cancer, upstream regulators are hardly known. Accumulating experimental and clinical evidence points to an important role of estrogens in deregulating centrosome homeostasis and promoting karyotype instability. Here, we will summarize existing literature of how natural and synthetic estrogens might contribute to structural and numerical centrosome defects, genomic instability and human carcinogenesis.
Collapse
|
32
|
Corrê MDS, de Freitas BS, Machado GDB, Pires VN, Bromberg E, Hallak JEC, Zuardi AW, Crippa JAS, Schröder N. Cannabidiol reverses memory impairments and activates components of the Akt/GSK3β pathway in an experimental model of estrogen depletion. Behav Brain Res 2022; 417:113555. [PMID: 34450240 DOI: 10.1016/j.bbr.2021.113555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/16/2021] [Accepted: 08/23/2021] [Indexed: 11/28/2022]
Abstract
Clinical and preclinical evidence has indicated that estrogen depletion leads to memory impairments and increases the susceptibility to neural damage. Here, we have sought to investigate the effects of Cannabidiol (CBD) a non-psychotomimetic compound from Cannabis sativa, on memory deficits induced by estrogen depletion in rats, and its underlying mechanisms. Adult rats were subjected to bilateral ovariectomy, an established estrogen depletion model in rodents, or sham surgery and allowed to recover for three weeks. After that, they received daily injections of CBD (10 mg/kg) for fourteen days. Rats were tested in the inhibitory avoidance task, a type of emotionally-motivated memory. After behavioral testing they were euthanized, and their hippocampi were isolated for analysis of components of the Akt/GSK3β survival pathway and the antiapoptotic protein Bcl2. Results revealed that ovariectomy impaired avoidance memory, and CBD was able to completely reverse estrogen depletion-induced memory impairment. Ovariectomy also reduced Akt/GSK3β pathway's activation by decreasing the phosphorylation levels of Akt and GSK3β and Bcl2 levels, which were ameliorated by CBD. The present results indicate that CBD leads to a functional recovery accompanied by the Akt/GSK3β survival pathway's activation, supporting its potential as a treatment for estrogen decline-induced deterioration of neural functioning and maintenance.
Collapse
Affiliation(s)
- Márcio da Silveira Corrê
- Neurobiology and Developmental Biology Laboratory, School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil; Faculty of Medicine, Department of Health, Integrated Regional University of Upper Uruguay and Missions, Erechim, Brazil
| | - Betânia Souza de Freitas
- Neurobiology and Developmental Biology Laboratory, School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Gustavo Dalto Barroso Machado
- Neurobiology and Developmental Biology Laboratory, School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Vivian Naziaseno Pires
- Neurobiology and Developmental Biology Laboratory, School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Elke Bromberg
- Neurobiology and Developmental Biology Laboratory, School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil; National Institute of Science and Technology for Translational Medicine (INCT-TM), Conselho Nacional de Desenvolvimento Cientifico e Tecnologico (CNPq), Brasília, Brazil
| | - Jaime E C Hallak
- National Institute of Science and Technology for Translational Medicine (INCT-TM), Conselho Nacional de Desenvolvimento Cientifico e Tecnologico (CNPq), Brasília, Brazil; Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Antônio Waldo Zuardi
- National Institute of Science and Technology for Translational Medicine (INCT-TM), Conselho Nacional de Desenvolvimento Cientifico e Tecnologico (CNPq), Brasília, Brazil; Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - José Alexandre S Crippa
- National Institute of Science and Technology for Translational Medicine (INCT-TM), Conselho Nacional de Desenvolvimento Cientifico e Tecnologico (CNPq), Brasília, Brazil; Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Nadja Schröder
- National Institute of Science and Technology for Translational Medicine (INCT-TM), Conselho Nacional de Desenvolvimento Cientifico e Tecnologico (CNPq), Brasília, Brazil; Department of Physiology, Institute for Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil.
| |
Collapse
|
33
|
The impact of G protein-coupled oestrogen receptor 1 on male breast cancer: a retrospective analysis. Contemp Oncol (Pozn) 2021; 25:204-212. [PMID: 34729041 PMCID: PMC8547179 DOI: 10.5114/wo.2021.110010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 09/23/2021] [Indexed: 01/02/2023] Open
Abstract
Introduction The G protein-coupled oestrogen receptor 1 (GPER-1) is a potential prognostic marker in breast cancer. However, its role in male breast cancer (MBC) is still unknown. This study evaluates the expression of GPER-1 in MBC samples and correlates these data with clinical and pathological parameters including patients' survival. Material and methods For this retrospective analysis of a prospectively maintained cohort of patients with MBC, we examined 161 specimens for GPER-1 expression using immunohistochemistry. An immunoreactive score (IRS) was calculated based on staining intensity and the percentage of positive tumour cells. Then, we correlated GPER-1 IRS with clinical and pathological parameters, and overall and relapse-free survival. Results About 40% of MBC samples were positive for GPER-1 expression (IRS ≥ 4). There was no significant correlation with clinicopathological parameters, such as hormone receptor status or grading. However, a statistical trend was observed for tumour size (≥ 2 cm, p = 0.093). Kaplan-Meier survival analysis revealed no significant correlation with relapse-free survival. However, there was a significant correlation with overall survival, but when we adjusted the log-rank p-value to compensate for the cut-off point optimization method, it rose above 0.1. Additionally, GPER-1-positive patients were older at diagnosis. When adjusted for age by multivariable Cox regression analysis, the significance of GPER-1 status for survival was further reduced. Conclusions We found no significant prognostic value of GPER-1 in this MBC cohort as anticipated from studies on female BC. Future studies with higher sample size are needed to further verify a potential sex-specific role of GPER-1.
