1
|
Zhang Y, Du C, Zhang SQ, Yu HX, Mo HL, Yang QY, Li Y. Missense mutations of GPER1 in breast invasive carcinoma: Exploring gene expression, signal transduction and immune cell infiltration with insights from cellular pharmacology. Biomed Rep 2025; 22:22. [PMID: 39720300 PMCID: PMC11668130 DOI: 10.3892/br.2024.1900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 10/15/2024] [Indexed: 12/26/2024] Open
Abstract
G protein-coupled estrogen receptor 1 (GPER1) plays a crucial role in the progression of breast cancer and has emerged as a promising therapeutic target. However, while missense mutations in GPER1 have been detected in breast invasive carcinoma (BIC) samples, the resulting molecular, cellular and pharmacological changes remain unclear. The present study categorized BIC samples from The Cancer Genome Atlas database based on mutation information available in the cBioPortal database. Subsequently, survival analysis was conducted and the samples screened for differentially expressed genes (DEGs). Using these DEGs, the present study performed Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses, protein-protein interaction network analysis and hub gene selection. After assessing the prognostic value of hub genes, the immune cell infiltration between mutant and wild-type (WT) groups was analyzed. Finally, a luciferase reporter system was used to assess the cyclic AMP (cAMP) production mediated by GPER1 following treatment with the agonist G-1 for each mutation. The results revealed a significant decrease in progression-free survival and disease-specific survival in the GPER1 mutant group compared with the WT group. Gene expression analysis identified 60 DEGs, all of which were upregulated and significantly enriched in GO terms related to tumor progression, such as organic anion transport, glycosaminoglycan binding and monoatomic ion-gated channel activity. DEGs were also significantly enriched in the PI3K-Akt signaling pathway in KEGG. Hub gene selection and prognostic evaluation identified three genes significantly associated with survival: IL33, STAB2 and CFTR. Immune cell infiltration analysis revealed a significant decrease in CD8 T cell content in the GPER1 mutant group compared with the WT group. Luciferase reporter assays demonstrated that four missense mutations in GPER1 (L129M, E218Q, S235F and A345G) significantly attenuated the induction of cyclic adenosine monophosphate production mediated by its agonist. These findings provided valuable insights for the design of breast cancer drugs targeting GPER1 and for precision medicine initiatives.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Chong Du
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Shu-Qun Zhang
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Hui-Xia Yu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, P.R. China
| | - Hao-Lin Mo
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, P.R. China
| | - Qi-Yuan Yang
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Yang Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, P.R. China
| |
Collapse
|
2
|
Tirado-Garibay AC, Ruiz-Barcenas B, Rescala-Ponce de León JI, Ochoa-Zarzosa A, López-Meza JE. The GPR30 Receptor Is Involved in IL-6-Induced Metastatic Properties of MCF-7 Luminal Breast Cancer Cells. Int J Mol Sci 2024; 25:8988. [PMID: 39201674 PMCID: PMC11354767 DOI: 10.3390/ijms25168988] [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: 07/23/2024] [Revised: 08/12/2024] [Accepted: 08/17/2024] [Indexed: 09/03/2024] Open
Abstract
Luminal breast cancer has a high incidence worldwide and poses a severe health threat. Estrogen receptor alpha (ER-α) is activated by 17β-estradiol (E2), and its overexpression promotes cancerous characteristics. Luminal breast cancer is an epithelial type; however, the cytokine IL-6, secreted by cells within the tumor microenvironment, stimulates the epithelial-to-mesenchymal transition (EMT) and promotes metastasis. Also, IL-6 decreases ER-α levels, favoring the tamoxifen (TMX) resistance development. However, genes under E2 regulation continue to be expressed even though this receptor is absent. GPR30 is an alternative E2 receptor present in both luminal and aggressive triple-negative breast cancer and is related to TMX resistance and cancer progression. The roles of GPR30 and IL-6 in metastasis have been individually established; however, their interplay remains unexplored. This study aims to elucidate the role of GPR30 in IL-6-induced metastatic properties of MCF-7 luminal breast cancer cells. Results showed that GPR30 contributes to the E2-induced MCF-7 proliferation because its inhibition with the antagonist G15 and the Pertussis toxin (PTX) reduced it. Besides, GPR30 upregulated vimentin and downregulated E-cadherin levels in MCF-7 and TMX-resistant (R-TMX) cells and is also involved in the IL-6-induced migration, invasion, and TMX resistance in MCF-7 cells. In addition, in MDA-MB-231 triple-negative cells, both basal and IL-6-induced metastatic properties were related to GPR30 activity. These results indicate that the GPR30 receptor regulates the EMT induced by IL-6 in breast cancer cells.
