1
|
Wojciechowska AM, Zając P, Gogola-Mruk J, Kowalik MK, Ptak A. Myo-Inositol and D-Chiro-Inositol Reduce DHT-Stimulated Changes in the Steroidogenic Activity of Adult Granulosa Cell Tumors. Int J Mol Sci 2024; 25:10974. [PMID: 39456756 PMCID: PMC11507073 DOI: 10.3390/ijms252010974] [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: 09/30/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
Considering the properties of myo-inositol (MI) and D-chiro-inositol (DCI), which are well known in polycystic ovary syndrome therapy, and the limitations of adult granulosa cell tumor (AGCT) treatment, especially for androgen-secreting tumors, we studied the role of MI and DCI in the androgen-rich environment of AGCTs. For this purpose, we analyzed the mRNA expression of steroidogenic genes and the secretion of progesterone (P4) and 17β-estradiol (E2) in an unstimulated and/or dihydrotestosterone (DHT)-stimulated environment under MI and DCI influence. Thus, we used the HGrC1 and KGN cell lines as in vitro models of healthy and cancerous granulosa cells. We found that DHT, the most potent androgen, increased E2 secretion and steroidogenic acute regulatory protein (StAR) and cytochrome P450 side-chain cleavage gene (CYP11A1) mRNA expression without affecting 450 aromatase (CYP19A1) in AGCTs. However, after the MI and DCI treatment of KGN cells, both compounds strongly reduced StAR and CYP11A1 expression. Interestingly, in DHT-stimulated KGN cells, only DCI alone and its cotreatment with MI reduced both CYP11A1 mRNA and E2 secretion. These findings suggest that CYP11A1 is responsible for the antiestrogenic effect of DCI in the androgen-rich environment of AGCTs. Therefore, MI and DCI could be used as effective agents in the adjuvant treatment of AGCT, but further detailed studies are needed.
Collapse
Affiliation(s)
- Anna Maria Wojciechowska
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-082 Olsztyn, Poland
| | - Paulina Zając
- Department of Physiology and Toxicology, Institute of Animal Reproduction and Food Research of the Polish Academy of Sciences, 10-747 Olsztyn, Poland; (P.Z.); (M.K.K.)
| | - Justyna Gogola-Mruk
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University, 30-387 Krakow, Poland; (J.G.-M.); (A.P.)
| | - Magdalena Karolina Kowalik
- Department of Physiology and Toxicology, Institute of Animal Reproduction and Food Research of the Polish Academy of Sciences, 10-747 Olsztyn, Poland; (P.Z.); (M.K.K.)
| | - Anna Ptak
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University, 30-387 Krakow, Poland; (J.G.-M.); (A.P.)
| |
Collapse
|
2
|
Monteiro FL, Stepanauskaite L, Archer A, Williams C. Estrogen receptor beta expression and role in cancers. J Steroid Biochem Mol Biol 2024; 242:106526. [PMID: 38657699 DOI: 10.1016/j.jsbmb.2024.106526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 04/06/2024] [Accepted: 04/21/2024] [Indexed: 04/26/2024]
Abstract
Estrogen drives the growth of some cancers, such as breast cancer, via estrogen receptor alpha (ERα). Estrogen also activates ERβ, but whether ERβ is expressed and has a role in different cancers is debated. The use of nonspecific antibodies has contributed to the confusion, and this review delves into ERβ's controversial role in cancer and focuses on tumor expression that can be supported by non-antibody-dependent assays. We discuss its expression at the transcript level and focus on its potential role in lymphoma, granulosa cell tumors, testicular, and adrenal cancers, emphasizing recent findings and the complexities that necessitate further research.
Collapse
Affiliation(s)
- Fátima L Monteiro
- SciLifeLab, Department of Protein Science, KTH Royal Institute of Technology, Solna 171 21, Sweden; Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge 141 83, Sweden
| | - Lina Stepanauskaite
- SciLifeLab, Department of Protein Science, KTH Royal Institute of Technology, Solna 171 21, Sweden; Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge 141 83, Sweden
| | - Amena Archer
- SciLifeLab, Department of Protein Science, KTH Royal Institute of Technology, Solna 171 21, Sweden; Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge 141 83, Sweden
| | - Cecilia Williams
- SciLifeLab, Department of Protein Science, KTH Royal Institute of Technology, Solna 171 21, Sweden; Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge 141 83, Sweden.