Collapse
|
34
|
Molecular Characterization of Membrane Steroid Receptors in Hormone-Sensitive Cancers. Cells 2021; 10:cells10112999. [PMID: 34831222 PMCID: PMC8616056 DOI: 10.3390/cells10112999] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 10/29/2021] [Accepted: 11/02/2021] [Indexed: 12/20/2022] Open
Abstract
Cancer is one of the most common causes of death worldwide, and its development is a result of the complex interaction of genetic factors, environmental cues, and aging. Hormone-sensitive cancers depend on the action of one or more hormones for their development and progression. Sex steroids and corticosteroids can regulate different physiological functions, including metabolism, growth, and proliferation, through their interaction with specific nuclear receptors, that can transcriptionally regulate target genes via their genomic actions. Therefore, interference with hormones’ activities, e.g., deregulation of their production and downstream pathways or the exposition to exogenous hormone-active substances such as endocrine-disrupting chemicals (EDCs), can affect the regulation of their correlated pathways and trigger the neoplastic transformation. Although nuclear receptors account for most hormone-related biologic effects and their slow genomic responses are well-studied, less-known membrane receptors are emerging for their ability to mediate steroid hormones effects through the activation of rapid non-genomic responses also involved in the development of hormone-sensitive cancers. This review aims to collect pre-clinical and clinical data on these extranuclear receptors not only to draw attention to their emerging role in cancer development and progression but also to highlight their dual role as tumor microenvironment players and potential candidate drug targets.
Collapse
|
35
|
Maric D, Paterek A, Delaunay M, López IP, Arambasic M, Diviani D. A-Kinase Anchoring Protein 2 Promotes Protection against Myocardial Infarction. Cells 2021; 10:2861. [PMID: 34831084 PMCID: PMC8616452 DOI: 10.3390/cells10112861] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 10/20/2021] [Accepted: 10/21/2021] [Indexed: 12/25/2022] Open
Abstract
Myocardial infarction (MI) is a leading cause of maladaptive cardiac remodeling and heart failure. In the damaged heart, loss of function is mainly due to cardiomyocyte death and remodeling of the cardiac tissue. The current study shows that A-kinase anchoring protein 2 (AKAP2) orchestrates cellular processes favoring cardioprotection in infarcted hearts. Induction of AKAP2 knockout (KO) in cardiomyocytes of adult mice increases infarct size and exacerbates cardiac dysfunction after MI, as visualized by increased left ventricular dilation and reduced fractional shortening and ejection fraction. In cardiomyocytes, AKAP2 forms a signaling complex with PKA and the steroid receptor co-activator 3 (Src3). Upon activation of cAMP signaling, the AKAP2/PKA/Src3 complex favors PKA-mediated phosphorylation and activation of estrogen receptor α (ERα). This results in the upregulation of ER-dependent genes involved in protection against apoptosis and angiogenesis, including Bcl2 and the vascular endothelial growth factor a (VEGFa). In line with these findings, cardiomyocyte-specific AKAP2 KO reduces Bcl2 and VEGFa expression, increases myocardial apoptosis and impairs the formation of new blood vessels in infarcted hearts. Collectively, our findings suggest that AKAP2 organizes a transcriptional complex that mediates pro-angiogenic and anti-apoptotic responses that protect infarcted hearts.
Collapse
Affiliation(s)
- Darko Maric
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland; (D.M.); (A.P.); (M.D.); (I.P.L.); (M.A.)
- Section of Medicine, Department of Endocrinology, Metabolism and Cardiovascular System, University of Fribourg, 1700 Fribourg, Switzerland
| | - Aleksandra Paterek
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland; (D.M.); (A.P.); (M.D.); (I.P.L.); (M.A.)
| | - Marion Delaunay
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland; (D.M.); (A.P.); (M.D.); (I.P.L.); (M.A.)
| | - Irene Pérez López
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland; (D.M.); (A.P.); (M.D.); (I.P.L.); (M.A.)
| | - Miroslav Arambasic
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland; (D.M.); (A.P.); (M.D.); (I.P.L.); (M.A.)
| | - Dario Diviani
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland; (D.M.); (A.P.); (M.D.); (I.P.L.); (M.A.)
| |
Collapse
|
36
|
Gjorgoska M, Rižner TL. Estrogens and the Schrödinger's Cat in the Ovarian Tumor Microenvironment. Cancers (Basel) 2021; 13:cancers13195011. [PMID: 34638494 PMCID: PMC8508344 DOI: 10.3390/cancers13195011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/29/2021] [Accepted: 10/02/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Ovarian cancer is a complex pathology for which we require effective screening and therapeutical strategies. Apart from the cancer cell portion, there exist plastic immune and non-immune cell populations, jointly constituting the context-adaptive tumor microenvironment, which is pivotal in tumorigenesis. Estrogens might be synthesized in the ovarian tumor tissue and actively contribute to the shaping of an immunosuppressive microenvironment. Current immune therapies have limited effectiveness as a multitude of factors influence the outcome. A thorough understanding of the ovarian cancer biology is crucial in the efforts to reestablish homeostasis. Abstract Ovarian cancer is a heterogeneous disease affecting the aging ovary, in concert with a complex network of cells and signals, together representing the ovarian tumor microenvironment. As in the “Schrödinger’s cat” thought experiment, the context-dependent constituents of the—by the time of diagnosis—well-established tumor microenvironment may display a tumor-protective and -destructive role. Systemic and locally synthesized estrogens contribute to the formation of a pro-tumoral microenvironment that enables the sustained tumor growth, invasion and metastasis. Here we focus on the estrogen biosynthetic and metabolic pathways in ovarian cancer and elaborate their actions on phenotypically plastic, estrogen-responsive, aging immune cells of the tumor microenvironment, altogether highlighting the multicomponent-connectedness and complexity of cancer, and contributing to a broader understanding of the ovarian cancer biology.