Collapse
Affiliation(s)
| | | | | | | | - Joel E. López-Meza
- Centro Multidisciplinario de Estudios en Biotecnología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Michoacana de San Nicolás de Hidalgo, Km 9.5 Carretera Morelia-Zinapécuaro, Posta Veterinaria, Morelia 58893, Michoacán, Mexico; (A.C.T.-G.); (B.R.-B.); (J.I.R.-P.d.L.); (A.O.-Z.)
| |
Collapse
|
3
|
Abbas MA, Al-Kabariti AY, Sutton C. Comprehensive understanding of the role of GPER in estrogen receptor-alpha negative breast cancer. J Steroid Biochem Mol Biol 2024; 241:106523. [PMID: 38636681 DOI: 10.1016/j.jsbmb.2024.106523] [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: 09/03/2023] [Revised: 11/01/2023] [Accepted: 04/14/2024] [Indexed: 04/20/2024]
Abstract
G protein-coupled estrogen receptor (GPER) plays a prominent role in facilitating the rapid, non-genomic signaling of estrogens in breast cancer cells. Herein, a comprehensive overview of the role of GPER in ER-ɑ-negative breast cancer is provided. Activation of GPER affected proliferation, metastasis and epithelial mesenchymal transition in ER-ɑ negative breast cancer cells. Clinical studies have demonstrated that GPER positivity was strongly correlated with larger tumor size and advanced clinical stage, suggesting that GPER/ERK signaling may play a role in promoting tumor progression. Strong evidence existed that environmental contaminants like bisphenol A have a carcinogenic potential mediated by GPER activation. The complexity of the cross talk between GPER and other receptors including ER-β, ER-α36, Estrogen-related receptor α (ERRα) and androgen receptor has been discussed. The potential utility of small molecules and phytoestrogens targeting GPER, adds valuable insights into its therapeutic potential. This review holds promises in advancing our understanding of GPER role in ER-ɑ-negative breast cancer. Overall, the consequences of GPER activation are still an area of active research and the implication are not entirely clear.
Collapse
Affiliation(s)
- Manal A Abbas
- Department of Medical Laboratory Sciences, Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman 19328, Jordan; Pharmacological and Diagnostic Research Centre, Al-Ahliyya Amman University, Amman 19328, Jordan
| | - Aya Y Al-Kabariti
- Department of Biopharmaceutics and Clinical Pharmacy, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman 19328, Jordan; Pharmacological and Diagnostic Research Centre, Al-Ahliyya Amman University, Amman 19328, Jordan.
| | - Chris Sutton
- School of Chemistry and Biosciences, University of Bradford, Bradford BD7 1DP, UK
| |
Collapse
|
4
|
Tutzauer J, Serafin DS, Schmidt T, Olde B, Caron KM, Leeb-Lundberg LMF. G protein-coupled estrogen receptor (GPER)/GPR30 forms a complex with the β 1-adrenergic receptor, a membrane-associated guanylate kinase (MAGUK) scaffold protein, and protein kinase A anchoring protein (AKAP) 5 in MCF7 breast cancer cells. Arch Biochem Biophys 2024; 752:109882. [PMID: 38211639 PMCID: PMC11481754 DOI: 10.1016/j.abb.2024.109882] [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: 08/03/2023] [Revised: 12/26/2023] [Accepted: 01/08/2024] [Indexed: 01/13/2024]
Abstract
G protein-coupled receptor 30 (GPR30), also named G protein-coupled estrogen receptor (GPER), and the β1-adrenergic receptor (β1AR) are G protein-coupled receptors (GPCR) that are implicated in breast cancer progression. Both receptors contain PSD-95/Discs-large/ZO-1 homology (PDZ) motifs in their C-terminal tails through which they interact in the plasma membrane with membrane-associated guanylate kinase (MAGUK) scaffold proteins, and in turn protein kinase A anchoring protein (AKAP) 5. GPR30 constitutively and PDZ-dependently inhibits β1AR-mediated cAMP production. We hypothesized that this inhibition is a consequence of a plasma membrane complex of these receptors. Using co-immunoprecipitation, confocal immunofluorescence microscopy, and bioluminescence resonance energy transfer (BRET), we show that GPR30 and β1AR reside in close proximity in a plasma membrane complex when transiently expressed in HEK293. Deleting the GPR30 C-terminal PDZ motif (-SSAV) does not interfere with the receptor complex, indicating that the complex is not PDZ-dependent. MCF7 breast cancer cells express GPR30, β1AR, MAGUKs, and AKAP5 in the plasma membrane, and co-immunoprecipitation revealed that these proteins exist in close proximity also under native conditions. Furthermore, expression of GPR30 in MCF7 cells constitutively and PDZ-dependently inhibits β1AR-mediated cAMP production. AKAP5 also inhibits β1AR-mediated cAMP production, which is not additive with GPR30-promoted inhibition. These results argue that GPR30 and β1AR form a PDZ-independent complex in MCF7 cells through which GPR30 constitutively and PDZ-dependently inhibits β1AR signaling via receptor interaction with MAGUKs and AKAP5.
