1
|
Gülseren V, Dolanbay M, Çağli F, Dağgez M, Topaloğlu N, Öztürk F, Özçelik B, Serin İS, Güngördük K. The Role of hCG Expression in Endometrial Cancer Prognosis. Int J Gynecol Pathol 2025:00004347-990000000-00225. [PMID: 39869090 DOI: 10.1097/pgp.0000000000001102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
This study aims to investigate the expression pattern of human chorionic gonadotropin (hCG) in the tissue of endometrioid type endometrial cancer (EEC) using immunohistochemistry, and also to investigate the effect of hCG expression pattern on prognosis and survival in EEC. We evaluated patients who were operated between 2010 and 2020 in the obstetrics and gynecology clinic of our center due to EEC. In total, 194 women were determined to be in either the hCG-negative group (n=137) or the hCG-positive group (n=57). The detection rate of deep myometrial invasion (16.8% vs. 36.8%; P=0.002), lymphovascular space invasion (10.9% vs. 24.6%; P=0.015), and metastatic lymph node (6.7% vs. 21.8%; P= 0.003) in patients with hCG-positive staining were analyzed to be significantly higher. Five-year disease-free survival (DFS) (P= 0.015) and overall survival (OS) (P= 0.024) rates were found to be higher in the hCG-negative group. hCG expression was found to be an independent risk factor for recurrence, and DFS in grade I-II EEC was limited to the uterus and with superficial myometrial invasion (low risk). No independent risk factors for OS were analyzed. hCG positivity is a risk factor with poor prognostic factors in endometrial cancer. It was concluded that hCG expression in low-risk EEC is a valuable negative prognostic factor for recurrence and DFS.
Collapse
Affiliation(s)
- Varol Gülseren
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology
| | | | | | - Mine Dağgez
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology
| | - Nahit Topaloğlu
- Medical Pathology, Faculty of Medicine, Erciyes University, Kayseri
| | - Figen Öztürk
- Medical Pathology, Faculty of Medicine, Erciyes University, Kayseri
| | - Bülent Özçelik
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology
| | | | - Kemal Güngördük
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Faculty of Medicine, Muğla Sitki Koçman University, Muğla, Turkey
| |
Collapse
|
2
|
Pant A, Moar K, Maurya PK. Impact of estradiol in inducing endometrial cancer using RL95-2. Pathol Res Pract 2024; 263:155640. [PMID: 39383736 DOI: 10.1016/j.prp.2024.155640] [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: 08/11/2024] [Revised: 09/27/2024] [Accepted: 10/02/2024] [Indexed: 10/11/2024]
Abstract
BACKGROUND Endometrial cancer is the most common gynecological malignancy that originates from the inner lining of the uterus and predominantly affects postmenopausal women. Prolonged exposure to estrogen, family history of endometrial cancer, obesity, and hormonal imbalance are some of the risk factors associated with endometrial cancer. In our study, we investigated the effect of estradiol, a potent form of estrogen at various concentrations on endometrial cell line RL95-2. METHODS Endometrial cell RL95-2 were cultured in DMEM medium with optimal conditions required to maintain the cells. MTT assay and colony formation assay were further performed after treating the cells with different concentrations of estradiol (1, 10, and 100 nM) and TAM (100 nM). Moreover, the effect of genes regulated by estradiol was also examined using microarray and validated using real-time polymerase chain reaction (qRT-PCR). RESULTS Time-dependent MTT assay shows a significant change in the ability of the cells to survive relative to concentrations. Colony formation was found to be directly proportional to the concentration of the estradiol (p < 0.05). Among genes, MMP14 (p = 0.03), SPARCL1 (p = 0.005), and CLU (p = 0.06) showed a significant up-regulation in their expression after estradiol treatment while NRN1 (p < 0.001) showed significant downregulation in expression pattern compared to control. However, the TAM treatment was found to be significantly effective after 72 h (p < 0.001) compared to control and 100 nM E2 (p = 0.0206). CONCLUSION Our study suggests that estradiol significantly contributes to regulating the viability, colony formation, and expression of genes associated with endometrial cancer.
Collapse
Affiliation(s)
- Anuja Pant
- Department of Biochemistry, Central University of Haryana, Mahendergarh 123031, India
| | - Kareena Moar
- Department of Biochemistry, Central University of Haryana, Mahendergarh 123031, India
| | - Pawan Kumar Maurya
- Department of Biochemistry, Central University of Haryana, Mahendergarh 123031, India.
| |
Collapse
|
3
|
Cavalcante L, Deshmukh SK, Ribeiro JR, Carneiro BA, Dizon DS, Angara K, Mattox T, Wu S, Xiu J, Walker P, Oberley M, Nabhan C, Huang H, Antonarakis ES. Opposing Roles of SPOP Mutations in Human Prostate and Endometrial Cancers. JCO Precis Oncol 2023; 7:e2300088. [PMID: 37677121 DOI: 10.1200/po.23.00088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 05/09/2023] [Accepted: 07/12/2023] [Indexed: 09/09/2023] Open
Abstract
PURPOSE Recurrent gene mutations in speckle-type POZ protein (SPOP), the substrate-binding component of E3 ubiquitin ligase, are associated with tumor progression in prostate and endometrial cancers. Here, we characterized SPOP mutations in these cancers and explored their association with molecular and immune signatures and patient outcomes. METHODS There were 7,398 prostate cancer and 19,188 endometrial cancer samples analyzed for clinical and molecular profiles at Caris Life Sciences. Overall survival (OS) was analyzed using Kaplan-Meier survival curves. Statistical significance was determined using chi-square and Mann-Whitney U tests, with P values adjusted for multiple comparisons. RESULTS SPOP mutations were identified in 9.2% of prostate and 4.3% of endometrial cancers. Mutations clustered in the SPOP meprin and TRAF-C homology domain, with no significant overlap between cancer types. SPOP mutation was associated with differential comutation profiles and opposing tumor immune microenvironment signatures for each cancer, with greater immune infiltration in SPOP-mutated endometrial cancer. SPOP-mutated prostate and endometrial cancers displayed altered epigenetic gene expression, including opposite regulation of BRD2 transcripts. In SPOP-mutant prostate cancer, higher expression of androgen receptor-regulated transcripts and improved OS after treatment with hormonal agents were observed. In endometrial cancer, hormone receptor expression was significantly lower in SPOP-mutated tumors and differences in OS were highly dependent on the particular hotspot mutation and histologic subtype. CONCLUSION These data indicate that SPOP mutations drive opposing molecular and immune landscapes in prostate and endometrial cancers-suggesting a loss-of-function mechanism in prostate cancer and gain-of-function mechanism in endometrial cancer-and provide a rationale for tailored therapeutic approaches.