Collapse
|
37
|
GPR30 Activation by 17β-Estradiol Promotes p62 Phosphorylation and Increases Estrogen Receptor α Protein Expression by Inducing Its Release from a Complex Formed with KEAP1. J Pers Med 2021; 11:jpm11090906. [PMID: 34575683 PMCID: PMC8468056 DOI: 10.3390/jpm11090906] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 08/21/2021] [Accepted: 09/09/2021] [Indexed: 12/13/2022] Open
Abstract
Estrogens can elicit rapid cellular responses via the G-protein-coupled receptor 30 (GPR30), followed by estrogen receptor α (ERα/ESR1)-mediated genomic effects. Here, we investigated whether rapid estrogen signaling via GRP30 may affect ESR1 expression, and we examined the underlying molecular mechanisms. The exposure of human endometrial cancer cells to 17β-estradiol promoted p62 phosphorylation and increased ESR1 protein expression. However, both a GPR30 antagonist and GPR30 silencing abrogated this phenomenon. GPR30 activation by 17β-estradiol elicited the SRC/EGFR/PI3K/Akt/mTOR signaling pathway. Intriguingly, unphosphorylated p62 and ESR1 were found to form an intracellular complex with the substrate adaptor protein KEAP1. Upon phosphorylation, p62 promoted ESR1 release from the complex, to increase its protein expression. Given the critical role played by p62 in autophagy, we also examined how this process affected ESR1 expression. The activation of autophagy by everolimus decreased ESR1 by promoting p62 degradation, whereas autophagy inhibition with chloroquine increased ESR1 expression. The treatment of female C57BL/6 mice with the autophagy inhibitor hydroxychloroquine—which promotes p62 expression—increased both phosphorylated p62 and ESR1 expression in uterine epithelial cells. Collectively, our results indicate that 17β-estradiol-mediated GPR30 activation elicits the SRC/EGFR/PI3K/Akt/mTOR signaling pathway and promotes p62 phosphorylation. In turn, phosphorylated p62 increased ESR1 expression by inducing its release from complexes that included KEAP1. Our findings may lead to novel pharmacological strategies aimed at decreasing ESR1 expression in estrogen-sensitive cells.
Collapse
|
38
|
Xu J, Zhou Y, Yan C, Wang X, Lou J, Luo Y, Gao S, Wang J, Wu L, Gao X, Shao A. Neurosteroids: A novel promise for the treatment of stroke and post-stroke complications. J Neurochem 2021; 160:113-127. [PMID: 34482541 DOI: 10.1111/jnc.15503] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 08/22/2021] [Accepted: 08/23/2021] [Indexed: 01/14/2023]
Abstract
Stroke is the primary reason for death and disability worldwide, with few treatment strategies to date. Neurosteroids, which are natural molecules in the brain, have aroused great interest in the field of stroke. Neurosteroids are a kind of steroid that acts on the nervous system, and are synthesized in the mitochondria of neurons or glial cells using cholesterol or other steroidal precursors. Neurosteroids mainly include estrogen, progesterone (PROG), allopregnanolone, dehydroepiandrosterone (DHEA), and vitamin D (VD). Most of the preclinical studies have confirmed that neurosteroids can decrease the risk of stroke, and improve stroke outcomes. In the meantime, neurosteroids have been shown to have a positive therapeutic significance in some post-stroke complications, such as epilepsy, depression, anxiety, cardiac complications, movement disorders, and post-stroke pain. In this review, we report the historical background, modulatory mechanisms of neurosteroids in stroke and post-stroke complications, and emphasize on the application prospect of neurosteroids in stroke therapy.
Collapse
Affiliation(s)
- Jiawei Xu
- The First Affiliated Hospital of Zhejiang, Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yunxiang Zhou
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Caochong Yan
- The Key Laboratory of Reproductive Genetics, Ministry of Education, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaoyu Wang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jianyao Lou
- Department of General Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yi Luo
- The Second Affiliated Hospital of Zhejiang University School of Medicine (Changxing Branch), Changxing, Huzhou, Zhejiang, China
| | - Shiqi Gao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Junjie Wang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Liang Wu
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiangfu Gao
- The First Affiliated Hospital of Zhejiang, Chinese Medical University, Hangzhou, Zhejiang, China
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
39
|
Lopes ACR, Zavan B, Corrêa YJC, Vieira TM, Severs LJ, Oliveira LM, Soncini R. Impact of obesity and ovariectomy on respiratory function in female mice. Respir Physiol Neurobiol 2021; 294:103775. [PMID: 34416380 DOI: 10.1016/j.resp.2021.103775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 08/03/2021] [Accepted: 08/09/2021] [Indexed: 10/20/2022]
Abstract
Obesity and the corresponding variations in female sex hormones are associated with severe lung disease. We determined the potential effects of obesity and sex hormones in female mice by investigating changes in lung structure and respiratory function in an obesity model induced by postnatal overnutrition. Obese female mice exhibited pronounced weight gain, abdominal fat accumulation and collagen type I deposition in the airways. However, neither elastic tissue nor estrogen receptors-α/-β were affected in obese female mice after ovariectomy or sham-operated mice. Bronchoconstriction in response to methacholine challenge in obese sham-operated mice was higher than in the obese group after ovariectomy. Our results suggest that the coexistence of obesity and ovariectomy impacted on respiratory system and airway resistance (attenuates bronchoconstriction after methacholine), on collagen I deposition and on airway estrogen β-receptors of mice.