| |
Collapse
|
3
|
Pich K, Respekta N, Kurowska P, Rame C, Dobrzyń K, Smolińska N, Dupont J, Rak A. Omentin expression in the ovarian follicles of Large White and Meishan sows during the oestrous cycle and in vitro effect of gonadotropins and steroids on its level: Role of ERK1/2 and PI3K signaling pathways. PLoS One 2024; 19:e0297875. [PMID: 38408058 PMCID: PMC10896505 DOI: 10.1371/journal.pone.0297875] [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: 08/16/2023] [Accepted: 01/08/2024] [Indexed: 02/28/2024] Open
Abstract
Omentin (ITLN1) is a novel adipokine mainly expressed in the white adipose tissue. It plays a crucial role in the metabolic homeostasis and insulin sensitivity. Our last study documented that ITLN1 levels in the adipose tissue and plasma are lower in fat Meishan (MS) compared to normal weight Large White (LW) pigs. The aim of this study was to investigate transcript and protein concentrations of ITLN1 as well as its immunolocalisation in the ovarian follicles and examine the molecular mechanism involved in the regulation of its expression in response to gonadotropins (FSH, LH) and steroids (P4, T, E2). Ovarian follicles were collected from LW and MS sows on days 2-3, 10-12, and 14-16 of the oestrous. We found the elevated ITLN1 expression in the ovarian follicles and the increase of concentrations in follicular fluid (FF) of LW pigs vs MS pigs; in both breeds of pigs, the levels of ITLN1 increased with the oestrous progression. We noted ITLN1 signals in oocyte, granulosa and theca cells. Gonadotropins and steroids increased ITLN1 levels in the ovarian follicle cells of LW pigs, while in MS pigs, we observed only the stimulatory effect of LH and T. Both extracellular signal-regulated kinase (ERK1/2) and phosphatidylinositol 3'-kinase (PI3K) were involved in the regulation of ITLN1. Our study demonstrated the levels and regulation of ITLN1 in the porcine ovarian follicles through ERK1/2 and PI3K signaling pathways.
Collapse
Affiliation(s)
- Karolina Pich
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University in Krakow, Krakow, Poland
| | - Natalia Respekta
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University in Krakow, Krakow, Poland
| | - Patrycja Kurowska
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Christelle Rame
- INRAE, UMR85, Unité Physiologie de la Reproduction et des Comportements, Nouzilly, France
| | - Kamil Dobrzyń
- Department of Zoology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Olsztyn-Kortowo, Poland
| | - Nina Smolińska
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Olsztyn-Kortowo, Poland
| | - Joëlle Dupont
- INRAE, UMR85, Unité Physiologie de la Reproduction et des Comportements, Nouzilly, France
| | - Agnieszka Rak
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| |
Collapse
|
4
|
Khlebus E, Vuttaradhi VK, Welte T, Khurana N, Celestino J, Beird HC, Gumbs C, Little L, Legarreta AF, Fellman BM, Nguyen T, Lawson B, Ferri-Borgogno S, Mok SC, Broaddus RR, Gershenson DM, Futreal PA, Hillman RT. Comparative Tumor Microenvironment Analysis of Primary and Recurrent Ovarian Granulosa Cell Tumors. Mol Cancer Res 2023; 21:483-494. [PMID: 37068116 PMCID: PMC10150241 DOI: 10.1158/1541-7786.mcr-22-0623] [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: 08/09/2022] [Revised: 12/08/2022] [Accepted: 02/02/2023] [Indexed: 04/18/2023]
Abstract
Adult-type granulosa cell tumors (aGCT) are rare ovarian sex cord tumors with few effective treatments for recurrent disease. The objective of this study was to characterize the tumor microenvironment (TME) of primary and recurrent aGCTs and to identify correlates of disease recurrence. Total RNA sequencing (RNA-seq) was performed on 24 pathologically confirmed, cryopreserved aGCT samples, including 8 primary and 16 recurrent tumors. After read alignment and quality-control filtering, DESeq2 was used to identify differentially expressed genes (DEG) between primary and recurrent tumors. Functional enrichment pathway analysis and gene set enrichment analysis was performed using "clusterProfiler" and "GSVA" R packages. TME composition was investigated through the analysis and integration of multiple published RNA-seq deconvolution algorithms. TME analysis results were externally validated using data from independent previously published RNA-seq datasets. A total of 31 DEGs were identified between primary and recurrent aGCTs. These included genes with known function in hormone signaling such as LHCGR and INSL3 (more abundant in primary tumors) and CYP19A1 (more abundant in recurrent tumors). Gene set enrichment analysis revealed that primarily immune-related and hormone-regulated gene sets expression was increased in recurrent tumors. Integrative TME analysis demonstrated statistically significant depletion of cancer-associated fibroblasts in recurrent tumors. This finding was confirmed in multiple independent datasets. IMPLICATIONS Recurrent aGCTs exhibit alterations in hormone pathway gene expression as well as decreased infiltration of cancer-associated fibroblasts, suggesting dual roles for hormonal signaling and TME remodeling underpinning disease relapse.