Collapse
Affiliation(s)
| | | | | | - Benedito A Carneiro
- Lifespan Cancer Institute, Legorreta Cancer Center at Brown University, Providence, RI
| | - Don S Dizon
- Lifespan Cancer Institute, Legorreta Cancer Center at Brown University, Providence, RI
| | | | | | | | | | | | | | | | - Haojie Huang
- Mayo Clinic College of Medicine and Science, Rochester, MN
| | | |
Collapse
|
4
|
DSCAM-AS1 Long Non-Coding RNA Exerts Oncogenic Functions in Endometrial Adenocarcinoma via Activation of a Tumor-Promoting Transcriptome Profile. Biomedicines 2022; 10:biomedicines10071727. [PMID: 35885035 PMCID: PMC9313190 DOI: 10.3390/biomedicines10071727] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/12/2022] [Accepted: 07/13/2022] [Indexed: 11/16/2022] Open
Abstract
Accumulating evidence suggests that lncRNA DSCAM-AS1 acts tumor-promoting in various cancer entities. In breast cancer, DSCAM-AS1 was shown to be the lncRNA being most responsive to induction by estrogen receptor α (ERα). In this study, we examined the function of DSCAM-AS1 in endometrial adenocarcinoma using in silico and different in vitro approaches. Initial analysis of open-source data revealed DSCAM-AS1 overexpression in endometrial cancer (EC) (p < 0.01) and a significant association with shorter overall survival of EC patients (HR = 1.78, p < 0.01). In EC, DSCAM-AS1 was associated with endometrial tumor promotor gene PRL and with expression of ERα and its target genes TFF1 and PGR. Silencing of this lncRNA by RNAi in two EC cell lines was more efficient in ERα-negative HEC-1B cells and reduced their growth and the expression of proliferation activators like NOTCH1, PTK2 and EGR1. DSCAM-AS1 knockdown triggered an anti-tumoral transcriptome response as revealed by Affymetrix microarray analysis, emerging from down-regulation of tumor-promoting genes and induction of tumor-suppressive networks. Finally, several genes regulated upon DSCAM-AS1 silencing in vitro were found to be inversely correlated with this lncRNA in EC tissues. This study clearly suggests an oncogenic function of DSCAM-AS1 in endometrial adenocarcinoma via activation of a tumor-promoting transcriptome profile.
Collapse
|
5
|
Cheng Y, Zhang S, Qiang Y, Dong L, Li Y. Integrated bioinformatics data analysis reveals a risk signature and PKD1 induced progression in endometrial cancer patients with postmenopausal status. Aging (Albany NY) 2022; 14:5554-5570. [PMID: 35816294 PMCID: PMC9320543 DOI: 10.18632/aging.204168] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 06/23/2022] [Indexed: 11/25/2022]
Abstract
Background: Endometrial cancer (EC) is one of the most common type of female genital malignancies. The purpose of the present study was to reveal the underlying oncogene and mechanism that played a pivotal role in postmenopausal EC patients. Methods: Weighted gene co-expression network analysis (WGCNA) was conducted using the microarray dataset and clinical data of EC patients from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases to identify significant gene modules and hub genes associated with postmenopausal status in EC patients. LASSO regression was conducted to build and validate the risk model. Finally, expression of hub gene was validated in pre- and post-menopausal EC patients in our center. Results: 1240 common genes were used to construct the WGCNA model. According to the WGCNA results, we identified a brown module with 471 genes which was significantly associated with postmenopausal status in EC patients. Furthermore, we constructed an 11-gene risk signature to predict the overall survival of EC patients. The Kaplan–Meier curve and area under the ROC curve (AUC) of this model showed high accuracy in prediction. We also validate the risk model in patients in our center and it also has a high accuracy. Among the 11 genes, PKD1 was recognized as a potential biomarker in the progression of EC patients with postmenopausal status. Conclusion: Taken together, we uncovered a common PKD1-mediated mechanism underlying postmenopausal EC patients’ progression by integrated analyses. This finding may improve targeted therapy for EC patients.
Collapse
Affiliation(s)
- Yun Cheng
- Department of Gynecology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210000, Jiangsu Province, China
| | - Suyun Zhang
- Department of Gynecology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210000, Jiangsu Province, China
| | - Yan Qiang
- Department of Gynecology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210000, Jiangsu Province, China
| | - Lingyan Dong
- Department of Gynecology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210000, Jiangsu Province, China
| | - Yujuan Li
- Department of Gynecology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210000, Jiangsu Province, China
| |
Collapse
|
6
|
Török P, Molnár S, Lampé R, Jakab A. The use of hysteroscopy in endometrial cancer: old questions and novel challenges. Climacteric 2021; 23:330-335. [PMID: 32648827 DOI: 10.1080/13697137.2020.1732914] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Endometrial cancer is the most common gynecological malignancy with a relatively good overall prognosis. It traditionally has two subtypes: type 1 (endometrioid carcinoma) and type 2 (non-endometrioid carcinoma). The prognosis is excellent for stage I endometrioid cancer, with a 5-year survival rate of 96%. However, the prognosis is much worse for women with high-risk endometrial cancer. Effective preoperative staging is important in order to tailor treatment and achieve optimal long-term survival. The majority of asymptomatic polyps detected by ultrasound are treated surgically. Conventionally, dilatation and curettage was performed to obtain a histological diagnosis, but nowadays hysteroscopy with biopsy is starting to be considered as the gold standard. Hysteroscopic resection seems to reduce the risk of underdiagnosed (atypical endometrial hyperplasia) endometrial cancer. To avoid the spread of malignant cells, hysteroscopy should be performed with concern to keep intrauterine pressure low. In comparison with cervical injection, the hysteroscopic method has a better detection rate in the para-aortic area during sentinel lymph node mapping. In the assessment of cervical involvement, the accuracy of magnetic resonance imaging is significantly higher than the accuracy of hysteroscopy. In fertility-sparing cases, hysteroscopic endometrium resection with progesterone therapy is an acceptable option.