Collapse
Affiliation(s)
- Ana C R Lopes
- Department of Physiology, Institute of Biomedical Science, Federal University of Alfenas, 37130-000, Alfenas, MG, Brazil
| | - Bruno Zavan
- Integrative Animal Biology Laboratory, Institute of Biomedical Science, Federal University of Alfenas, 37130-000, Alfenas, MG, Brazil
| | - Yuri J C Corrêa
- Department of Physiology, Institute of Biomedical Science, Federal University of Alfenas, 37130-000, Alfenas, MG, Brazil
| | - Tânia M Vieira
- Department of Physiology, Institute of Biomedical Science, Federal University of Alfenas, 37130-000, Alfenas, MG, Brazil
| | - Liza J Severs
- Center for Integrative Brain Research, Seattle Children's Research Institute, 1900 9th Avenue, JMB10, Seattle, WA, 98101, USA
| | - Luiz M Oliveira
- Center for Integrative Brain Research, Seattle Children's Research Institute, 1900 9th Avenue, JMB10, Seattle, WA, 98101, USA
| | - Roseli Soncini
- Department of Physiology, Institute of Biomedical Science, Federal University of Alfenas, 37130-000, Alfenas, MG, Brazil.
| |
Collapse
|
40
|
Delgado NTB, Rouver WDN, Freitas-Lima LC, Vieira-Alves I, Lemos VS, dos Santos RL. Sex Differences in the Vasodilation Mediated by G Protein-Coupled Estrogen Receptor (GPER) in Hypertensive Rats. Front Physiol 2021; 12:659291. [PMID: 34393807 PMCID: PMC8359777 DOI: 10.3389/fphys.2021.659291] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 06/22/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The protective effect of estrogen on the vasculature cannot be explained only by its action through the receptors ERα and ERβ. G protein-coupled estrogen receptors (GPER)-which are widely distributed throughout the cardiovascular system-may also be involved in this response. However, little is known about GPER actions in hypertension. Therefore, in this study we evaluated the vascular response mediated by GPER using a specific agonist, G-1, in spontaneously hypertensive rats (SHR). We hypothesized that G-1 would induce a relaxing response in resistance mesenteric arteries from SHR of both sexes. METHODS G-1 concentration-response curves (1 nM-10 μM) were performed in mesenteric arteries from SHR of both sexes (10-12-weeks-old, weighing 180-250 g). The effects of G-1 were evaluated before and after endothelial removal and incubation for 30 min with the inhibitors L-NAME (300 μM) and indomethacin (10 μM) alone or combined with clotrimazole (0.75 μM) or catalase (1,000 units/mL). GPER immunolocalization was also investigated, and vascular hydrogen peroxide (H2O2) and ROS were evaluated using dichlorofluorescein (DCF) and dihydroethidium (DHE) staining, respectively. RESULTS GPER activation promoted a similar relaxing response in resistance mesenteric arteries of female and male hypertensive rats, but with the participation of different endothelial mediators. Males appear to be more dependent on the NO pathway, followed by the H2O2 pathway, and females on the endothelium and H2O2 pathway. CONCLUSION These findings show that the GPER agonist G-1 can induce a relaxing response in mesenteric arteries from hypertensive rats of both sexes in a similar way, albeit with differential participation of endothelial mediators. These results contribute to the understanding of GPER activation on resistance mesenteric arteries in essential hypertension.
Collapse
Affiliation(s)
| | - Wender do Nascimento Rouver
- Department of Physiological Sciences, Health Sciences Center, Federal University of Espirito Santo, Vitoria, Brazil
| | | | - Ildernandes Vieira-Alves
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Virgínia Soares Lemos
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Roger Lyrio dos Santos
- Department of Physiological Sciences, Health Sciences Center, Federal University of Espirito Santo, Vitoria, Brazil
| |
Collapse
|
41
|
Lee D, Kim YM, Chin YW, Kang KS. Schisandrol A Exhibits Estrogenic Activity via Estrogen Receptor α-Dependent Signaling Pathway in Estrogen Receptor-Positive Breast Cancer Cells. Pharmaceutics 2021; 13:pharmaceutics13071082. [PMID: 34371773 PMCID: PMC8308983 DOI: 10.3390/pharmaceutics13071082] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/12/2021] [Accepted: 07/12/2021] [Indexed: 01/29/2023] Open
Abstract
The aim of this study was to examine the estrogen-like effects of gentiopicroside, macelignan, γ-mangostin, and three lignans (schisandrol A, schisandrol B, and schisandrin C), and their possible mechanism of action. Their effects on the proliferation of the estrogen receptor (ER)-positive breast cancer cell line (MCF-7) were evaluated using Ez-Cytox reagents. The expression of extracellular signal-regulated kinase (ERK), phosphatidylinositol 3-kinase (PI3K), AKT, and estrogen receptor α (ERα) was measured by performing Western blot analysis. 17β-estradiol (E2), also known as estradiol, is an estrogen steroid and was used as a positive control. ICI 182,780 (ICI), an ER antagonist, was used to block the ER function. Our results showed that, except for gentiopicroside, all the compounds promoted proliferation of MCF-7 cells, with schisandrol A being the most effective; this effect was better than that of E2 and was mitigated by ICI. Consistently, the expression of ERK, PI3K, AKT, and ERα increased following treatment with schisandrol A; this effect was slightly better than that of E2 and was mitigated by ICI. Taken together, the ERα induction via the PI3K/AKT and ERK signaling pathways may be a potential mechanism underlying the estrogen-like effects of schisandrol A. This study provides an experimental basis for the application of schisandrol A as a phytoestrogen for the prevention of menopausal symptoms.