Collapse
Affiliation(s)
- Eleonora Khlebus
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Veena K. Vuttaradhi
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Thomas Welte
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Namrata Khurana
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Joseph Celestino
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hannah C. Beird
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Curtis Gumbs
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Latasha Little
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alejandra Flores Legarreta
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Bryan M. Fellman
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Tri Nguyen
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Barrett Lawson
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sammy Ferri-Borgogno
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Samuel C. Mok
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Russell R. Broaddus
- Department of Pathology and Laboratory Medicine, The University of North Carolina, Chapel Hill, North Carolina
| | - David M. Gershenson
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - P. Andrew Futreal
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - R. Tyler Hillman
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
- CPRIT Scholar in Cancer Research, Houston, Texas
| |
Collapse
|
5
|
Andersson N, Haltia UM, Färkkilä A, Wong SC, Eloranta K, Wilson DB, Unkila-Kallio L, Pihlajoki M, Kyrönlahti A, Heikinheimo M. Analysis of Non-Relapsed and Relapsed Adult Type Granulosa Cell Tumors Suggests Stable Transcriptomes during Tumor Progression. Curr Issues Mol Biol 2022; 44:686-698. [PMID: 35723333 PMCID: PMC8928977 DOI: 10.3390/cimb44020048] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/17/2022] [Accepted: 01/24/2022] [Indexed: 11/16/2022] Open
Abstract
Adult-type granulosa cell tumor (AGCT) is a rare ovarian malignancy characterized by slow growth and hormonal activity. The prognosis of AGCT is generally favorable, but one-third of patients with low-stage disease experience a late relapse, and over half of them die of AGCT. To identify markers that would distinguish patients at risk for relapse, we performed Lexogen QuantSeq 3′ mRNA sequencing on formalin-fixed paraffin-embedded, archival AGCT tissue samples tested positive for the pathognomonic Forkhead Box L2 (FOXL2) mutation. We compared the transcriptomic profiles of 14 non-relapsed archival primary AGCTs (follow-up time 17–26 years after diagnosis) with 13 relapsed primary AGCTs (follow-up time 1.7–18 years) and eight relapsed tumors (follow-up time 2.8–18.9 years). Non-relapsed and relapsed primary AGCTs had similar transcriptomic profiles. In relapsed tumors three genes were differentially expressed: plasmalemma vesicle associated protein (PLVAP) was upregulated (p = 0.01), whereas argininosuccinate synthase 1 (ASS1) (p = 0.01) and perilipin 4 (PLIN4) (p = 0.02) were downregulated. PLVAP upregulation was validated using tissue microarray RNA in situ hybridization. In our patient cohort with extremely long follow-up, we observed similar gene expression patterns in both primary AGCT groups, suggesting that relapse is not driven by transcriptomic changes. These results reinforce earlier findings that molecular markers do not predict AGCT behavior or risk of relapse.
Collapse
Affiliation(s)
- Noora Andersson
- HUSLAB, Helsinki University Hospital, Haartmaninkatu 4, 00290 Helsinki, Finland;
- Pediatric Research Center, Children’s Hospital, University of Helsinki and Helsinki University Hospital, Tukholmankatu 8, 00290 Helsinki, Finland; (K.E.); (A.K.); (M.H.)
| | - Ulla-Maija Haltia
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Haartmaninkatu 2, 00290 Helsinki, Finland; (U.-M.H.); (A.F.); (L.U.-K.)
| | - Anniina Färkkilä
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Haartmaninkatu 2, 00290 Helsinki, Finland; (U.-M.H.); (A.F.); (L.U.-K.)