Collapse
Affiliation(s)
- P Török
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - S Molnár
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - R Lampé
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - A Jakab
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
7
|
Carugno J, Marbin SJ, LaganÀ AS, Vitale SG, Alonso L, DI Spiezio Sardo A, Haimovich S. New development on hysteroscopy for endometrial cancer diagnosis: state of the art. Minerva Med 2021; 112:12-19. [PMID: 33438376 DOI: 10.23736/s0026-4806.20.07123-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Endometrial cancer (EC) is the most common gynecologic cancer diagnosed in developed countries and represents the second most frequent gynecologic cancer-related cause of death following ovarian cancer. There are 2 subtypes of EC. Type I tumors (endometrioid adenocarcinoma) representing 85-90% of the cases. They are likely to be low-grade tumors and are thought to have a link to estrogen exposure. Type II tumors represent 10-15% of EC. They are characterized as high-grade carcinomas, with serous or clear cell histology type, and carry poor prognoses. The benefits of hysteroscopy in achieving a targeted endometrial biopsy under direct visualization over blind biopsy techniques are widely accepted. Hysteroscopic endometrial biopsy is performed under direct visualization and is the only technique that allows for the selective biopsy of targeted areas of the endometrium. There is no screening protocol for the early detection of EC. Among the general population, advanced age, obesity, nulliparity and the use of exogenous hormones are known as risk factors for EC. There are additional situations that portend an increased risk of EC that deserve special consideration such as in patients diagnosed with Lynch Syndrome, using tamoxifen, obese, or the young patient with a desire for future fertility. We presented a narrative review of the current role of hysteroscopy for the diagnosis of endometrial cancer.
Collapse
Affiliation(s)
- Jose Carugno
- Miller School of Medicine, Unit of Minimally Invasive Gynecology, Division of Obstetrics, Department of Gynecology and Reproductive Sciences, University of Miami, Miami, FL, USA -
| | - Staci J Marbin
- Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Antonio S LaganÀ
- Department of Obstetrics and Gynecology, Filippo del Ponte Hospital, University of Insubria, Varese, Italy
| | - Salvatore G Vitale
- Unit of Obstetrics and Gynecology, Department of General Surgery and Medical Surgical Specialties, University of Catania, Catania, Italy
| | - Luis Alonso
- Unit of Gynecology Endoscopy, Gutenberg Center, Malaga, Spain
| | | | - Sergio Haimovich
- Rappaport Faculty of Medicine, Hillel Yaffe Medical Center, Technion, Israel
| |
Collapse
|
8
|
Treeck O, Skrzypczak M, Schüler-Toprak S, Weber F, Ortmann O. Long non-coding RNA CCAT1 is overexpressed in endometrial cancer and regulates growth and transcriptome of endometrial adenocarcinoma cells. Int J Biochem Cell Biol 2020; 122:105740. [PMID: 32173521 DOI: 10.1016/j.biocel.2020.105740] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 02/18/2020] [Accepted: 03/09/2020] [Indexed: 01/24/2023]
Abstract
BACKGROUND Long non-coding RNAs (lncRNAs) play important roles in regulation of gene expression and are involved in pathogenesis of different diseases including cancer. Recent studies suggested the lncRNA Colon cancer associated transcript-1 (CCAT1) to act as putative oncogene. In this study, to elucidate the role of this lncRNA in endometrial cancer, we examined its expression in normal endometrium and type 1 endometrial cancer and knocked down its expression in endometrial cancer cell lines followed by transcriptome and pathway analyses. METHODS CCAT1 expression was examined in 100 tissue samples of normal endometrium and type 1 endometrial cancer tissues by means of RT-qPCR. Knockdown of CCAT1 expression in HEC-1B and RL95/2 endometrial cancer cells was performed by siRNA transfection. Affymetrix GeneChip arrays were used to elucidate the effect of both lncRNAs on the transcriptome of these cell lines. RESULTS Median CCAT1 expression was found to be 9.3-fold higher in endometrial cancer when compared to normal endometrium (p < 0.05). In contrast to premenopausal endometrium and G1, G2 and G3 graded endometrial cancer, CCAT1 expression was nearly absent in postmenopausal tissue. Knockdown of CCAT1 by transient siRNA transfection significantly reduced proliferation of HEC-1B cancer cells in vitro by 35.5 % 6 days after transfection and notably reduced their colony formation ability. Affymetrix microarray and Ingenuity pathway analyses revealed a set of up- or down-regulated genes in transfected ERα-negative HEC-1B cells forming a network controlled by the key regulators TNF and TP53, including genes known to be involved in growth control, providing putative molecular mechanisms underlying the observed growth inhibition of HEC-1B cells. In contrast, CCAT1 knockdown in ERα-positive RL95/2 cells did not significantly affect proliferation, but resulted in down-regulation of a network of ERα target genes. CONCLUSIONS Given that the lncRNA CCAT1 was found to be overexpressed in endometrial cancer, affected the growth of HEC-1B cells and the expression of growth regulatory genes, our data suggest CCAT1 to exert oncogenic functions in endometrial cancer and encourage further studies to examine to what extent this lncRNA might be a potential therapy target in this cancer entity.
Collapse
Affiliation(s)
- Oliver Treeck
- Department of Obstetrics and Gynecology, University Medical Center Regensburg, Landshuter Str. 65, 93053, Regensburg, Germany.
| | - Maciej Skrzypczak
- Second Department of Gynecology, Medical University of Lublin, Jaczewskiego 8, PL 20-090, Lublin, Poland.
| | - Susanne Schüler-Toprak
- Department of Obstetrics and Gynecology, University Medical Center Regensburg, Landshuter Str. 65, 93053, Regensburg, Germany.
| | - Florian Weber
- Department of Pathology, University Medical Center Regensburg, Franz-Josef Strauß Allee 11, 93053, Regensburg, Germany.
| | - Olaf Ortmann
- Department of Obstetrics and Gynecology, University Medical Center Regensburg, Landshuter Str. 65, 93053, Regensburg, Germany.