Collapse
Affiliation(s)
- Julia Tutzauer
- Department of Experimental Medical Science, Lund University, 22184, Lund, Sweden
| | - D Stephen Serafin
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Tobias Schmidt
- Wallenberg Center for Molecular Medicine, Department of Clinical Sciences Lund, Division of Pediatrics, Lund University, 22184, Lund, Sweden
| | - Björn Olde
- Department of Clinical Sciences, Division of Cardiology, Lund University, 22184, Lund, Sweden
| | - Kathleen M Caron
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | | |
Collapse
|
5
|
Gopinath P, Oviya RP, Gopisetty G. Oestrogen receptor-independent actions of oestrogen in cancer. Mol Biol Rep 2023; 50:9497-9509. [PMID: 37731028 DOI: 10.1007/s11033-023-08793-8] [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: 06/08/2023] [Accepted: 08/30/2023] [Indexed: 09/22/2023]
Abstract
Oestrogen, the primary female sex hormone, plays a significant role in tumourigenesis. The major pathway for oestrogen is via binding to its receptor [oestrogen receptor (ERα or β)], followed by nuclear translocation and transcriptional regulation of target genes. Almost 70% of breast tumours are ER + , and endocrine therapies with selective ER modulators (tamoxifen) have been successfully applied. As many as 25% of tamoxifen-treated patients experience disease relapse within 5 years upon completion of chemotherapy. In such cases, the ER-independent oestrogen actions provide a plausible explanation for the resistance, as well as expands the existing horizon of available drug targets. ER-independent oestrogen signalling occurs via one of the following pathways: signalling through membrane receptors, oxidative catabolism giving rise to genotoxic metabolites, effects on mitochondria and redox balance, and induction of inflammatory cytokines. The current review focuses on the non-classical oestrogen signalling, its role in cancer, and its clinical significance.
Collapse
Affiliation(s)
- Prarthana Gopinath
- Department of Molecular Oncology, Cancer Institute (WIA), Adyar, Chennai, 600020, India
| | - Revathi Paramasivam Oviya
- Department of Biotechnology, School of Bio and Chemical Engineering, Sathyabama Institute of Science and Technology, Chennai, 600119, India
| | - Gopal Gopisetty
- Department of Molecular Oncology, Cancer Institute (WIA), Adyar, Chennai, 600020, India.
| |
Collapse
|
6
|
Locomotion Outcome Improvement in Mice with Glioblastoma Multiforme after Treatment with Anastrozole. Brain Sci 2023; 13:brainsci13030496. [PMID: 36979306 PMCID: PMC10046174 DOI: 10.3390/brainsci13030496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/09/2023] [Accepted: 03/10/2023] [Indexed: 03/17/2023] Open
Abstract
Glioblastoma Multiforme (GBM) is a tumor that infiltrates several brain structures. GBM is associated with abnormal motor activities resulting in impaired mobility, producing a loss of functional motor independence. We used a GBM xenograft implanted in the striatum to analyze the changes in Y (vertical) and X (horizontal) axis displacement of the metatarsus, ankle, and knee. We analyzed the steps dissimilarity factor between control and GBM mice with and without anastrozole. The body weight of the untreated animals decreased compared to treated mice. Anastrozole reduced the malignant cells and decreased GPR30 and ERα receptor expression. In addition, we observed a partial recovery in metatarsus and knee joint displacement (dissimilarity factor). The vertical axis displacement of the GBM+anastrozole group showed a difference in the right metatarsus, right knee, and left ankle compared to the GBM group. In the horizontal axis displacement of the right metatarsus, ankle, and knee, the GBM+anastrozole group exhibited a difference at the last third of the step cycle compared to the GBM group. Thus, anastrozole partially modified joint displacement. The dissimilarity factor and the vertical and horizontal displacements study will be of interest in GBM patients with locomotion alterations. Hindlimb displacement and gait locomotion analysis could be a valuable methodological tool in experimental and clinical studies to help diagnose locomotive deficits related to GBM.
Collapse
|
7
|
Roles of Estrogen, Estrogen Receptors, and Estrogen-Related Receptors in Skeletal Muscle: Regulation of Mitochondrial Function. Int J Mol Sci 2023; 24:ijms24031853. [PMID: 36768177 PMCID: PMC9916347 DOI: 10.3390/ijms24031853] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/13/2023] [Accepted: 01/14/2023] [Indexed: 01/19/2023] Open
Abstract
Estrogen is an essential sex steroid hormone that functions primarily in female reproductive system, as well as in a variety of tissues and organs with pleiotropic effects, such as in cardiovascular, nervous, immune, and musculoskeletal systems. Women with low estrogen, as exemplified by those in postmenopause, are therefore prone to suffer from various disorders, i.e., cardiovascular disease, dementia, metabolic syndrome, osteoporosis, sarcopenia, frailty, and so on. Estrogen regulates the expression of its target genes by binding to its cognate receptors, estrogen receptors (ERs) α and β. Notably, the estrogen-related receptors (ERRs) α, β, and γ are originally identified as orphan receptors that share substantial structural homology and common transcriptional targets with ERs. Accumulating evidence suggests that ERs and ERRs play crucial roles in skeletal muscles, such as muscle mass maintenance, muscle exercise physiology, and muscle regeneration. In this article, we review potential regulatory roles of ERs and ERRs in muscle physiology, particularly with regard to mitochondrial function and metabolism.