Collapse
Affiliation(s)
- Dahae Lee
- College of Korean Medicine, Gachon University, Seongnam 13120, Korea;
| | - Young-Mi Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1, Gwanak-ro, Gwanak-gu, Seoul 08826, Korea;
| | - Young-Won Chin
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1, Gwanak-ro, Gwanak-gu, Seoul 08826, Korea;
- Correspondence: (Y.-W.C.); (K.S.K.); Tel.: +82-2-880-7859 (Y.-W.C.); +82-31-750-5402 (K.S.K.)
| | - Ki Sung Kang
- College of Korean Medicine, Gachon University, Seongnam 13120, Korea;
- Correspondence: (Y.-W.C.); (K.S.K.); Tel.: +82-2-880-7859 (Y.-W.C.); +82-31-750-5402 (K.S.K.)
| |
Collapse
|
42
|
Sahebnasagh A, Saghafi F, Negintaji S, Hu T, Shabani-Boroujeni M, Safdari M, Ghaleno HR, Miao L, Qi Y, Wang M, Liao P, Sureda A, Simal-Gándara J, Nabavi SM, Xiao J. Nitric Oxide and Immune Responses in Cancer: Searching for New Therapeutic Strategies. Curr Med Chem 2021; 29:1561-1595. [PMID: 34238142 DOI: 10.2174/0929867328666210707194543] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 05/05/2021] [Accepted: 05/15/2021] [Indexed: 02/08/2023]
Abstract
In recent years, there has been an increasing interest in understanding the mysterious functions of nitric oxide (NO) and how this pleiotropic signaling molecule contributes to tumorigenesis. This review attempts to expose and discuss the information available on the immunomodulatory role of NO in cancer and recent approaches to the role of NO donors in the area of immunotherapy. To address the goal, the following databases were searched to identify relevant literature concerning empirical evidence: The Cochrane Library, Pubmed, Medline, EMBASE from 1980 through March 2020. Valuable attempts have been made to develop distinctive NO-based cancer therapy. Although the data do not allow generalization, the evidence seems to indicate that low / moderate levels may favor tumorigenesis while higher levels would exert anti-tumor effects. In this sense, the use of NO donors could have an important therapeutic potential within immunotherapy, although there are still no clinical trials. The emerging understanding of NO-regulated immune responses in cancer may help unravel the recent features of this "double-edged sword" in cancer physiological and pathologic processes and its potential use as a therapeutic agent for cancer treatment. In short, in this review, we discuss the complex cellular mechanism in which NO, as a pleiotropic signaling molecule, participates in cancer pathophysiology. We also debate the dual role of NO in cancer and tumor progression, and clinical approaches for inducible nitric oxide synthase (iNOS) based therapy against cancer.
Collapse
Affiliation(s)
- Adeleh Sahebnasagh
- Clinical Research Center, Department of Internal Medicine, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Fatemeh Saghafi
- Department of Clinical Pharmacy, Faculty of Pharmacy and Pharmaceutical Sciences Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Sina Negintaji
- Student Research Committee, School of Pharmacy, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Tingyan Hu
- School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Mojtaba Shabani-Boroujeni
- Department of Clinical Pharmacy, Faculty of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammadreza Safdari
- Department of Orthopedic Surgery, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Hassan Rezai Ghaleno
- Department of Surgery, Faculty of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Lingchao Miao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Yaping Qi
- Purdue Quantum Science and Engineering Institute, Purdue University, West Lafayette, IN 47907, United States
| | - Mingfu Wang
- School of Biological Sciences, The University of Hong Kong, Pokfulam Road. Hong Kong, China
| | - Pan Liao
- Department of Biochemistry, Purdue University, West Lafayette, IN 47907, United States
| | - Antoni Sureda
- Research Group on Community Nutrition and Oxidative Stress, University of the Balearic Islands, Palma de Mallorca, Spain
| | - Jesus Simal-Gándara
- Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Faculty of Food Science and Technology, University of Vigo - Ourense Campus, E-32004 Ourense, Spain
| | - Seyed Mohammad Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Jianbo Xiao
- Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Faculty of Food Science and Technology, University of Vigo - Ourense Campus, E-32004 Ourense, Spain
| |
Collapse
|
43
|
Liu L, Liu S, Luo H, Chen C, Zhang X, He L, Tu G. GPR30-mediated HMGB1 upregulation in CAFs induces autophagy and tamoxifen resistance in ERα-positive breast cancer cells. Aging (Albany NY) 2021; 13:16178-16197. [PMID: 34182538 PMCID: PMC8266353 DOI: 10.18632/aging.203145] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 05/17/2021] [Indexed: 01/30/2023]
Abstract
Tamoxifen (TAM) resistance constitutes a challenge in managing estrogen receptor (ER)α+ breast cancer patients. G-protein-coupled estrogen receptor (GPR30/GPER), which reportedly initiates TAM resistance in ERα+/ GPR30+ breast cancers, is detected in the breast cancer microenvironment, especially cancer associated fibroblasts (CAFs). Herein, considering that GPR30 mediates transcriptional regulation in different cell backgrounds, a microarray strategy was applied in immortalized CAFs derived from primary breast cancer samples, resulting in the identification of 165 GPR30 target genes, among which HMGB1 was confirmed to be upregulated by 17-β estradiol(E2)- and TAM-triggered GPR30 activation in CAFs. Activated GPR30 increased extracellular HMGB1 secretion by CAFs, which was reduced by blocking PI3K/AKT signaling using G15 or LY294002. GPR30-induced HMGB1 upregulation triggered MEK/ERK signaling, leading to increased autophagic behavior to protect cancer cells from TAM-induced apoptosis, mimicking the recombinant HMGB1-mediated increase in cancer cell resistance potential to TAM. MEK/ERK signaling blockage by U0126 decreased the autophagic behavior and resistance ability of cancer cells to TAM. CAF-expressed GPR30 induced TAM resistance via HMGB1 in vivo. Overall, TAM upregulated HMGB1 expression and secretion in CAFs via GPR30/PI3K/AKT signaling, and the secreted HMGB1 induced autophagy to enhance TAM resistance in MCF-7 cells in an ERK-dependent manner. Thus, targeting GPR30 and downstream cascades may be an effective strategy to attenuate the resistance of ERα-positive breast tumors to endocrine therapy.