| |
Collapse
|
9
|
Deng J, Wang W, Yu G, Ma X. MicroRNA‑195 inhibits epithelial‑mesenchymal transition by targeting G protein‑coupled estrogen receptor 1 in endometrial carcinoma. Mol Med Rep 2019; 20:4023-4032. [PMID: 31545414 PMCID: PMC6797983 DOI: 10.3892/mmr.2019.10652] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 06/25/2019] [Indexed: 12/12/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) has been shown to exert promoting effects on the progression of a number of cancer types, including endometrial carcinoma (EC). MicroRNA (miRNA or miR)-195 has been shown to function as a tumor suppressor. This study aimed to explore the potential role of miR-195 in the EMT process of EC. miR-195 overexpression (Mimics) and mimics control (Mock) vectors were constructed and transfected into human endometrial cancer cells (AN3-CA and Hec1A) using Lipofectamine 2000, and cell viability was detected using the Cell Counting kit-8 (CCK-8). The invasive and migratory capacities of the cells transfected with the Mimics or Mock vectors were assessed by Transwell and wound healing assays. Relative mRNA and protein levels were analyzed by reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blot analysis, respectively. Using TargetScan prediction, the potential target of miR-195 was identified and was further verified by dual-luciferase reporter assay. Following transfection with miR-195 mimics, the viability of the AN3-CA and Hec1A cells decreased in a time-dependent manner, specifically at 24 h. The wound closure rate and the number of invaded cells in the Mimics group were much lower than those in the Control and Mock groups (P<0.01). miR-195 overexpression significantly upregulated the mRNA and protein levels of tissue inhibitor of metalloproteinase 2 (TIMP-2), while it downregulated the expression levels of matrix metalloproteinase (MMP)-2 and MMP-9. Moreover, the phosphorylation levels of PI3K and AKT were also notably decreased (P<0.01). G protein-coupled estrogen receptor 1 (GPER) was identified as a target of miR-195. On the whole, the findings of this study indicate that the inhibitory effects of miR195 on EC cell migration and invasion are associated with the PI3K/AKT signaling pathway and GPER expression.
Collapse
Affiliation(s)
- Junfeng Deng
- Department of Obstetrics and Gynecology, Yantai Hospital of Traditional Chinese Medicine, Yantai, Shandong 264012, P.R. China
| | - Weihua Wang
- Department of Obstetrics and Gynecology, Yantai Hospital of Traditional Chinese Medicine, Yantai, Shandong 264012, P.R. China
| | - Guangyu Yu
- Department of Obstetrics and Gynecology, Yantai Hospital of Traditional Chinese Medicine, Yantai, Shandong 264012, P.R. China
| | - Xiuzhen Ma
- Department of Obstetrics and Gynecology, Yantai Hospital of Traditional Chinese Medicine, Yantai, Shandong 264012, P.R. China
| |
Collapse
|
10
|
Knockdown of estrogen receptor β increases proliferation and affects the transcriptome of endometrial adenocarcinoma cells. BMC Cancer 2019; 19:745. [PMID: 31357971 PMCID: PMC6664594 DOI: 10.1186/s12885-019-5928-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 07/12/2019] [Indexed: 01/29/2023] Open
Abstract
Background Estrogen receptor β (ERβ) has been repeatedly suggested to play important roles in hormone-dependent cancer like in tumors of the breast, ovary or prostate. In this study, we intended to further elucidate its role in endometrial cancer. Methods For this purpose, we knocked down ERβ expression in two endometrial cancer cell lines, the ERα-negative/ERβ-positive line HEC-1A and the ERα/β-positive cell line RL95/2, by means of siRNA transfection. Cell proliferation after transfection was assessed using the fluorescent CTB Assay (Promega). In order to elucidate possible molecular mechanisms which might underlie the effect on proliferation, we performed transcriptome analyses by means of human Affymetrix Human Gene Chip 2.0. Additionally, we treated the employed cell lines with different ERβ modulators to examine their effect on proliferation. Results siRNA-mediated knockdown of ERβ significantly increased proliferation of both endometrial cancer cell lines. In HEC-1A cells, proliferation was significantly increased 4, 5 and 6 days after transfection, with a maximum of about 1.7-fold (p < 0.05) on day 6. Endometrial RL95/2 cells with an ERβ knockdown exhibited a clearly enhanced proliferation on day 3 and days 4 to 8, when even 2.4-fold higher numbers of viable cells were detected (p < 0.01). Transcriptome analysis revealed that this was accompanied by increased expression of several genes being known to be upregulated in cancer, including proliferation-associated genes and oncogenes, and by repression of genes associated with differentiation, apoptosis or growth inhibition. Corroborating the observed knockdown effects, treatment with the ERβ antagonists PHTTP and (R, R) THC was also able to induce proliferation of both cell lines. Conclusions Our data clearly support the putative role of ERβ as tumor suppressor in endometrium as previously suggested in studies on other tissues and encourage further studies to find out to what extent this molecule might be a potential therapy target in this cancer entity. Electronic supplementary material The online version of this article (10.1186/s12885-019-5928-2) contains supplementary material, which is available to authorized users.
Collapse
|
11
|
Ayakannu T, Taylor AH, Konje JC. Cannabinoid receptor expression in estrogen-dependent and estrogen-independent endometrial cancer. J Recept Signal Transduct Res 2018; 38:385-392. [PMID: 30569804 DOI: 10.1080/10799893.2018.1531890] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The lack of good diagnostic/prognostic biomarkers and the often late presentation of endometrial cancer (EC) hinders the amelioration of the morbidity and mortality rates associated with this primarily estrogen-driven disease, a disease that is becoming more prevalent in the population. Previous studies on the expression of the classical cannabinoid receptors, CB1 and CB2, suggest these could provide good diagnostic/prognostic biomarkers for EC but those observations have been contradictory. In this study, we sought to resolve the inconsistency of CB1 and CB2 expression levels in different EC studies. To that end, we used qRT-PCR and immunohistochemistry (IHC) for CB1 and CB2 in endometrial biopsies from women with or without EC and found that transcript levels for both CB1 and CB2 were significantly decreased by 90 and 80%, respectively in EC. These observations were supported by histomorphometric studies where CB1 and CB2 staining intensity was decreased in all types of EC. These data suggest that the loss of both types of CB receptors is potentially involved in the development of or progression of EC and that CB1 and CB2 receptor expression could serve as useful histological markers and therapeutic targets in the treatment of or prevention of EC.