Collapse
|
8
|
Muhammad A, Forcados GE, Yusuf AP, Abubakar MB, Sadiq IZ, Elhussin I, Siddique MAT, Aminu S, Suleiman RB, Abubakar YS, Katsayal BS, Yates CC, Mahavadi S. Comparative G-Protein-Coupled Estrogen Receptor (GPER) Systems in Diabetic and Cancer Conditions: A Review. Molecules 2022; 27:molecules27248943. [PMID: 36558071 PMCID: PMC9786783 DOI: 10.3390/molecules27248943] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/05/2022] [Accepted: 12/08/2022] [Indexed: 12/23/2022] Open
Abstract
For many patients, diabetes Mellitus and Malignancy are frequently encountered comorbidities. Diabetes affects approximately 10.5% of the global population, while malignancy accounts for 29.4 million cases each year. These troubling statistics indicate that current treatment approaches for these diseases are insufficient. Alternative therapeutic strategies that consider unique signaling pathways in diabetic and malignancy patients could provide improved therapeutic outcomes. The G-protein-coupled estrogen receptor (GPER) is receiving attention for its role in disease pathogenesis and treatment outcomes. This review aims to critically examine GPER' s comparative role in diabetes mellitus and malignancy, identify research gaps that need to be filled, and highlight GPER's potential as a therapeutic target for diabetes and malignancy management. There is a scarcity of data on GPER expression patterns in diabetic models; however, for diabetes mellitus, altered expression of transport and signaling proteins has been linked to GPER signaling. In contrast, GPER expression in various malignancy types appears to be complex and debatable at the moment. Current data show inconclusive patterns of GPER expression in various malignancies, with some indicating upregulation and others demonstrating downregulation. Further research should be conducted to investigate GPER expression patterns and their relationship with signaling pathways in diabetes mellitus and various malignancies. We conclude that GPER has therapeutic potential for chronic diseases such as diabetes mellitus and malignancy.
Collapse
Affiliation(s)
- Aliyu Muhammad
- Center for Cancer Research, Department of Biology, Tuskegee University, Tuskegee, AL 36088, USA
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria P.M.B. 1044, Nigeria
| | | | - Abdurrahman Pharmacy Yusuf
- Department of Biochemistry, School of Life Sciences, Federal University of Technology, Minna P.M.B. 65, Nigeria
| | - Murtala Bello Abubakar
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, Usmanu Danfodiyo University, Sokoto P.M.B. 2254, Nigeria
- Centre for Advanced Medical Research & Training (CAMRET), Usmanu Danfodiyo University, Sokoto P.M.B. 2254, Nigeria
| | - Idris Zubairu Sadiq
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria P.M.B. 1044, Nigeria
| | - Isra Elhussin
- Center for Cancer Research, Department of Biology, Tuskegee University, Tuskegee, AL 36088, USA
| | - Md Abu Talha Siddique
- Center for Cancer Research, Department of Biology, Tuskegee University, Tuskegee, AL 36088, USA
| | - Suleiman Aminu
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria P.M.B. 1044, Nigeria
| | - Rabiatu Bako Suleiman
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria P.M.B. 1044, Nigeria
| | - Yakubu Saddeeq Abubakar
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria P.M.B. 1044, Nigeria
| | - Babangida Sanusi Katsayal
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria P.M.B. 1044, Nigeria
| | - Clayton C Yates
- Center for Cancer Research, Department of Biology, Tuskegee University, Tuskegee, AL 36088, USA
| | - Sunila Mahavadi
- Center for Cancer Research, Department of Biology, Tuskegee University, Tuskegee, AL 36088, USA
| |
Collapse
|
9
|
Xu T, Ma D, Chen S, Tang R, Yang J, Meng C, Feng Y, Liu L, Wang J, Luo H, Yu K. High GPER expression in triple-negative breast cancer is linked to pro-metastatic pathways and predicts poor patient outcomes. NPJ Breast Cancer 2022; 8:100. [PMID: 36042244 PMCID: PMC9427744 DOI: 10.1038/s41523-022-00472-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 08/09/2022] [Indexed: 11/09/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a particularly aggressive and heterogeneous disease with few effective targeted therapies and precision therapeutic options over a long period. It is generally considered that TNBC is an estrogen-independent breast cancer, while a new estrogen receptor, namely G protein-coupled estrogen receptor (GPER), is demonstrated to mediate estrogenic actions in TNBC. Based on our transcriptomic analysis, expression of GPER was correlated with clinicopathological variables and survival of 360 TNBC patients. GPER expression at mRNA level was significantly correlated with immunohistochemistry scoring in 12 randomly chosen samples. According to the cutoff value, 26.4% (95/360) of patients showed high GPER expression and significant correlation with the mRNA subtype of TNBC (P = 0.001), total metastatic events (P = 0.019) and liver metastasis (P = 0.011). In quantitative comparison, GPER abundance is correlated with the high-risk subtype of TNBC. At a median follow-up interval of 67.1 months, a significant trend towards reduced distant metastasis-free survival (DMFS) (P = 0.014) was found by Kaplan–Meier analysis in patients with high GPER expression. Furthermore, univariate analysis confirmed that GPER was a significant prognostic factor for DMFS in TNBC patients. Besides, high GPER expression was significantly linked to the worse survival in patients with lymph node metastasis, TNM stage III as well as nuclear grade G3 tumors. Transcriptome-based bioinformatics analysis revealed that GPER was linked to pro-metastatic pathways in our cohort. These results may supply new insights into GPER-mediated estrogen carcinogenesis in TNBC, thus providing a potential strategy for endocrine therapy of TNBC.