Collapse
Affiliation(s)
- Li Liu
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Yu-Zhong 400016, Chongqing, China
| | - Shengchun Liu
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Yu-Zhong 400016, Chongqing, China
| | - Haojun Luo
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Chenxi Chen
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Yu-Zhong 400016, Chongqing, China
| | - Xiaoling Zhang
- Maternal and Child Care Center Service of Kaizhou, Chongqing 405400, China
| | - Lin He
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Yu-Zhong 400016, Chongqing, China
| | - Gang Tu
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Yu-Zhong 400016, Chongqing, China
| |
Collapse
|
44
|
Reyes-García J, Montaño LM, Carbajal-García A, Wang YX. Sex Hormones and Lung Inflammation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1304:259-321. [PMID: 34019274 DOI: 10.1007/978-3-030-68748-9_15] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Inflammation is a characteristic marker in numerous lung disorders. Several immune cells, such as macrophages, dendritic cells, eosinophils, as well as T and B lymphocytes, synthetize and release cytokines involved in the inflammatory process. Gender differences in the incidence and severity of inflammatory lung ailments including asthma, chronic obstructive pulmonary disease (COPD), pulmonary fibrosis (PF), lung cancer (LC), and infectious related illnesses have been reported. Moreover, the effects of sex hormones on both androgens and estrogens, such as testosterone (TES) and 17β-estradiol (E2), driving characteristic inflammatory patterns in those lung inflammatory diseases have been investigated. In general, androgens seem to display anti-inflammatory actions, whereas estrogens produce pro-inflammatory effects. For instance, androgens regulate negatively inflammation in asthma by targeting type 2 innate lymphoid cells (ILC2s) and T-helper (Th)-2 cells to attenuate interleukin (IL)-17A-mediated responses and leukotriene (LT) biosynthesis pathway. Estrogens may promote neutrophilic inflammation in subjects with asthma and COPD. Moreover, the activation of estrogen receptors might induce tumorigenesis. In this chapter, we summarize the most recent advances in the functional roles and associated signaling pathways of inflammatory cellular responses in asthma, COPD, PF, LC, and newly occurring COVID-19 disease. We also meticulously deliberate the influence of sex steroids on the development and progress of these common and severe lung diseases.
Collapse
Affiliation(s)
- Jorge Reyes-García
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico.,Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Luis M Montaño
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico
| | - Abril Carbajal-García
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico
| | - Yong-Xiao Wang
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA.
| |
Collapse
|
45
|
Linares-Anaya O, Avila-Sorrosa A, Díaz-Cedillo F, Gil-Ruiz LÁ, Correa-Basurto J, Salazar-Mendoza D, Orjuela AL, Alí-Torres J, Ramírez-Apan MT, Morales-Morales D. Synthesis, Characterization, and Preliminary In Vitro Cytotoxic Evaluation of a Series of 2-Substituted Benzo [ d] [1,3] Azoles. Molecules 2021; 26:molecules26092780. [PMID: 34066820 PMCID: PMC8125891 DOI: 10.3390/molecules26092780] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 04/09/2021] [Accepted: 04/13/2021] [Indexed: 11/16/2022] Open
Abstract
A series of benzo [d] [1,3] azoles 2-substituted with benzyl- and allyl-sulfanyl groups were synthesized, and their cytotoxic activities were in vitro evaluated against a panel of six human cancer cell lines. The results showed that compounds BTA-1 and BMZ-2 have the best inhibitory effects, compound BMZ-2 being comparable in some cases with the reference drug tamoxifen and exhibiting a low cytotoxic effect against healthy cells. In silico molecular coupling studies at the tamoxifen binding site of ERα and GPER receptors revealed affinity and the possible mode of interaction of both compounds BTA-1 and BMZ-2.
Collapse
Affiliation(s)
- Ozvaldo Linares-Anaya
- Instituto Politécnico Nacional, Departamento de Química Orgánica, Carpio y Plan de Ayala S/N, Escuela Nacional de Ciencias Biológicas, Colonia Santo Tomás, Ciudad de México 11340, Mexico; (O.L.-A.); (F.D.-C.); (L.Á.G.-R.)
| | - Alcives Avila-Sorrosa
- Instituto Politécnico Nacional, Departamento de Química Orgánica, Carpio y Plan de Ayala S/N, Escuela Nacional de Ciencias Biológicas, Colonia Santo Tomás, Ciudad de México 11340, Mexico; (O.L.-A.); (F.D.-C.); (L.Á.G.-R.)
- Correspondence: ; Tel.: +52-555-729-6000
| | - Francisco Díaz-Cedillo
- Instituto Politécnico Nacional, Departamento de Química Orgánica, Carpio y Plan de Ayala S/N, Escuela Nacional de Ciencias Biológicas, Colonia Santo Tomás, Ciudad de México 11340, Mexico; (O.L.-A.); (F.D.-C.); (L.Á.G.-R.)
| | - Luis Ángel Gil-Ruiz
- Instituto Politécnico Nacional, Departamento de Química Orgánica, Carpio y Plan de Ayala S/N, Escuela Nacional de Ciencias Biológicas, Colonia Santo Tomás, Ciudad de México 11340, Mexico; (O.L.-A.); (F.D.-C.); (L.Á.G.-R.)
