1
|
Wang SE, Viallon V, Lee M, Dimou N, Hamilton F, Biessy C, O'Mara T, Kyrgiou M, Crosbie EJ, Truong T, Severi G, Kaaks R, Fortner RT, Schulze MB, Bendinelli B, Sabina S, Tumino R, Sacerdote C, Panico S, Crous-Bou M, Sánchez MJ, Aizpurua A, Palacios DR, Guevara M, Travis RC, Tsilidis KK, Heath A, Yarmolinsky J, Rinaldi S, Gunter MJ, Dossus L. Circulating inflammatory and immune response proteins and endometrial cancer risk: a nested case-control study and Mendelian randomization analyses. EBioMedicine 2024; 108:105341. [PMID: 39278107 PMCID: PMC11418138 DOI: 10.1016/j.ebiom.2024.105341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 08/30/2024] [Accepted: 09/01/2024] [Indexed: 09/17/2024] Open
Abstract
BACKGROUND Inflammation and immune dysregulation are hypothesized contributors to endometrial carcinogenesis; however, the precise underlying mechanisms remain unclear. METHODS We measured pre-diagnostically 152 plasma protein biomarkers in 624 endometrial cancer case-control pairs nested within the European Prospective Investigation into Cancer and Nutrition (EPIC) cohort. Odds ratios (ORs) were estimated using conditional logistic regression, accounting for confounding and multiple comparisons. Proteins considered as associated with endometrial cancer risk were further tested in a two-sample Mendelian randomization (MR) analysis using summary data from the UK Biobank (n = 52,363) and the Endometrial Cancer Association Consortium (12,270 cases and 46,126 controls). FINDINGS In the EPIC nested case-control study, IL-6 [OR per NPX (doubling of concentration) = 1.28 (95% confidence interval (CI) 1.03-1.57)], HGF [1.48 (1.06-2.07)], PIK3AP1 [1.22 (1.00-1.50)] and CLEC4G [1.52 (1.00-2.32)] were positively associated; HSD11B1 [0.67 (0.49-0.91)], SCF [0.68 (0.49-0.94)], and CCL25 [0.80 (0.65-0.99)] were inversely associated with endometrial cancer risk; all estimates had multiple comparisons adjusted P-value > 0.05. In complementary MR analysis, IL-6 [OR per inverse-rank normalized NPX = 1.19 (95% CI 1.04-1.36)] and HSD11B1 [0.91 (0.84-0.99)] were associated with endometrial cancer risk. INTERPRETATION Altered IL-6 signalling and reduced glucocorticoid activity via HSD11B1 might play important roles in endometrial carcinogenesis. FUNDING Funding for IIG_FULL_2021_008 was obtained from Wereld Kanker Onderzoek Fonds (WKOF), as part of the World Cancer Research Fund International grant programme; Funding for INCA_15849 was obtained from Institut National du Cancer (INCa).
Collapse
Affiliation(s)
- Sabrina E Wang
- International Agency for Research on Cancer, World Health Organization, Lyon, France.
| | - Vivian Viallon
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Matthew Lee
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Niki Dimou
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Fergus Hamilton
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom; Infection Science, North Bristol NHS Trust, Bristol, United Kingdom
| | - Carine Biessy
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Tracy O'Mara
- Cancer Research Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Maria Kyrgiou
- Department of Metabolism, Digestion & Reproduction - Surgery and Cancer, Faculty of Medicine, Imperial College London, London, United Kingdom; West London Gynaecological Cancer Centre, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Emma J Crosbie
- Gynaecological Oncology Research Group, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, St Mary's Hospital, Manchester, United Kingdom; Department of Obstetrics and Gynaecology, St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Therese Truong
- Paris-Saclay University, UVSQ, Inserm, Gustave Roussy, CESP, Villejuif, France
| | - Gianluca Severi
- Paris-Saclay University, UVSQ, Inserm, Gustave Roussy, CESP, Villejuif, France; Department of Statistics, Computer Science, Applications "G. Parenti", University of Florence, Florence, Italy
| | - Rudolf Kaaks
- Division of Cancer Epidemiology, German Cancer Research Center, Heidelberg, Germany
| | - Renée Turzanski Fortner
- Division of Cancer Epidemiology, German Cancer Research Center, Heidelberg, Germany; Department of Research, Cancer Registry of Norway, Norwegian Institute of Public Health, Oslo, Norway
| | - Matthias B Schulze
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany; Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
| | - Benedetta Bendinelli
- Clinical Epidemiology Unit, Institute for Cancer Research, Prevention and Clinical Network (ISPRO), Florence, Italy
| | - Sieri Sabina
- Epidemiology and Prevention Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Rosario Tumino
- Hyblean Association for Epidemiological Research, Aire Onlus, Ragusa, Italy
| | - Carlotta Sacerdote
- Unit of Cancer Epidemiology, Città della Salute e della Scienza Hospital, University of Turin and CPO Piemonte, Turin, Italy