Collapse
Affiliation(s)
- Ting Xu
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Road, Chongqing, 400010, People's Republic of China
| | - Ding Ma
- Department of Breast Surgery, Precision Cancer Medicine Center, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai, 200032, People's Republic of China
| | - Sheng Chen
- Department of Breast Surgery, Precision Cancer Medicine Center, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai, 200032, People's Republic of China
| | - Rui Tang
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Jianling Yang
- Department of Thyroid and Breast Surgery, Weihai Municipal Hospital, 70 Heping Road, Huancui District, Weihai, Shandong, 264200, People's Republic of China
| | - Chunhui Meng
- Department of Thyroid and Breast Surgery, Heze Municipal Hospital, 2888 Caozhou West Road, Heze, Shandong, 274031, People's Republic of China
| | - Yang Feng
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Road, Chongqing, 400010, People's Republic of China
| | - Li Liu
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Road, Chongqing, 400010, People's Republic of China
| | - Jiangfen Wang
- Department of General Surgery, Shanxi Provincial People's Hospital, Taiyuan, Shanxi, 030000, People's Republic of China
| | - Haojun Luo
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Road, Chongqing, 400010, People's Republic of China.
| | - Keda Yu
- Department of Breast Surgery, Precision Cancer Medicine Center, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai, 200032, People's Republic of China.
| |
Collapse
|
10
|
Bubb M, Beyer ASL, Dasgupta P, Kaemmerer D, Sänger J, Evert K, Wirtz RM, Schulz S, Lupp A. Assessment of G Protein-Coupled Oestrogen Receptor Expression in Normal and Neoplastic Human Tissues Using a Novel Rabbit Monoclonal Antibody. Int J Mol Sci 2022; 23:ijms23095191. [PMID: 35563581 PMCID: PMC9099907 DOI: 10.3390/ijms23095191] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/03/2022] [Accepted: 05/04/2022] [Indexed: 12/23/2022] Open
Abstract
In addition to the classical oestrogen receptors, ERα and ERβ, a G protein-coupled oestrogen receptor (GPER) has been identified that primarily mediates the rapid, non-genomic signalling of oestrogens. Data on GPER expression at the protein level are contradictory; therefore, the present study was conducted to re-evaluate GPER expression by immunohistochemistry to obtain broad GPER expression profiles in human non-neoplastic and neoplastic tissues, especially those not investigated in this respect so far. We developed and thoroughly characterised a novel rabbit monoclonal anti-human GPER antibody, 20H15L21, using Western blot analyses and immunocytochemistry. The antibody was then applied to a large series of formalin-fixed, paraffin-embedded human tissue samples. In normal tissue, GPER was identified in distinct cell populations of the cortex and the anterior pituitary; islets and pancreatic ducts; fundic glands of the stomach; the epithelium of the duodenum and gallbladder; hepatocytes; proximal tubules of the kidney; the adrenal medulla; and syncytiotrophoblasts and decidua cells of the placenta. GPER was also expressed in hepatocellular, pancreatic, renal, and endometrial cancers, pancreatic neuroendocrine tumours, and pheochromocytomas. The novel antibody 20H15L21 will serve as a valuable tool for basic research and the identification of GPER-expressing tumours during histopathological examinations.
Collapse
Affiliation(s)
- Maria Bubb
- Institute of Pharmacology and Toxicology, Jena University Hospital, 07747 Jena, Germany; (M.B.); (A.-S.L.B.); (P.D.); (S.S.)
| | - Anna-Sophia Lieselott Beyer
- Institute of Pharmacology and Toxicology, Jena University Hospital, 07747 Jena, Germany; (M.B.); (A.-S.L.B.); (P.D.); (S.S.)
| | - Pooja Dasgupta
- Institute of Pharmacology and Toxicology, Jena University Hospital, 07747 Jena, Germany; (M.B.); (A.-S.L.B.); (P.D.); (S.S.)
| | - Daniel Kaemmerer
- Department of General and Visceral Surgery, Zentralklinik Bad Berka, 99438 Bad Berka, Germany;
| | - Jörg Sänger
- Laboratory of Pathology and Cytology Bad Berka, 99438 Bad Berka, Germany;
| | - Katja Evert
- Department of Pathology, University of Regensburg, 93053 Regensburg, Germany;
- Institute of Pathology, University Medicine of Greifswald, 17475 Greifswald, Germany
| | - Ralph M. Wirtz
- STRATIFYER Molecular Pathology GmbH, 50935 Cologne, Germany;
| | - Stefan Schulz
- Institute of Pharmacology and Toxicology, Jena University Hospital, 07747 Jena, Germany; (M.B.); (A.-S.L.B.); (P.D.); (S.S.)
| | - Amelie Lupp
- Institute of Pharmacology and Toxicology, Jena University Hospital, 07747 Jena, Germany; (M.B.); (A.-S.L.B.); (P.D.); (S.S.)