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotecnológica, Instituto Politécnico Nacional, Escuela Superior de Medicina, Ciudad de México 11340, Mexico;
| | - José Correa-Basurto
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotecnológica, Instituto Politécnico Nacional, Escuela Superior de Medicina, Ciudad de México 11340, Mexico;
| | - Domingo Salazar-Mendoza
- Carretera a Acatlima, Huajuapan de León, Universidad Tecnológica de la Mixteca, Oaxaca 69000, Mexico;
| | - Adrian L. Orjuela
- Departamento de Química, Universidad Nacional de Colombia-Sede, Bogotá 111321, Colombia; (A.L.O.); (J.A.-T.)
| | - Jorge Alí-Torres
- Departamento de Química, Universidad Nacional de Colombia-Sede, Bogotá 111321, Colombia; (A.L.O.); (J.A.-T.)
| | - María Teresa Ramírez-Apan
- Instituto de Química, Universidad Nacional Autónoma de México, Circuito Exterior, Ciudad Universitaria, Ciudad de México 04510, Mexico; (M.T.R.-A.); (D.M.-M.)
| | - David Morales-Morales
- Instituto de Química, Universidad Nacional Autónoma de México, Circuito Exterior, Ciudad Universitaria, Ciudad de México 04510, Mexico; (M.T.R.-A.); (D.M.-M.)
| |
Collapse
|
46
|
Zahra A, Dong Q, Hall M, Jeyaneethi J, Silva E, Karteris E, Sisu C. Identification of Potential Bisphenol A (BPA) Exposure Biomarkers in Ovarian Cancer. J Clin Med 2021; 10:jcm10091979. [PMID: 34062972 PMCID: PMC8125610 DOI: 10.3390/jcm10091979] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/16/2021] [Accepted: 04/24/2021] [Indexed: 02/07/2023] Open
Abstract
Endocrine-disrupting chemicals (EDCs) can exert multiple deleterious effects and have been implicated in carcinogenesis. The xenoestrogen Bisphenol A (BPA) that is found in various consumer products has been involved in the dysregulation of numerous signalling pathways. In this paper, we present the analysis of a set of 94 genes that have been shown to be dysregulated in presence of BPA in ovarian cancer cell lines since we hypothesised that these genes might be of biomarker potential. This study sought to identify biomarkers of disease and biomarkers of disease-associated exposure. In silico analyses took place using gene expression data extracted from The Cancer Genome Atlas (TCGA) and the Genotype-Tissue Expression (GTEx) databases. Differential expression was further validated at protein level using immunohistochemistry on an ovarian cancer tissue microarray. We found that 14 out of 94 genes are solely dysregulated in the presence of BPA, while the remaining 80 genes are already dysregulated (p-value < 0.05) in their expression pattern as a consequence of the disease. We also found that seven genes have prognostic power for the overall survival in OC in relation to their expression levels. Out of these seven genes, Keratin 4 (KRT4) appears to be a biomarker of exposure-associated ovarian cancer, whereas Guanylate Binding Protein 5 (GBP5), long intergenic non-protein coding RNA 707 (LINC00707) and Solute Carrier Family 4 Member 11 (SLC4A11) are biomarkers of disease. BPA can exert a plethora of effects that can be tissue- or cancer-specific. Our in silico findings generate a hypothesis around biomarkers of disease and exposure that could potentially inform regulation and policy making.
Collapse
Affiliation(s)
- Aeman Zahra
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK; (A.Z.); (Q.D.); (M.H.); (J.J.); (E.S.)
| | - Qiduo Dong
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK; (A.Z.); (Q.D.); (M.H.); (J.J.); (E.S.)
| | - Marcia Hall
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK; (A.Z.); (Q.D.); (M.H.); (J.J.); (E.S.)
- Mount Vernon Cancer Centre, Northwood HA6 2RN, UK
| | - Jeyarooban Jeyaneethi
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK; (A.Z.); (Q.D.); (M.H.); (J.J.); (E.S.)
| | - Elisabete Silva
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK; (A.Z.); (Q.D.); (M.H.); (J.J.); (E.S.)
| | - Emmanouil Karteris
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK; (A.Z.); (Q.D.); (M.H.); (J.J.); (E.S.)
- Correspondence: (E.K.); (C.S.)
| | - Cristina Sisu
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK; (A.Z.); (Q.D.); (M.H.); (J.J.); (E.S.)
- Correspondence: (E.K.); (C.S.)
| |
Collapse
|
47
|
Qiu YA, Xiong J, Yu T. Role of G Protein-Coupled Estrogen Receptor in Digestive System Carcinomas: A Minireview. Onco Targets Ther 2021; 14:2611-2622. [PMID: 33888991 PMCID: PMC8055353 DOI: 10.2147/ott.s291896] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 03/16/2021] [Indexed: 12/14/2022] Open
Abstract
Digestive system carcinomas are one of the leading causes of cancer-related deaths worldwide. G protein-coupled estrogen receptor (GPER), a novel estrogen receptor, has been recognized as an important mediator in numerous cancer types. Recently, the function and clinical significance of GPER in digestive system carcinomas has been a subject of interest. Increasing evidence has revealed that GPER plays an important role as a potential biomarker in digestive system carcinomas. This work summarizes the recent literature and focuses on the emerging functional role of GPER in digestive system carcinomas, including gastric cancer, hepatocellular carcinoma, pancreatic cancer, and colorectal cancer. The potential application of GPER in novel strategies for the diagnosis and treatment of digestive system carcinomas is discussed and highlighted.