| | | | - Marta Crous-Bou
- Unit of Nutrition and Cancer, Cancer Epidemiology Research Program, Catalan Institute of Oncology (ICO) - Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, USA
| | - Maria-Jose Sánchez
- Escuela Andaluza de Salud Pública (EASP), Granada, Spain; Instituto de Investigación Biosanitaria, Granada, Spain; Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Amaia Aizpurua
- Sub Directorate for Public Health and Addictions of Gipuzkoa, Ministry of Health of the Basque Government, San Sebastian, Spain; Epidemiology of Chronic and Communicable Diseases Group, Biodonostia Health Research Institute, San Sebastián, Spain
| | - Daniel Rodriguez Palacios
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain; Department of Epidemiology, Murcia Regional Health Council-IMIB, Murcia, Spain
| | - Marcela Guevara
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain; Instituto de Salud Pública y Laboral de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Ruth C Travis
- Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Konstantinos K Tsilidis
- Cancer Epidemiology and Prevention Research Unit, School of Public Health, Imperial College London, London, United Kingdom; Department of Hygiene and Epidemiology, University of Ioannina School of Medicine, Ioannina, Greece
| | - Alicia Heath
- Cancer Epidemiology and Prevention Research Unit, School of Public Health, Imperial College London, London, United Kingdom
| | - James Yarmolinsky
- Cancer Epidemiology and Prevention Research Unit, School of Public Health, Imperial College London, London, United Kingdom
| | - Sabina Rinaldi
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Marc J Gunter
- International Agency for Research on Cancer, World Health Organization, Lyon, France; Cancer Epidemiology and Prevention Research Unit, School of Public Health, Imperial College London, London, United Kingdom
| | - Laure Dossus
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| |
Collapse
|
2
|
Ruiz-Ochoa D, Guerra-Ruiz AR, García-Unzueta MT, Muñoz-Cacho P, Rodriguez-Montalvan B, Amado-Diago CA, Lavín-Gómez BA, Cano-García ME, Pablo-Marcos D, Vázquez LA. Sex hormones and the total testosterone:estradiol ratio as predictors of severe acute respiratory syndrome coronavirus 2 infection in hospitalized men. Andrology 2024; 12:1381-1388. [PMID: 38212146 DOI: 10.1111/andr.13581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 12/07/2023] [Accepted: 12/12/2023] [Indexed: 01/13/2024]
Abstract
BACKGROUND The predictive ability of the early determination of sex steroids and the total testosterone:estradiol ratio for the risk of severe coronavirus disease 2019 or the potential existence of a biological gradient in this relationship has not been evaluated. OBJECTIVES To assess the relationship of sex steroid levels and the total testosterone:estradiol ratio with the risk of severe acute respiratory syndrome coronavirus 2 infection in men, defined as the need for intensive care unit admission or death, and the predictive ability of each biomarker. MATERIALS AND METHODS This was a prospective observational study. We included all consecutive adult men with severe acute respiratory syndrome coronavirus 2 infections in a single center admitted to a general hospital ward or to the intensive care unit. Sex steroids were evaluated at the centralized laboratory of our hospital. RESULTS We recruited 98 patients, 54 (55.1%) of whom developed severe coronavirus disease in 2019. Compared to patients with nonsevere coronavirus disease 2019, patients with severe coronavirus disease 2019 had significantly lower serum levels of total testosterone (111 ± 89 vs. 191 ± 143 ng/dL; p < 0.001), dehydroepiandrosterone (1.69 ± 1.26 vs. 2.96 ± 2.64 ng/mL; p < 0.001), and dehydroepiandrosterone sulfate (91.72 ± 76.20 vs. 134.28 ± 98.261 μg/dL; p = 0.009), significantly higher levels of estradiol (64.61 ± 59.35 vs. 33.78 ± 13.78 pg/mL; p = 0.001), and significantly lower total testosterone:estradiol ratio (0.28 ± 0.31 vs. 0.70 ± 0.75; p < 0.001). The lower the serum level of androgen and the lower the total testosterone:estradiol ratio values, the higher the likelihood of developing severe coronavirus disease 2019, with the linear trend in the adjusted analyses being statistically significant for all parameters except for androstenedione (p = 0.064). In the receiver operating characteristic analysis, better predictive performance was shown by the total testosterone:estradiol ratio, with an area under the curve of 0.77 (95% confidence interval 0.68-0.87; p < 0.001). DISCUSSION AND CONCLUSION Our results suggest that men with severe acute respiratory syndrome coronavirus 2 infection, decreased androgen levels and increased estradiol levels have a higher likelihood of developing an unfavorable outcome. The total testosterone:estradiol ratio showed the best predictive ability.