- Research Program for Systems Oncology, University of Helsinki and Helsinki University Hospital, Haartmaninkatu 8, 00290 Helsinki, Finland
| | | | - Katja Eloranta
- Pediatric Research Center, Children’s Hospital, University of Helsinki and Helsinki University Hospital, Tukholmankatu 8, 00290 Helsinki, Finland; (K.E.); (A.K.); (M.H.)
| | - David B. Wilson
- Department of Pediatrics, Washington University in St. Louis, 660 S Euclid Ave, St. Louis, MO 63110, USA;
- Department of Developmental Biology, Washington University School of Medicine, 660 S. Euclid Avenue Campus Box 8103, St. Louis, MO 63110, USA
| | - Leila Unkila-Kallio
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Haartmaninkatu 2, 00290 Helsinki, Finland; (U.-M.H.); (A.F.); (L.U.-K.)
| | - Marjut Pihlajoki
- Pediatric Research Center, Children’s Hospital, University of Helsinki and Helsinki University Hospital, Tukholmankatu 8, 00290 Helsinki, Finland; (K.E.); (A.K.); (M.H.)
- Correspondence:
| | - Antti Kyrönlahti
- Pediatric Research Center, Children’s Hospital, University of Helsinki and Helsinki University Hospital, Tukholmankatu 8, 00290 Helsinki, Finland; (K.E.); (A.K.); (M.H.)
| | - Markku Heikinheimo
- Pediatric Research Center, Children’s Hospital, University of Helsinki and Helsinki University Hospital, Tukholmankatu 8, 00290 Helsinki, Finland; (K.E.); (A.K.); (M.H.)
- Department of Pediatrics, Washington University in St. Louis, 660 S Euclid Ave, St. Louis, MO 63110, USA;
| |
Collapse
|
6
|
Chauvin S, Cohen-Tannoudji J, Guigon CJ. Estradiol Signaling at the Heart of Folliculogenesis: Its Potential Deregulation in Human Ovarian Pathologies. Int J Mol Sci 2022; 23:ijms23010512. [PMID: 35008938 PMCID: PMC8745567 DOI: 10.3390/ijms23010512] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/23/2021] [Accepted: 12/24/2021] [Indexed: 01/26/2023] Open
Abstract
Estradiol (E2) is a major hormone controlling women fertility, in particular folliculogenesis. This steroid, which is locally produced by granulosa cells (GC) within ovarian follicles, controls the development and selection of dominant preovulatory follicles. E2 effects rely on a complex set of nuclear and extra-nuclear signal transduction pathways principally triggered by its nuclear receptors, ERα and ERβ. These transcription factors are differentially expressed within follicles, with ERβ being the predominant ER in GC. Several ERβ splice isoforms have been identified and display specific structural features, which greatly complicates the nature of ERβ-mediated E2 signaling. This review aims at providing a concise overview of the main actions of E2 during follicular growth, maturation, and selection in human. It also describes the current understanding of the various roles of ERβ splice isoforms, especially their influence on cell fate. We finally discuss how E2 signaling deregulation could participate in two ovarian pathogeneses characterized by either a follicular arrest, as in polycystic ovary syndrome, or an excess of GC survival and proliferation, leading to granulosa cell tumors. This review emphasizes the need for further research to better understand the molecular basis of E2 signaling throughout folliculogenesis and to improve the efficiency of ovarian-related disease therapies.
Collapse
|
7
|
Cluzet V, Devillers MM, Petit F, Pierre A, Giton F, Airaud E, L'Hôte D, Leary A, Genestie C, Treilleux I, Mayeur A, Katzenellenbogen JA, Kim SH, Cohen-Tannoudji J, Chauvin S, Guigon CJ. Estradiol promotes cell survival and induces Greb1 expression in granulosa cell tumors of the ovary through an ERα-dependent mechanism. J Pathol 2021; 256:335-348. [PMID: 34860414 DOI: 10.1002/path.5843] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 11/10/2021] [Accepted: 11/30/2021] [Indexed: 12/12/2022]
Abstract