Collapse
Affiliation(s)
- Thangesweran Ayakannu
- a Reproductive Sciences Section, Department of Cancer Studies and Molecular Medicine , University of Leicester , Leicester , UK.,b Department of Gynaecology Oncology , Royal Surrey County Hospital , Guildford , UK.,c Department of Clinical and Experimental Medicine , University of Surrey , Guildford , UK
| | - Anthony H Taylor
- a Reproductive Sciences Section, Department of Cancer Studies and Molecular Medicine , University of Leicester , Leicester , UK.,d Department of Molecular and Cell Biology , University of Leicester, Leicester , UK
| | - Justin C Konje
- a Reproductive Sciences Section, Department of Cancer Studies and Molecular Medicine , University of Leicester , Leicester , UK.,e Department of Obstetrics and Gynaecology , Sidra Medical and Research Centre , Doha , Qatar
| |
Collapse
|
12
|
Lim S, Kim YH, Lee KB, Lee JM. The influence of hormone therapy with drospirenone-estradiol on endometrioid type endometrial cancer patients. J Gynecol Oncol 2018; 29:e72. [PMID: 30022635 PMCID: PMC6078887 DOI: 10.3802/jgo.2018.29.e72] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 04/17/2018] [Accepted: 04/22/2018] [Indexed: 02/04/2023] Open
Abstract
Objective To determine whether drospirenone/estradiol (DRSP/E2) has an adverse effect on clinical outcomes in surgically staged International Federation of Gynecology and Obstetrics (FIGO) stage I/II endometrial cancer (EC) patients. Methods In a retrospective case-controlled study, 58 women with EC who had received DRSP/E2 postoperatively were compared with 116 women who had not. And, oncologic safety of postoperative hormone therapy with DRSP/E2 in EC survivors were compared between the 2 groups after propensity score matching using a logistic regression model. Results The median ages were 47.7 years and 53.6 years for the study and the control groups, respectively (p<0.001). The study group had similar parity (p=0.71), lower body mass index (p=0.03) and more premenopausal women (p<0.001) than the control group. The stages were completely matched. The grades (p=0.42), lymphovascular space invasion (p=0.23), preoperative cancer antigen 125 (CA 125) level (p=0.89), and hormone receptor status (p=0.07) were similar in both groups. The median tumor diameter was statistically larger in the study group than in the control group (p<0.001). Both group received similar adjuvant therapy (p=0.80). In the propensity matching, only hormone receptor status was significantly different (p=0.03). In the univariate analysis, only stage was significantly associated with disease-free survival (DFS) and there was no variable associated with overall survival (OS). And, there was no significant factor identified in multivariate analysis. The difference in the DFS (p=0.63) and in the OS (p=0.32) was not significant. The same results were obtained after propensity score matching. Conclusion Postoperative hormone therapy with DRSP/E2 in EC survivors did not increase recurrence or the death rate.
Collapse
Affiliation(s)
- Soyi Lim
- Department of Obstetrics and Gynecology, Gachon University Gil Medical Center, Gachon University College of Medicine, Incheon, Korea
| | - Yun Hwan Kim
- Department of Obstetrics and Gynecology, Ewha Womans University Mokdong Hospital, Ewha Womans University College of Medicine, Seoul, Korea
| | - Kwang Beom Lee
- Department of Obstetrics and Gynecology, Gachon University Gil Medical Center, Gachon University College of Medicine, Incheon, Korea.
| | - Jong Min Lee
- Department of Obstetrics and Gynecology, Kyung-Hee University Hospital at Gangdong, Kyung Hee University School of Medicine, Seoul, Korea
| |
Collapse
|
13
|
Expression Pattern of G-Protein-Coupled Estrogen Receptor in Myometrium of Uteri with and without Adenomyosis. BIOMED RESEARCH INTERNATIONAL 2017; 2017:5974693. [PMID: 29109960 PMCID: PMC5646294 DOI: 10.1155/2017/5974693] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Accepted: 08/27/2017] [Indexed: 01/19/2023]
Abstract
Objective To compare the expression of G-protein-coupled estrogen receptor (GPER) in the junctional zone and outer myometrium of the proliferative and secretory phases of women with and without adenomyosis. Methods A total of 76 women were included in this study, 42 with adenomyosis (proliferative phase, n = 23; secretory phases, n = 19) and 34 controls (proliferative phase, n = 16; secretory phases, n = 18). Protein and total RNA were extracted from the junctional zone (JZ) and outer myometrium (OM). GPER protein and mRNA expression levels were evaluated by the use of western blotting and real-time quantitative polymerase chain reaction (RT-qPCR). Results The expression of GPER protein and mRNA in women with adenomyosis was significantly higher than that of control subjects, both in the junctional zone and in the outer myometrium and both in the proliferative and in the secretory phases. Conclusion The significant and consistent increase in GPER expression in adenomyosis compared with control subjects, regardless of whether it was in the proliferative or secretory phases and regardless of whether it was in the JZ or OM, suggests that GPER plays an important role in the pathogenesis of the adenomyosis.
Collapse
|
14
|
Zhang L, Li Y, Lan L, Liu R, Wu Y, Qu Q, Wen K. Tamoxifen has a proliferative effect in endometrial carcinoma mediated via the GPER/EGFR/ERK/cyclin D1 pathway: A retrospective study and an in vitro study. Mol Cell Endocrinol 2016; 437:51-61. [PMID: 27519631 DOI: 10.1016/j.mce.2016.08.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Revised: 07/12/2016] [Accepted: 08/08/2016] [Indexed: 10/21/2022]
Abstract
Tamoxifen has been widely used to treat breast cancer as an endocrine therapy. However, tamoxifen is known to enhance the risk of developing endometrial cancer. We want to examine the effect of tamoxifen on endometrial cancer. In our retrospective study, we found that high grade, high stage, and lymph node metastasis were more common in tamoxifen users. In vitro 4-hydroxytamoxifen (OHT) induced cell proliferation and cell cycle promotion in type I and type II endometrial cancer cell lines, and this proliferation was blocked by GPER silencing. Treatment with OHT increased EGFR and ERK phosphorylation and the mRNA and protein levels of cyclin D1 and GPER. Taken together, our data demonstrate that endometrial cancer patients with tamoxifen treatment exhibit more aggressive histological subtypes and worse prognosis. OHT is a proliferation-inducing agent for endometrial cancer cells, and the GPER/EFGR/ERK/cyclin D1 pathway is involved in this process.