- Correspondence: ; Tel.: +49-3641-9325678; Fax: +49-3641-9325652
| |
Collapse
|
11
|
Torres-López L, Olivas-Aguirre M, Villatoro-Gómez K, Dobrovinskaya O. The G-Protein–Coupled Estrogen Receptor Agonist G-1 Inhibits Proliferation and Causes Apoptosis in Leukemia Cell Lines of T Lineage. Front Cell Dev Biol 2022; 10:811479. [PMID: 35237599 PMCID: PMC8882838 DOI: 10.3389/fcell.2022.811479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 01/13/2022] [Indexed: 11/25/2022] Open
Abstract
The G-protein–coupled estrogen receptor (GPER) mediates non-genomic action of estrogen. Due to its differential expression in some tumors as compared to the original healthy tissues, the GPER has been proposed as a therapeutic target. Accordingly, the non-steroidal GPER agonist G-1, which has often demonstrated marked cytotoxicity in experimental models, has been suggested as a novel anticancer agent for several sensitive tumors. We recently revealed that cell lines derived from acute T-cell (query) lymphoblastic leukemia (T-ALL) express the GPER. Here, we address the question whether G-1 is cytotoxic to T-ALL. We have shown that G-1 causes an early rise of intracellular Ca2+, arrests the cell cycle in G2/M, reduces viability, and provokes apoptosis in T-ALL cell lines. Importantly, G-1 caused destabilization and depolymerization of microtubules. We assume that it is a disturbance of the cytoskeleton that causes G-1 cytotoxic and cytostatic effects in our model. The observed cytotoxic effects, apparently, were not triggered by the interaction of G-1 with the GPER as pre-incubation with the highly selective GPER antagonist G-36 was ineffective in preventing the cytotoxicity of G-1. However, G-36 prevented the intracellular Ca2+ rise provoked by G-1. Finally, G-1 showed only a moderate negative effect on the activation of non-leukemic CD4+ lymphocytes. We suggest G-1 as a potential antileukemic drug.
Collapse
|
12
|
Zhang D, Wang J, Chen H, Yan S. Cytoplasmic G Protein-Coupled Estrogen Receptor 1 as a Prognostic Indicator of Breast Cancer: A Meta-Analysis. Technol Cancer Res Treat 2022; 21:15330338221131664. [PMID: 36254557 PMCID: PMC9580104 DOI: 10.1177/15330338221131664] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Purpose: To determine whether G protein-coupled estrogen receptor 1 (GPER1) is a suitable biomarker to predict the treatment outcome of breast cancer (BC). Methods: A meta-analysis of the literature was performed to clarify the correlation between GPER1 protein expression and BC outcome. The relationship between GPER1 mRNA expression and survival was analyzed using Breast Cancer Gene-Expression Miner (bc-GenExMiner) v4.6 software. Results: Six studies involving 2697 patients were included in the meta-analysis. Four studies reported the correlation between GPER1 protein expression and relapse-free survival (RFS) and 4 others reported the impact of GPER1 protein expression on overall survival (OS). The results showed that high GPER1 protein expression was not associated with RFS (hazard ratio [HR] = 1.58; 95% confidence interval [CI] = 0.71-3.48; P = .26) or OS (HR = 1.18; 95% CI = 0.64-2.18; P = .60). Subgroup analysis suggested that nuclear expression of GPER1 was not associated with OS (HR = 0.91; 95% CI = 0.77-1.08; P = .30), but high expression of cytoplasmic GPER1 was significantly associated with longer OS (HR = 0.69; 95% CI = 0.55-0.86; P = .001). Furthermore, the association of GPER1 mRNA and OS of BC patients was analyzed using bc-GenExMiner v4.6. Two data sets involving 4016 patients were included in the analysis. The targeted prognostic analysis results showed that high mRNA expression of GPER1 was predictive of better OS in BC patients (HR = 0.71; 95% CI = 0.59-0.86; P = .0005), which was remarkably similar to the result of cytoplasmic GPER1. Further subgroup analysis demonstrated that high mRNA expression of GPER1 was predictive of better OS in estrogen receptor (ER)-positive, but not ER-negative or triple-negative BC patients. Conclusions: High mRNA and cytoplasmic protein expression of GPER1 were predictive of better OS of BC patients.