Granulosa cell tumor (GCT) is a form of ovarian tumor characterized by its tendency to recur years after surgical ablation. Little is known on the mechanisms involved in GCT development and progression. GCTs can produce estradiol (E2), but whether this hormone could play a role in this cancer through its nuclear receptors, i.e., ERα and ERβ, remains unknown. Here, we addressed this issue by cell-based and molecular studies on human GCTs and GCT cell lines. Importantly, we observed that E2 significantly increased the growth of GCT cells by promoting cell survival. The use of selective agonists of each type of receptor, together with Esr1 (ERα) or Esr2 (ERβ)-deleted GCT cells revealed that E2 mediated its effects through ERα-dependent genomic mechanisms and ERβ/ERα-dependent extra-nuclear mechanisms. Notably, the expression of Greb1, a prototypical ER target gene, was dose-dependently up-regulated by E2 specifically through ERα in GCT cells. Accordingly, using GCTs from patients, we found that GREB1 mRNA abundance was positively correlated to intra-tumoral E2 concentrations. Tissue microarrays analyses showed that there were various combinations of ER expression in primary and recurrent GCTs, and that ERα expression persisted only in combination with ERβ in ~40% of recurrent tumors. Altogether, this study demonstrates that E2 can promote the progression of GCTs, with a clear dependence on ERα. In addition to demonstrating that GCTs can be classified as a hormone-related cancer, our results also highlight that the nature of ER forms present in recurrent GCTs could underlie the variable efficiency of endocrine therapies. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Victoria Cluzet
- Université de Paris, BFA, UMR 8251, CNRS, ERL U1133, Inserm, F-75013, Paris, France
| | - Marie M Devillers
- Université de Paris, BFA, UMR 8251, CNRS, ERL U1133, Inserm, F-75013, Paris, France
| | - Florence Petit
- Université de Paris, BFA, UMR 8251, CNRS, ERL U1133, Inserm, F-75013, Paris, France
| | - Alice Pierre
- Université de Paris, BFA, UMR 8251, CNRS, ERL U1133, Inserm, F-75013, Paris, France
| | - Frank Giton
- AP-HP, Pôle biologie-Pathologie Henri Mondor, INSERM IMRB U955, Créteil, France
| | - Eloïse Airaud
- Université de Paris, BFA, UMR 8251, CNRS, ERL U1133, Inserm, F-75013, Paris, France
| | - David L'Hôte
- Université de Paris, BFA, UMR 8251, CNRS, ERL U1133, Inserm, F-75013, Paris, France
| | - Alexandra Leary
- Gustave Roussy Cancer Campus and University of Paris-Saclay, Villejuif, France
| | - Catherine Genestie
- Department of Pathology, University Paris-Saclay, Gustave Roussy Cancer Center, Villejuif, France
| | | | - Anne Mayeur
- Service de Médecine de la Reproduction et Préservation de la Fertilité, Hôpital Antoine Béclère, Clamart, France
| | - John A Katzenellenbogen
- Department of Chemistry and Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| | - Sung Hoon Kim
- Department of Chemistry and Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| | | | - Stéphanie Chauvin
- Université de Paris, BFA, UMR 8251, CNRS, ERL U1133, Inserm, F-75013, Paris, France
| | - Céline J Guigon
- Université de Paris, BFA, UMR 8251, CNRS, ERL U1133, Inserm, F-75013, Paris, France
| |
Collapse
|
8
|
Gambioli R, Montanino Oliva M, Nordio M, Chiefari A, Puliani G, Unfer V. New Insights into the Activities of D-Chiro-Inositol: A Narrative Review. Biomedicines 2021; 9:biomedicines9101378. [PMID: 34680494 PMCID: PMC8533370 DOI: 10.3390/biomedicines9101378] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 09/29/2021] [Accepted: 09/30/2021] [Indexed: 12/14/2022] Open
Abstract
D-chiro-inositol (DCI) is a natural compound detectable in cell membranes, which is highly conserved as a biological signaling molecule. In mammals, its function is primarily characterized in the intracellular transduction cascade of insulin. In particular, insulin signal promotes the release of pivotal DCI-containing molecules. In fact, impaired release of DCI is a common feature of insulin-resistant tissues, and insulin-sensitizing pharmaceuticals induce higher concentrations of free DCI. Moreover, it also plays important roles in several other processes. DCI is involved in the regulation of steroidogenesis, due to its regulatory effects on steroidogenic enzymes, including 17α-hydroxylase, 3β-hydroxysteroid dehydrogenase, and aromatase. Such regulation of various enzymes indicates a mechanism by which the body regulates different processes via a single molecule, depending on its concentration. DCI also reduces the expression of integrin β3, which is an adhesion molecule involved in embryo implantation and cellular phenomena such as survival, stemness, and invasiveness. In addition, DCI seems to have important anti-inflammatory activities, like its 3-O-methyl-ether, called pinitol. In vitro evidence demonstrates that treatment with both compounds induces a reduction in pro-inflammatory factors—such as Nf-κB—and cytokines—such as TNF-α. DCI then plays important roles in several fundamental processes in physiology. Therefore, research on such molecule is of primary importance.