Collapse
Affiliation(s)
- Lizhi Zhang
- Department of Obstetrics and Gynecology, Tianjin First Center Hospital, Tianjin, 300192, China.
| | - Yanmin Li
- Department of Pathology, Tianjin First Center Hospital, Tianjin, 300192, China.
| | - Lan Lan
- Department of Synergistic Chinese and Western Medicine Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China.
| | - Rong Liu
- Department of Obstetrics and Gynecology, Tianjin First Center Hospital, Tianjin, 300192, China.
| | - Yanhong Wu
- Department of Obstetrics and Gynecology, Tianjin First Center Hospital, Tianjin, 300192, China.
| | - Quanxin Qu
- Department of Obstetrics and Gynecology, Tianjin First Center Hospital, Tianjin, 300192, China.
| | - Ke Wen
- Department of Pharmacology, Tianjin Medical University, Tianjin, 300070, China.
| |
Collapse
|
15
|
Wang X, Simpson ER, Brown KA. Aromatase overexpression in dysfunctional adipose tissue links obesity to postmenopausal breast cancer. J Steroid Biochem Mol Biol 2015. [PMID: 26209254 DOI: 10.1016/j.jsbmb.2015.07.008] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The number of breast cancer cases has increased in the last a few decades and this is believed to be associated with the increased prevalence of obesity worldwide. The risk of breast cancer increases with age beyond menopause and the relationship between obesity and the risk of breast cancer in postmenopausal women is well established. The majority of postmenopausal breast cancers are estrogen receptor (ER) positive and estrogens produced in the adipose tissue promotes tumor formation. Obesity results in the secretion of inflammatory factors that stimulate the expression of the aromatase enzyme, which converts androgens into estrogens in the adipose tissue. Evidence demonstrating a link between obesity and breast cancer has led to the investigation of metabolic pathways as novel regulators of estrogen production, including pathways that can be targeted to inhibit aromatase specifically within the breast. This review aims to present some of the key findings in this regard.
Collapse
Affiliation(s)
- Xuyi Wang
- Metabolism & Cancer Laboratory, Centre for Cancer Research, Hudson Institute of Medical Research, Monash University, Clayton, Victoria, Australia; Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - Evan R Simpson
- Metabolism & Cancer Laboratory, Centre for Cancer Research, Hudson Institute of Medical Research, Monash University, Clayton, Victoria, Australia; Department of biochemistry & Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Kristy A Brown
- Metabolism & Cancer Laboratory, Centre for Cancer Research, Hudson Institute of Medical Research, Monash University, Clayton, Victoria, Australia; Department of Physiology, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
16
|
Ulrich L. HRT after endometrial cancer – Is it safe? Maturitas 2014; 79:237-8. [DOI: 10.1016/j.maturitas.2014.07.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 07/31/2014] [Indexed: 10/24/2022]
|
17
|
Gu Z, Ding G, Liang K, Zhang H, Guo G, Zhang L, Cui J. TESTIN suppresses tumor growth and invasion via manipulating cell cycle progression in endometrial carcinoma. Med Sci Monit 2014; 20:980-7. [PMID: 24929083 PMCID: PMC4067424 DOI: 10.12659/msm.890544] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The TESTIN gene was demonstrated to be a tumor suppressor in prostate and breast cancer through inhibiting tumor growth and invasion. Herein, we aimed to investigate the detailed functions of TESTIN in the highly sexual hormone (estrogen)-dependent malignancy, endometrial carcinoma. MATERIAL AND METHODS TESTIN mRNA and protein expression were measured by qRT-PCR, Western blot and immunohistochemistry. Upregulation of TESTIN was achieved by transfecting the pcDNA3.1-TESTIN plasmids into AN3CA cells. Knockdown of TESTIN was achieved by transfecting the shRNA-TESTIN into Ishikawa cells. MTT assay, colony formation assay, and Transwell assay were used to investigate the effects of TESTIN on cellular proliferation and invasion. The apoptotic status and cell cycle were analyzed using flow cytometry. MMP2 secretion was determined by ELISA assay. The xenograft assay was used to investigate the functions of TESTIN in nude mice. RESULTS Compared to the non-malignant adjacent endometrium, 54% of tumor samples presented downregulation of TESTIN (P<0.001). Loss of TESTIN protein was correlated with advanced tumor stage (P=0.047), high grade (P=0.034), and lymphatic vascular space invasion (P=0.036). In vitro, overexpression of TESTIN suppressed cell proliferation, induced dramatic G1 arrest, and inhibited tumor invasion through blocking the secretion of MMP2. Loss of TESTIN accelerated cellular proliferation, promoted cell cycle progression, and enhanced tumor invasion by increasing the secretion of MMP2. Consistently, TESTIN could significantly delay the growth of xenografts in nude mice. CONCLUSIONS TESTIN was commonly downregulated in human endometrial carcinoma and was associated with poor prognostic markers. Moreover, TESTIN significantly inhibited tumor growth and invasion via arresting cell cycle in in vitro and in vivo experiments. Therefore, we propose that TESTIN might be a prognostic marker and therapeutic target for endometrial carcinoma.
Collapse
Affiliation(s)
- Zhenpeng Gu
- Department of Obstetrics and Gynecology, Affiliated Hospital of Binzhou Medical College, Binzhou, China (mainland)
| | - Guofeng Ding
- Department of Infectious Diseases, Affiliated Hospital of Binzhou Medical College, Binzhou, China (mainland)
| | - Kuixiang Liang
- Department of Obstetrics and Gynecology, Affiliated Hospital of Binzhou Medical College, Binzhou, China (mainland)
| | - Hongtao Zhang
- Department of Obstetrics and Gynecology, Affiliated Hospital of Binzhou Medical College, Binzhou, China (mainland)
| | - Guanghong Guo
- Department of Obstetrics and Gynecology, Affiliated Hospital of Binzhou Medical College, Binzhou, China (mainland)
| | - Lili Zhang
- Department of Obstetrics and Gynecology, Affiliated Hospital of Binzhou Medical College, Binzhou, China (mainland)
| | - Jinxiu Cui
- Department of Obstetrics and Gynecology, Affiliated Hospital of Binzhou Medical College, Binzhou, China (mainland)
| |
Collapse
|
18
|
Wang J, Hu F, Luo Y, Luo H, Huang N, Cheng F, Deng Z, Deng W, Zou K. Estrogenic and anti-estrogenic activities of hispolon from Phellinus lonicerinus (Bond.) Bond. et sing. Fitoterapia 2014; 95:93-101. [PMID: 24637110 DOI: 10.1016/j.fitote.2014.03.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 03/06/2014] [Accepted: 03/09/2014] [Indexed: 01/25/2023]
Abstract
Hispolon was the main antitumor active ingredient in Phellinus sensu lato species. In order to confirm the dual regulating estrogenic ingredient and obtain more effective natural estrogen replacement drugs, hispolon was separated from Phellinus lonicerinus (Bond.) Bond. et sing. Hispolon exhibited significant anti-proliferative effect against estrogen-sensitive ER (+) MCF-7 cells in the absence of estrogen, and exhibits antagonistic effects on 17β-estradiol (E2)-induced MCF-7 cell proliferation when E2 and the different concentrations of hispolon were treated simultaneously. Hispolon also inhibited the proliferation of estrogen-negative ER (-) MDA-MB-231 cells at the concentration of 5.00×10(-5) M. The yeast two-hybrid experiments showed that hispolon had strong and non-selective effects on the estrogen receptor (ER) α and ERβ at a concentration of 1.00×10(-6) M. The ERβ-binding ability of hispolon was larger than ERα in the concentration range of 1.00×10(-9) M and 1.00×10(-7) M. Hispolon could increase the body weight coefficient, serum E2 and progesterone contents in immature female mice at dose of 9.10×10(-6) mol/kg, and increase coefficient of thymus and spleen in mice. The Gscores of hispolon-ERα and hispolon-ERβ docked complexes were -7.93 kcal/mol and -7.79 kcal/mol in docking simulations. Hispolon presented dual regulating estrogenic activities, which showed estrogenic agonist activity at low concentration or lack of endogenous estrogen, and the estrogenic antagonistic effect was stimulated at high concentrations or too much endogenous estrogen. Hispolon could be used for treating the estrogen deficiency-related disease with the benefit of non-toxic to normal cells, good antitumor effects and estrogenic activity.