Collapse
Affiliation(s)
- Duo Zhang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Jinpeng Wang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Hong Chen
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Shunchao Yan
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| |
Collapse
|
13
|
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
|
14
|
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
|
15
|
Tutzauer J, Gonzalez de Valdivia E, Swärd K, Alexandrakis Eilard I, Broselid S, Kahn R, Olde B, Leeb-Lundberg LMF. Ligand-independent G protein-coupled Estrogen Receptor (GPER)/GPR30 Activity: Lack of receptor-dependent effects of G-1 and 17β-estradiol.. Mol Pharmacol 2021; 100:271-282. [PMID: 34330822 PMCID: PMC8626787 DOI: 10.1124/molpharm.121.000259] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 06/06/2021] [Indexed: 11/22/2022] Open
Abstract
G protein–coupled receptor 30 (GPR30) is a membrane receptor reported to bind 17β-estradiol (E2) and mediate rapid nongenomic estrogen responses, hence also named G protein–coupled estrogen receptor. G-1 is a proposed GPR30-specific agonist that has been used to implicate the receptor in several pathophysiological events. However, controversy surrounds the role of GPR30 in G-1 and E2 responses. We investigated GPR30 activity in the absence and presence of G-1 and E2 in several eukaryotic systems ex vivo and in vitro in the absence and presence of the receptor. Ex vivo activity was addressed using the caudal artery from wild-type (WT) and GPR30 knockout (KO) mice, and in vitro activity was addressed using a HeLa cell line stably expressing a synthetic multifunctional promoter (nuclear factor κB, signal transducer and activator of transcription, activator protein 1)–luciferase construct (HFF11 cells) and a human GPR30-inducible T-REx system (T-REx HFF11 cells), HFF11 and human embryonic kidney 293 cells transiently expressing WT GPR30 and GPR30 lacking the C-terminal PDZ (postsynaptic density-95/discs-large /zonula occludens-1 homology) motif SSAV, and yeast Saccharomyces cerevisiae transformed to express GPR30. WT and KO arteries exhibited similar contractile responses to 60 mM KCl and 0.3 μM cirazoline, and G-1 relaxed both arteries with the same potency and efficacy. Furthermore, expression of GPR30 did not introduce any responses to 1 μM G-1 and 0.1 μM E2 in vitro. On the other hand, receptor expression caused considerable ligand-independent activity in vitro, which was receptor PDZ motif-dependent in mammalian cells. We conclude from these results that GPR30 exhibits ligand-independent activity in vitro but no G-1– or E2-stimulated activity in any of the systems used.
Collapse
Affiliation(s)
- Julia Tutzauer
- Department of Experimental Medical Science, Lund University, Sweden
| | | | - Karl Swärd
- Department of Experimental Medical Science, Lund University, Sweden
| | | | - Stefan Broselid
- Department of Experimental Medical Science, Lund University, Sweden
| | - Robin Kahn
- Department of Clinical Sciences Lund, Lund University, Sweden
| | - Björn Olde
- Department of Clinical Sciences Lund, Lund University, Sweden
| | | |
Collapse
|
16
|
Wolfson B, Padget MR, Schlom J, Hodge JW. Exploiting off-target effects of estrogen deprivation to sensitize estrogen receptor negative breast cancer to immune killing. J Immunother Cancer 2021; 9:jitc-2020-002258. [PMID: 34244306 PMCID: PMC8268928 DOI: 10.1136/jitc-2020-002258] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2021] [Indexed: 12/17/2022] Open
Abstract
Background There are highly effective treatment strategies for estrogen receptor (ER)+, progesterone receptor (PR)+, and HER2+ breast cancers; however, there are limited targeted therapeutic strategies for the 10%–15% of women who are diagnosed with triple-negative breast cancer. Here, we hypothesize that ER targeting drugs induce phenotypic changes to sensitize breast tumor cells to immune-mediated killing regardless of their ER status. Methods Real-time cell analysis, flow cytometry, qRT-PCR, western blotting, and multiplexed RNA profiling were performed to characterize ER+ and ER− breast cancer cells and to interrogate the phenotypic effects of ER targeting drugs. Sensitization of breast cancer cells to immune cell killing by the tamoxifen metabolite 4-hydroxytamoxifen (4-OHT) and fulvestrant was determined through in vitro health-donor natural killer cell 111IN-release killing assays. A syngeneic tumor study was performed to validate these findings in vivo. Results Pretreatment with tamoxifen metabolite 4-OHT or fulvestrant resulted in increased natural killer (NK)–mediated cell lysis of both ER+ and ER− breast cancer cells. Through multiplexed RNA profiling analysis of 4-OHT-treated ER+ and ER− cells, we identified increased activation of apoptotic and death receptor signaling pathways and identified G protein-coupled receptor for estrogen (GPR30) engagement as a putative mechanism for immunogenic modulation. Using the specific GPR30 agonist G-1, we demonstrate that targeted activation of GPR30 signaling resulted in increased NK cell killing. Furthermore, we show that knockdown of GPR30 inhibited 4-OHT and fulvestrant mediated increases to NK cell killing, demonstrating this is dependent on GPR30 expression. Moreover, we demonstrate that this mechanism remains active in a 4-OHT-resistant MCF7 cell line, showing that even in patient populations with ER+ tumors that are resistant to the cytotoxic effects of tamoxifen, 4-OHT treatment sensitizes them to immune-mediated killing. Moreover, we find that fulvestrant pretreatment of tumor cells synergizes with the IL-15 superagonist N-803 treatment of NK cells and sensitizes tumor cells to killing by programmed death-ligand 1 (PD-L1) targeting high-affinity natural killer (t-haNK) cells. Finally, we demonstrate that the combination of fulvestrant and N-803 is effective in triple-negative breast cancer in vivo. Conclusion Together, these findings demonstrate a novel effect of ER targeting drugs on the interaction of ER+ and, surprisingly, ER− tumors cells with the immune system. This study is the first to demonstrate the potential use of ER targeting drugs as immunomodulatory agents in an ER agnostic manner and may inform novel immunotherapy strategies in breast cancer.