Collapse
Affiliation(s)
| | - Mario Montanino Oliva
- The Experts Group on Inositol in Basic and Clinical Research (EGOI), 00161 Rome, Italy; (M.M.O.); (M.N.)
- Department of Obstetrics and Gynecology, Santo Spirito Hospital, 00193 Rome, Italy
| | - Maurizio Nordio
- The Experts Group on Inositol in Basic and Clinical Research (EGOI), 00161 Rome, Italy; (M.M.O.); (M.N.)
- Department of Experimental Medicine, Sapienza University, 00185 Rome, Italy
| | - Alfonsina Chiefari
- Oncological Endocrinology Unit, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (A.C.); (G.P.)
| | - Giulia Puliani
- Oncological Endocrinology Unit, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (A.C.); (G.P.)
| | - Vittorio Unfer
- The Experts Group on Inositol in Basic and Clinical Research (EGOI), 00161 Rome, Italy; (M.M.O.); (M.N.)
- System Biology Group Lab, 00161 Rome, Italy
- Correspondence:
| |
Collapse
|
9
|
Banerjee SN, Tang M, O'Connell RL, Sjoquist K, Clamp AR, Millan D, Nottley S, Lord R, Mullassery VM, Hall M, Gourley C, Bonaventura T, Goh JC, Sykes P, Grant PT, McNally O, Alexander L, Kelly C, Carty K, Divers L, Bradshaw N, Edmondson RJ, Friedlander M. A phase 2 study of anastrozole in patients with oestrogen receptor and/progesterone receptor positive recurrent/metastatic granulosa cell tumours/sex-cord stromal tumours of the ovary: The PARAGON/ANZGOG 0903 trial. Gynecol Oncol 2021; 163:72-78. [PMID: 34412908 DOI: 10.1016/j.ygyno.2021.07.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/11/2021] [Accepted: 07/13/2021] [Indexed: 12/28/2022]
Abstract
BACKGROUND Hormonal therapies are commonly prescribed to patients with metastatic granulosa cell tumours (GCT), based on high response rates in small retrospective studies. Aromatase inhibitors (AIs) are reported to have high response rates and an accepted treatment option. We report the results of a phase 2 trial of an AI in recurrent/metastatic GCTs. METHODS 41 patients with recurrent ER/PR + ve GCT received anastrozole 1 mg daily until progression or unacceptable toxicity. The primary endpoint was clinical benefit rate (CBR) at 12 weeks, evaluated by RECIST1.1 criteria. Secondary endpoints included progression-free survival (PFS), CBR duration, quality of life and toxicity. RESULTS The CBR at 12 weeks in 38 evaluable patients was 78.9%, which included one (2.6%; 95% CI: 0.5-13.5%) partial response and 76.3% stable disease. Two additional patients without measurable disease were stable, based on inhibin. Median PFS was 8.6 m (95% CI 5.5-13.5 m). There were delayed responses observed after 12 weeks with a total of 4 pts. (10.5%; 95% CI 4.2%-24.1%) with a RECIST partial response; 23 (59%) patients were progression-free at 6 months. The adverse effects were predominantly low grade. CONCLUSIONS This is the first prospective trial of hormonal therapy in GCTs. Although there was a high CBR, the objective response rate to anastrozole was much lower than the pooled response rates of >70% to AIs reported in most retrospective series and case reports. PARAGON demonstrates the importance of prospective trials in rare cancers and the need to reconsider the role of AIs as single agents in GCTs.
Collapse
Affiliation(s)
- Susana N Banerjee
- The Royal Marsden NHS Foundation Trust and Institute of Cancer Research, London, United Kingdom.