Collapse
Affiliation(s)
- Yu-An Qiu
- Department of Critical Care Medicine, Jiangxi Cancer Hospital, Nanchang University Cancer Hospital, Nanchang, 330029, People's Republic of China
| | - Jianping Xiong
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, People's Republic of China
| | - Tenghua Yu
- Department of Breast Surgery, Jiangxi Cancer Hospital, Nanchang University Cancer Hospital, Nanchang, 330029, People's Republic of China
| |
Collapse
|
48
|
Niță AR, Knock GA, Heads RJ. Signalling mechanisms in the cardiovascular protective effects of estrogen: With a focus on rapid/membrane signalling. Curr Res Physiol 2021; 4:103-118. [PMID: 34746830 PMCID: PMC8562205 DOI: 10.1016/j.crphys.2021.03.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 03/11/2021] [Accepted: 03/17/2021] [Indexed: 12/22/2022] Open
Abstract
In modern society, cardiovascular disease remains the biggest single threat to life, being responsible for approximately one third of worldwide deaths. Male prevalence is significantly higher than that of women until after menopause, when the prevalence of CVD increases in females until it eventually exceeds that of men. Because of the coincidence of CVD prevalence increasing after menopause, the role of estrogen in the cardiovascular system has been intensively researched during the past two decades in vitro, in vivo and in observational studies. Most of these studies suggested that endogenous estrogen confers cardiovascular protective and anti-inflammatory effects. However, clinical studies of the cardioprotective effects of hormone replacement therapies (HRT) not only failed to produce proof of protective effects, but also revealed the potential harm estrogen could cause. The "critical window of hormone therapy" hypothesis affirms that the moment of its administration is essential for positive treatment outcomes, pre-menopause (3-5 years before menopause) and immediately post menopause being thought to be the most appropriate time for intervention. Since many of the cardioprotective effects of estrogen signaling are mediated by effects on the vasculature, this review aims to discuss the effects of estrogen on vascular smooth muscle cells (VSMCs) and endothelial cells (ECs) with a focus on the role of estrogen receptors (ERα, ERβ and GPER) in triggering the more recently discovered rapid, or membrane delimited (non-genomic), signaling cascades that are vital for regulating vascular tone, preventing hypertension and other cardiovascular diseases.
Collapse
Affiliation(s)
- Ana-Roberta Niță
- School of Bioscience Education, Faculty of Life Sciences and Medicine, King’s College London, UK
| | - Greg A. Knock
- School of Bioscience Education, Faculty of Life Sciences and Medicine, King’s College London, UK
- School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, UK
| | - Richard J. Heads
- School of Bioscience Education, Faculty of Life Sciences and Medicine, King’s College London, UK
- Cardiovascular Research Section, King’s BHF Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, Faculty of Life Sciences and Medicine, King’s College London, UK
| |
Collapse
|
49
|
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
|
50
|
Qiu YA, Xiong J, Fu Q, Dong Y, Liu M, Peng M, Jin W, Zhou L, Xu X, Huang X, Fu A, Xu G, Tu G, Yu T. GPER-Induced ERK Signaling Decreases Cell Viability of Hepatocellular Carcinoma. Front Oncol 2021; 11:638171. [PMID: 33767999 PMCID: PMC7985169 DOI: 10.3389/fonc.2021.638171] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 01/25/2021] [Indexed: 12/15/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is an aggressive malignancy with a poor prognosis. Effective biomarkers and specific therapeutic targets for HCC are therefore urgently needed. G protein-coupled estrogen receptor (GPER) plays a crucial role in numerous cancer types; however, its functions in HCC require further exploration. In the present study, we found a remarkable difference in GPER staining between tumor tissue (100/141, 70.9%) and matched non-tumor tissue (27/30, 90.0%). Compared with the GPER-negative patients, the GPER-positive patients with HCC were closely associated with female sex, negative hepatitis B surface antigen, small tumor size, low serum alpha fetoprotein level, and longer overall survival. Treatment with GPER-specific agonist G1 led to the sustained and transient activation of the EGFR/ERK and EGFR/AKT signaling pathways, respectively, in the HCC cell lines HCCLM3 and SMMC-7721, which express high levels of GPER. Interestingly, G1-induced EGFR/ERK signaling, rather than EGFR/AKT signaling mediated by GPER, was involved in decreasing cell viability by blocking cell cycle progression, thereby promoting apoptosis and inhibiting cell growth. Clinical analysis indicated that simultaneous high expression of GPER and phosphorylated-ERK (p-ERK) predicted improved prognosis for HCC. Finally, the activation of GPER/ERK signaling remarkably suppressed tumor growth in an HCC xenograft model, and this result was consistent with the in vitro data. Our findings suggest that specific activation of the GPER/ERK axis may serve as a novel tumor-suppressive mechanism and that this axis could be a therapeutic target for HCC.
Collapse
Affiliation(s)
- Yu-An Qiu
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, China.,Department of Critical Care Medicine, Jiangxi Cancer Hospital, Nanchang University Cancer Hospital, Nanchang, China
| | - Jianping Xiong
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Qin Fu
- Department of Breast Surgery, Jiangxi Cancer Hospital, Nanchang University Cancer Hospital, Nanchang, China
| | - Yun Dong
- Department of Breast Surgery, Jiangxi Cancer Hospital, Nanchang University Cancer Hospital, Nanchang, China
| | - Manran Liu
- Key Laboratory of Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Meixi Peng
- Key Laboratory of Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Wenjian Jin
- Department of Elderly Oncology, Jiangxi Cancer Hospital, Nanchang University Cancer Hospital, Nanchang, China
| | - Lixia Zhou
- Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xue Xu
- Department of Ultrasonography, Jiangxi Cancer Hospital, Nanchang University Cancer Hospital, Nanchang, China
| | - Xianming Huang
- Department of Pathology, Jiangxi Cancer Hospital, Nanchang University Cancer Hospital, Nanchang, China
| | - Airong Fu
- Department of Pathology, Jiangxi Cancer Hospital, Nanchang University Cancer Hospital, Nanchang, China
| | - Guohui Xu
- Department of Hepatobiliary Surgery, Jiangxi Cancer Hospital, Nanchang University Cancer Hospital, Nanchang, China
| | - Gang Tu
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Tenghua Yu
- Department of Breast Surgery, Jiangxi Cancer Hospital, Nanchang University Cancer Hospital, Nanchang, China
| |
Collapse
|