Collapse
Affiliation(s)
- Benjamin Wolfson
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Michelle R Padget
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Jeffrey Schlom
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - James W Hodge
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
| |
Collapse
|
17
|
Zhou C, Li P, Han M, Gao X. Daidzein stimulates fatty acid-induced fat deposition in C2C12 myoblast cells via the G protein-coupled receptor 30 pathway. Anim Biotechnol 2020; 33:851-863. [PMID: 33164657 DOI: 10.1080/10495398.2020.1842749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Fat deposition in skeletal muscle is an important aspect of improving meat quality. Isoflavones can promote animal anabolism, but whether and how they regulate muscle fat deposition remain largely unclear. In this study, we explored the role and corresponding molecular mechanism of one of the major isoflavones, daidzein, in fat deposition in C2C12 myoblast cells. In the absence of fatty acids (FAs), daidzein did not promote triglyceride synthesis and lipid droplet formation in cells but increased sterol regulatory element-binding protein 1c (SREBP-1c) expression and maturation. In the presence of FAs, daidzein enhanced FAs-induced fat deposition and the SREBP-1c signaling. Daidzein promoted FAs-induced nuclear factor κB1 (NFκB1) phosphorylation and activated the SREBP-1c signaling in a PI3K-dependent manner. G protein-coupled receptor 30 (GPR30) knockdown but not estrogen receptor α (ERα) knockdown blocked the stimulation of daidzein on the PI3K-NFκB1-SREBP-1c signaling pathway, while both knockdown did not affect the stimulation of FAs on this signaling. qRT-PCR and ChIP-qPCR further detected that daidzein stimulated NFκB1-targeted SREBP-1c transcription. Daidzein did not affect ERα expression in cells, but it stimulated GPR30 expression and cytoplasmic localization. These results reveal that daidzein promotes FAs-induced fat deposition through the GPR30 signaling in C2C12 myoblast cells.
Collapse
Affiliation(s)
- Chengjian Zhou
- College of Animal Science, Yangtze University, Jingzhou, China
| | - Ping Li
- College of Animal Science, Yangtze University, Jingzhou, China
| | - Meihong Han
- College of Animal Science, Yangtze University, Jingzhou, China
| | - Xuejun Gao
- College of Animal Science, Yangtze University, Jingzhou, China
| |
Collapse
|
18
|
Chevalier N, Hinault C, Clavel S, Paul-Bellon R, Fenichel P. GPER and Testicular Germ Cell Cancer. Front Endocrinol (Lausanne) 2020; 11:600404. [PMID: 33574796 PMCID: PMC7870790 DOI: 10.3389/fendo.2020.600404] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 12/07/2020] [Indexed: 12/12/2022] Open
Abstract
The G protein-coupled estrogen receptor (GPER), also known as GPR30, is a widely conserved 7-transmembrane-domain protein which has been identified as a novel 17β-estradiol-binding protein that is structurally distinct from the classic oestrogen receptors (ERα and ERβ). There are still conflicting data regarding the exact role and the natural ligand of GPER/GPR30 in reproductive tracts as both male and female knock-out mice are fertile and have no abnormalities of reproductive organs. Testicular germ cell cancers (TGCCs) are the most common malignancy in young males and the most frequent cause of death from solid tumors in this age group. Clinical and experimental studies suggested that estrogens participate in the physiological and pathological control of male germ cell proliferation. In human seminoma cell line, while 17β-estradiol (E2) inhibits in vitro cell proliferation through an ERβ-dependent mechanism, an impermeable E2 conjugate (E2 coupled to BSA), in vitro cell proliferation is stimulated by activating ERK1/2 and protein kinase A through a membrane GPCR that we further identified as GPER/GPR30. The same effect was observed with low but environmentally relevant doses of BPA, an estrogenic endocrine disrupting compound. Furthermore, GPER/GPR30 is specifically overexpressed in seminomas but not in non-seminomas and this overexpression is correlated with an ERβ-downregulation. This GPER/GPR30 overexpression could be linked to some genetic variations, as single nucleotide polymorphisms, which was also reported in other hormone-dependent cancers. We will review here the implication of GPER/GPR30 in TGCCs pathophysiology and the arguments to consider GPER/GPR30 as a potential therapeutic target in humans.
Collapse
Affiliation(s)
- Nicolas Chevalier
- Université Côte d’Azur, CHU, INSERM U1065, C3M, Nice, France
- Université Côte d’Azur, INSERM U1065, C3M, Nice, France
- *Correspondence: Nicolas Chevalier, ;
| | - Charlotte Hinault
- Université Côte d’Azur, CHU, INSERM U1065, C3M, Nice, France
- Université Côte d’Azur, INSERM U1065, C3M, Nice, France
| | | | | | - Patrick Fenichel
- Université Côte d’Azur, CHU, INSERM U1065, C3M, Nice, France
- Université Côte d’Azur, INSERM U1065, C3M, Nice, France
| |
Collapse
|