| | - Monica Tang
- NHMRC Clinical Trials Centre, University of Sydney, NSW 2050, Australia
| | | | - Katrin Sjoquist
- NHMRC Clinical Trials Centre, University of Sydney, NSW 2050, Australia
| | - Andrew R Clamp
- The Christie NHS Foundation Trust and University of Manchester, Manchester, United Kingdom
| | - David Millan
- Queen Elizabeth University Hospital, Glasgow, Scotland, United Kingdom
| | - Steven Nottley
- Queen Elizabeth University Hospital, Glasgow, Scotland, United Kingdom
| | - Rosemary Lord
- The National Cancer Research Institute and the Clatterbridge Cancer Centre NHS Foundation Trust, Wirral, United Kingdom
| | | | - Marcia Hall
- Mount Vernon Cancer Centre, Middlesex, United Kingdom
| | - Charlie Gourley
- Cancer Research UK Edinburgh Centre, MRC IGMM, University of Edinburgh, Edinburgh, United Kingdom
| | | | - Jeffrey C Goh
- Royal Brisbane and Women's Hospital, Brisbane, QLD 4029, Australia
| | - Peter Sykes
- Christchurch Women's Hospital, Christchurch, New Zealand
| | - Peter T Grant
- Mercy Hospital for Women, Melbourne, VIC 3084, Australia
| | - Orla McNally
- Royal Women's Hospital, Melbourne, VIC 3052, Australia
| | - Laura Alexander
- Cancer Research UK Clinical Trials Unit, Institute of Cancer Sciences, University of Glasgow, United Kingdom
| | - Caroline Kelly
- Cancer Research UK Clinical Trials Unit, Institute of Cancer Sciences, University of Glasgow, United Kingdom
| | - Karen Carty
- Cancer Research UK Clinical Trials Unit, Institute of Cancer Sciences, University of Glasgow, United Kingdom
| | - Laura Divers
- Cancer Research UK Clinical Trials Unit, Institute of Cancer Sciences, University of Glasgow, United Kingdom
| | - N Bradshaw
- NHMRC Clinical Trials Centre, University of Sydney, NSW 2050, Australia
| | - Richard J Edmondson
- Manchester Academic Health Science Centre, St Mary's Hospital, Manchester, United Kingdom
| | - Michael Friedlander
- Prince of Wales Clinical School UNSW and Royal Hospital for Women, Sydney, NSW 2031, Australia
| | | |
Collapse
|
10
|
Roze JF, van Meurs HS, Monroe GR, Veldhuis WB, van Lonkhuijzen LRCW, Bennink RJ, Groeneweg JW, Witteveen PO, Jonges GN, Zweemer RP, Braat AJAT. [ 18F]FDG and [ 18F]FES positron emission tomography for disease monitoring and assessment of anti-hormonal treatment eligibility in granulosa cell tumors of the ovary. Oncotarget 2021; 12:665-673. [PMID: 33868587 PMCID: PMC8021033 DOI: 10.18632/oncotarget.27925] [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: 01/30/2021] [Accepted: 03/15/2021] [Indexed: 01/08/2023] Open
Abstract
Purpose: Adult granulosa cell tumors (AGCTs) of the ovary represent a rare malignancy in which timing and choice of treatment is a clinical challenge. This study investigates the value of FDG-PET/CT and FES-PET/CT in monitoring recurrent AGCTs and assessing eligibility for anti-hormonal treatment. Materials and Methods: We evaluated 22 PET/CTs from recurrent AGCT patients to determine tumor FDG (n = 16) and FES (n = 6) uptake by qualitative and quantitative analysis. We included all consecutive patients from two tertiary hospitals between 2003-2020. Expression of ERα and ERβ and mitoses per 2 mm2 were determined by immunohistochemistry and compared to FES and FDG uptake, respectively. Results: Qualitative assessment showed low-to-moderate FDG uptake in most patients (14/16), and intense uptake in 2/16. One patient with intense tumor FDG uptake had a high mitotic rate (18 per 2 mm2) Two out of six patients showed FES uptake on PET/CT at qualitative analysis. Lesion-based quantitative assessment showed a mean SUVmax of 2.4 (± 0.9) on FDG-PET/CT and mean SUVmax of 1.7 (± 0.5) on FES-PET/CT. Within patients, expression of ERα and ERβ varied and did not seem to correspond with FES uptake. In one FES positive patient, tumor locations with FES uptake remained stable or decreased in size during anti-hormonal treatment, while all FES negative locations progressed. Conclusions: This study shows that in AGCTs, FDG uptake is limited and therefore FDG-PET/CT is not advised. FES-PET/CT may be useful to non-invasively capture the estrogen receptor expression of separate tumor lesions and thus assess the potential eligibility for hormone treatment in AGCT patients.