Collapse
Affiliation(s)
- Junzhi Wang
- Hubei Key Laboratory of Natural Products Research and Development, China Three Gorges University, Yichang 443002, China; Hubei Tujia Institute of Medicine, China Three Gorges University, Yichang 443002, China
| | - Fang Hu
- Hubei Key Laboratory of Natural Products Research and Development, China Three Gorges University, Yichang 443002, China
| | - Youcheng Luo
- Hubei Key Laboratory of Natural Products Research and Development, China Three Gorges University, Yichang 443002, China
| | - Huajun Luo
- Hubei Key Laboratory of Natural Products Research and Development, China Three Gorges University, Yichang 443002, China
| | - Nianyu Huang
- Hubei Key Laboratory of Natural Products Research and Development, China Three Gorges University, Yichang 443002, China.
| | - Fan Cheng
- Hubei Key Laboratory of Natural Products Research and Development, China Three Gorges University, Yichang 443002, China
| | - Zhangshuang Deng
- Hubei Key Laboratory of Natural Products Research and Development, China Three Gorges University, Yichang 443002, China
| | - Weiqiao Deng
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Kun Zou
- Hubei Key Laboratory of Natural Products Research and Development, China Three Gorges University, Yichang 443002, China
| |
Collapse
|
19
|
G protein-coupled estrogen receptor-selective ligands modulate endometrial tumor growth. Obstet Gynecol Int 2013; 2013:472720. [PMID: 24379833 PMCID: PMC3863501 DOI: 10.1155/2013/472720] [Citation(s) in RCA: 122] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Accepted: 09/17/2013] [Indexed: 01/28/2023] Open
Abstract
Endometrial carcinoma is the most common cancer of the female reproductive tract. GPER/GPR30 is a 7-transmembrane spanning G protein-coupled receptor that has been identified as the third estrogen receptor, in addition to ERα and ERβ. High GPER expression is predictive of poor survival in endometrial and ovarian cancer, but despite this, the estrogen-mediated signaling pathways and specific estrogen receptors involved in endometrial cancer remain unclear. Here, employing ERα-negative Hec50 endometrial cancer cells, we demonstrate that GPER mediates estrogen-stimulated activation of ERK and PI3K via matrix metalloproteinase activation and subsequent transactivation of the EGFR and that ER-targeted therapeutic agents (4-hydroxytamoxifen, ICI182,780/fulvestrant, and Raloxifene), the phytoestrogen genistein, and the “ERα-selective” agonist propylpyrazole triol also function as GPER agonists. Furthermore, xenograft tumors of Hec50 cells yield enhanced growth with G-1 and estrogen, the latter being inhibited by GPER-selective pharmacologic antagonism with G36. These results have important implications with respect to the use of putatively ER-selective ligands and particularly for the widespread long-term use of “ER-targeted” therapeutics. Moreover, our findings shed light on the potential mechanisms of SERM/SERD side effects reported in many clinical studies. Finally, our results provide the first demonstration that pharmacological inhibition of GPER activity in vivo prevents estrogen-mediated tumor growth.
Collapse
|
20
|
Expression of estrogen receptors (α, β), cyclooxygenase-2 and aromatase in normal endometrium and endometrioid cancer of uterus. Adv Med Sci 2013; 58:96-103. [PMID: 23625281 DOI: 10.2478/v10039-012-0055-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
PURPOSE Endometrial cancer (EC) is one of the most common malignancies of the female genital tract, but the etiology, especially its metabolism is still investigated. The aim of this study was to evaluate the presence and relative expression of Estrogen Receptors (α, β), Cyclooxygenase-2 and Aromatase in both endometrial cancer and normal mucosa. MATERIAL/METHODS Two groups of women were selected for the study: 1) patients with endometrioid endometrial cancer (FIGO I; G1 - G3) (n=35) and 2) subjects with normal endometrial tissue (control group, n=29). The expression of Estrogen Receptors (ERα, β), Cyclooxygenase-2 (COX-2), Aromatase were estimated by Western blot analysis. Furthermore, the associations between FIGO classification (stage: Ia, Ib), tumor grade (G) and expression of ERα, β, COX-2, aromatase proteins were evaluated. Overall and disease-free survival curves were generated according to the Kaplan-Meier method. Median follow-up time of the patients examined in this study was 39 months. RESULTS The relative expression of each examined protein was markedly higher in the endometrial cancer tissue as compared to the healthy endometrium. The trends towards greater expression along with a tumor progression was noticed (FIGO stage: Ia vs. Ib). Analysis of endometrial cancer risk factors and their influence on survival curves showed only an inverse significant correlations between obesity (BMI: 36.2; n=21) and disease-free survival in EC group (p=0.00872), but there was no significant association between obesity and overall survival (p=0.358). CONCLUSIONS Endometrioid endometrial cancer shows relatively higher expression of either ER, COX-2 and aromatase comparing to healthy mucosa, suggesting their involvement in tumor development and progression.