Collapse
Affiliation(s)
- Joline F Roze
- Department of Gynecological Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Hannah S van Meurs
- Department of Gynecological Oncology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Glen R Monroe
- Department of Gynecological Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Wouter B Veldhuis
- Department of Radiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Luc R C W van Lonkhuijzen
- Department of Gynecological Oncology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Roel J Bennink
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Jolijn W Groeneweg
- Department of Gynecological Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Petronella O Witteveen
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Geertruida N Jonges
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Ronald P Zweemer
- Department of Gynecological Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Arthur J A T Braat
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
11
|
Liu T, Huang Y, Lin H. Estrogen disorders: Interpreting the abnormal regulation of aromatase in granulosa cells (Review). Int J Mol Med 2021; 47:73. [PMID: 33693952 PMCID: PMC7952251 DOI: 10.3892/ijmm.2021.4906] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 01/27/2021] [Indexed: 12/11/2022] Open
Abstract
Ovarian granulosa cells (GCs) are the most important source of estrogen. Therefore, aromatase (estrogen synthase), which is the key enzyme in estrogen synthesis, is not only an important factor of ovarian development, but also the key to estrogen secretion by GCs. Disorders of the ovarian estrogen secretion are more likely to induce female estrogen-dependent diseases and fertility issues, such as ovarian cancer and polycystic ovary syndrome. Hence, aromatase is an important drug target; treatment with its inhibitors in estrogen-dependent diseases has attracted increasing attention. The present review article focuses on the regulation and mechanism of the aromatase activity in the GCs, as well as the specific regulation of aromatase promoters. In GCs, follicle-stimulating hormone (FSH) is dependent on the cyclic adenosine monophosphate (cAMP) pathway to regulate the aromatase activity, and the regulation of this enzyme is related to the activation of signaling pathways, such as phosphatidylinositol 3-kinase (PI3K) and extracellular signal-regulated kinase (ERK). In addition, endocrine-disrupting substance and other related factors affect the expression of aromatase, which eventually create an imbalance in the estrogen secretion by the target tissues. The present review highlights these useful factors as potential inhibitors for target therapy.
Collapse
Affiliation(s)
- Ting Liu
- Department of Pathophysiology, School of Basic Medicine Sciences, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yifei Huang
- First Clinical Medical School, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Hui Lin
- Department of Pathophysiology, School of Basic Medicine Sciences, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
12
|
Secchi C, Benaglio P, Mulas F, Belli M, Stupack D, Shimasaki S. FOXO1 mitigates the SMAD3/FOXL2 C134W transcriptomic effect in a model of human adult granulosa cell tumor. J Transl Med 2021; 19:90. [PMID: 33639972 PMCID: PMC7913442 DOI: 10.1186/s12967-021-02754-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 02/16/2021] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Adult granulosa cell tumor (aGCT) is a rare type of stromal cell malignant cancer of the ovary characterized by elevated estrogen levels. aGCTs ubiquitously harbor a somatic mutation in FOXL2 gene, Cys134Trp (c.402C < G); however, the general molecular effect of this mutation and its putative pathogenic role in aGCT tumorigenesis is not completely understood. We previously studied the role of FOXL2C134W, its partner SMAD3 and its antagonist FOXO1 in cellular models of aGCT. METHODS In this work, seeking more comprehensive profiling of FOXL2C134W transcriptomic effects, we performed an RNA-seq analysis comparing the effect of FOXL2WT/SMAD3 and FOXL2C134W/SMAD3 overexpression in an established human GC line (HGrC1), which is not luteinized, and bears normal alleles of FOXL2. RESULTS Our data shows that FOXL2C134W/SMAD3 overexpression alters the expression of 717 genes. These genes include known and novel FOXL2 targets (TGFB2, SMARCA4, HSPG2, MKI67, NFKBIA) and are enriched for neoplastic pathways (Proteoglycans in Cancer, Chromatin remodeling, Apoptosis, Tissue Morphogenesis, Tyrosine Kinase Receptors). We additionally expressed the FOXL2 antagonistic Forkhead protein, FOXO1. Surprisingly, overexpression of FOXO1 mitigated 40% of the altered genome-wide effects specifically related to FOXL2C134W, suggesting it can be a new target for aGCT treatment. CONCLUSIONS Our transcriptomic data provide novel insights into potential genes (FOXO1 regulated) that could be used as biomarkers of efficacy in aGCT patients.
Collapse
Affiliation(s)
- Christian Secchi
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Diego School of Medicine, 9500 Gilman Drive, La Jolla, CA, 92093, USA.
| | - Paola Benaglio
- Department of Pediatrics, University of California San Diego, School of Medicine, La Jolla, CA, USA
| | - Francesca Mulas
- Department of Pediatrics, University of California San Diego, School of Medicine, La Jolla, CA, USA
| | - Martina Belli
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Diego School of Medicine, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Dwayne Stupack
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Diego School of Medicine, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Shunichi Shimasaki
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Diego School of Medicine, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| |
Collapse
|