Collapse
|
21
|
Dreisler E, Poulsen LG, Antonsen SL, Ceausu I, Depypere H, Erel CT, Lambrinoudaki I, Pérez-López FR, Simoncini T, Tremollieres F, Rees M, Ulrich LG. EMAS clinical guide: Assessment of the endometrium in peri and postmenopausal women. Maturitas 2013; 75:181-90. [DOI: 10.1016/j.maturitas.2013.03.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
22
|
Abstract
Most postmenopausal vaginal bleeding is benign; however, it merits diagnostic evaluation by transvaginal ultrasound or endometrial biopsy after emergency department evaluation. Patients and physicians may treat menopausal symptoms with hormone replacement therapy or other agents, such as venlafaxine or gabapentin. Hormone replacement therapy, when initiated close to the start of menopause and continued at the lowest possible dose for the shortest possible duration, carries less risk than previously believed. Pelvic organ prolapse affects millions of women and may contribute to poor body image and difficulty with urinary, gastrointestinal, and sexual function. Treatment options include Kegel exercises, pessaries, and surgery.
Collapse
|
23
|
Hursting SD, Digiovanni J, Dannenberg AJ, Azrad M, Leroith D, Demark-Wahnefried W, Kakarala M, Brodie A, Berger NA. Obesity, energy balance, and cancer: new opportunities for prevention. Cancer Prev Res (Phila) 2012; 5:1260-72. [PMID: 23034147 DOI: 10.1158/1940-6207.capr-12-0140] [Citation(s) in RCA: 137] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Obesity is associated with increased risk and poor prognosis for many types of cancer. The mechanisms underlying the obesity-cancer link are becoming increasingly clear and provide multiple opportunities for primary to tertiary prevention. Several obesity-related host factors can influence tumor initiation, progression and/or response to therapy, and these have been implicated as key contributors to the complex effects of obesity on cancer incidence and outcomes. These host factors include insulin, insulin-like growth factor-I, leptin, adiponectin, steroid hormones, cytokines, and inflammation-related molecules. Each of these host factors is considered in the context of energy balance and as potential targets for cancer prevention. The possibility of prevention at the systems level, including energy restriction, dietary composition, and exercise is considered as is the importance of the newly emerging field of stem cell research as a model for studying energy balance and cancer prevention.
Collapse
|
24
|
Current world literature. Curr Opin Oncol 2012; 24:587-95. [PMID: 22886074 DOI: 10.1097/cco.0b013e32835793f1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
25
|
p53 is correlated with low BMI negative progesterone receptor status and recurring disease in patients with endometrial cancer. Gynecol Oncol 2011; 125:200-7. [PMID: 22210468 DOI: 10.1016/j.ygyno.2011.12.443] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2011] [Revised: 12/18/2011] [Accepted: 12/19/2011] [Indexed: 01/13/2023]
Abstract
OBJECTIVE P53 tumor suppressor gene plays a role in endometrial carcinogenesis. Former studies described correlations between p53 protein overexpression in endometrial cancer and prognostic factors, measured by immunohistochemistry. But data is still controversial. The aim of this study was to measure p53 and phospho-p53 overexpression by Western blot and evaluate correlations between overexpression and prognostic and clinical factors. Phospho-p53 seems to be the functional p53 protein and was examined for the first time in endometrial cancer. METHODS 40 patients with endometrial cancer were included in the study. A control group of 20 patients with normal endometrial tissue samples was used. Western blot was performed for detection of p53 and phospho-p53. Clinical and pathological parameters were obtained from medical records. Statistical analysis was performed using the log-rank test, the Mann-Whitney test for two independent groups and the Fisher's exact test for dichotomous groupings. RESULTS In 17.5% of the patients with endometrial cancer a p53 overexpression could be evaluated. There was a correlation between a p53 overexpression and recurring disease (p: 0.014), a negative progesterone receptor status (p: 0.021) and a low BMI (p: 0.022). Only one of 40 patients had a phospho-p53 expression. CONCLUSION Western blot is a valid method for the detection of p53 overexpression. As other authors described before, p53 overexpression seems to correlate with negative prognostic factors. The correlation between p53 overexpression and a low BMI may underline the relationship between p53 alterations and biological aggressive endometrial carcinomas.
Collapse
|
26
|
SNP285C modulates oestrogen receptor/Sp1 binding to the MDM2 promoter and reduces the risk of endometrial but not prostatic cancer. Eur J Cancer 2011; 48:1988-96. [PMID: 22119201 DOI: 10.1016/j.ejca.2011.10.024] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Revised: 09/30/2011] [Accepted: 10/18/2011] [Indexed: 11/24/2022]
Abstract
INTRODUCTION The MDM2 promoter polymorphism (SNP309T > G) extends a binding site for the transcription factor Sp1 and has been linked to elevated cancer risk and/or young age at cancer diagnosis, especially in females. Recently, we reported an adjacent polymorphism (SNP285G > C). SNP285C antagonises the effect of SNP309G by reducing Sp1 binding and lowers the risk of breast and ovarian cancer. METHODS We assessed the potential gender specificity in the effect of this polymorphism. We performed in silico predictions of transcription factor binding sites in the MDM2 promoter and analysed MDM2 SNP285 and SNP309 status in two independent cohorts of endometrial (n = 438 and 472) and 666 prostatic cancer patients, and compared to 3.140 healthy controls. RESULTS We identified three oestrogen-receptor binding elements (EREs) within the MDM2 intronic promoter, one of which overlapping the Sp1 binding-site harbouring SNP285. The SNP285C/309G haplotype was associated with a reduced Odds Ratio (OR) for endometrial cancer (OR1: 0.55; Confidence Interval (CI) 0.32-0.97; OR2: 0.65; CI 0.40-1.08, especially for ER+ tumours; OR: 0.48; CI 0.28-0.87) but not for prostatic cancer among SNP309TG heterozygotes. SNP309G (SNP309TG or SNP309GG genotype) was associated with a moderately increased risk of endometrial cancer (OR: 1.17; CI 1.00-1.37) compared to SNP309TT homozygotes. Removing individuals harbouring the SNP309G-counteracting SNP285C polymorphism from the analysis strengthened this association (OR: 1.20; CI 1.02-1.41). CONCLUSION The finding of an ERE overlapping with the Sp1-binding site affected by SNP285, taken together with the significant impact of SNP285 on the risk of breast, ovarian and now endometrial cancer but not prostatic cancer, suggests a gender specific effect of SNP285C on cancer risk.
Collapse
|