Collapse
Affiliation(s)
- David Ruiz-Ochoa
- Department of Endocrinology and Nutrition, Marqués de Valdecilla University Hospital, Santander, Spain
| | - Armando-Raúl Guerra-Ruiz
- Department of Clinical Biochemistry, Marqués de Valdecilla University Hospital, Santander, Spain
- IDIVAL Health Research Institute, Santander, Spain
- University of Cantabria, Santander, Spain
| | - María-Teresa García-Unzueta
- Department of Clinical Biochemistry, Marqués de Valdecilla University Hospital, Santander, Spain
- IDIVAL Health Research Institute, Santander, Spain
- University of Cantabria, Santander, Spain
| | - Pedro Muñoz-Cacho
- IDIVAL Health Research Institute, Santander, Spain
- Department of Medicine and Psychiatry, Gerencia de Atención Primaria, Servicio Cántabro de Salud, Santander, Spain
| | | | - Carlos Antonio Amado-Diago
- IDIVAL Health Research Institute, Santander, Spain
- University of Cantabria, Santander, Spain
- Department of Pneumology, Marqués de Valdecilla University Hospital, Santander, Spain
| | - Bernardo-Alio Lavín-Gómez
- Department of Clinical Biochemistry, Marqués de Valdecilla University Hospital, Santander, Spain
- IDIVAL Health Research Institute, Santander, Spain
| | - María-Eliecer Cano-García
- Department of Microbiology, Marqués de Valdecilla University Hospital, Servicio Cántabro de Salud, Santander, Spain
| | - Daniel Pablo-Marcos
- Department of Microbiology, Marqués de Valdecilla University Hospital, Servicio Cántabro de Salud, Santander, Spain
| | - Luis Alberto Vázquez
- Department of Endocrinology and Nutrition, Marqués de Valdecilla University Hospital, Santander, Spain
- IDIVAL Health Research Institute, Santander, Spain
- University of Cantabria, Santander, Spain
| |
Collapse
|
3
|
Baker Frost D, Savchenko A, Takamura N, Wolf B, Fierkens R, King K, Feghali-Bostwick C. A Positive Feedback Loop Exists between Estradiol and IL-6 and Contributes to Dermal Fibrosis. Int J Mol Sci 2024; 25:7227. [PMID: 39000334 PMCID: PMC11241801 DOI: 10.3390/ijms25137227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 06/23/2024] [Accepted: 06/28/2024] [Indexed: 07/16/2024] Open
Abstract
Systemic sclerosis (SSc) is characterized by dermal fibrosis with a female predominance, suggesting a hormonal influence. Patients with SSc have elevated interleukin (IL)-6 levels, and post-menopausal women and older men also have high estradiol (E2) levels. In the skin, IL-6 increases the enzymatic activity of aromatase, thereby amplifying the conversion of testosterone to E2. Therefore, we hypothesized that an interplay between E2 and IL-6 contributes to dermal fibrosis. We used primary dermal fibroblasts from healthy donors and patients with diffuse cutaneous (dc)SSc, and healthy donor skin tissues stimulated with recombinant IL-6 and its soluble receptor (sIL-6R) or E2. Primary human dermal fibroblasts and tissues from healthy donors stimulated with IL-6+sIL-6R produced E2, while E2-stimulated dermal tissues and fibroblasts produced IL-6. Primary dermal fibroblasts from healthy donors treated with IL-6+sIL-6R and the aromatase inhibitor anastrozole (ANA) and dcSSc fibroblasts treated with ANA produced less fibronectin (FN), type III collagen A1 (Col IIIA1), and type V collagen A1 (Col VA1). Finally, dcSSc dermal fibroblasts treated with the estrogen receptor inhibitor fulvestrant also generated less FN, Col IIIA1, and Col VA1. Our data show that IL-6 exerts its pro-fibrotic influence in human skin in part through E2 and establish a positive feedback loop between E2 and IL-6.
Collapse
Affiliation(s)
- DeAnna Baker Frost
- Department of Medicine, Division of Rheumatology and Immunology, Medical University of South Carolina, 96 Jonathan Lucas Street, Suite 822, MSC 637, Charleston, SC 29425, USA;
| | - Alisa Savchenko
- College of Osteopathic Medicine, Rocky Vista University, 4130 Rocky Vista Way, Billings, MT 59106, USA;
| | - Naoko Takamura
- Department of Environmental Immuno-Dermatology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Kanagawa, Japan;
| | - Bethany Wolf
- Department of Public Health Sciences, Medical University of South Carolina, 135 Cannon Street, Room 305F, Charleston, SC 29425, USA;
| | - Roselyn Fierkens
- Barabara Davis Center, Department of Pediatrics, University of Colorado, School of Medicine, M20-3201N, 1775 Aurora Court, Aurora, CO 80045, USA;
| | - Kimberly King
- School of Medicine, Morehouse College, 720 Westview Drive, Atlanta, GA 30310, USA;
| | - Carol Feghali-Bostwick
- Department of Medicine, Division of Rheumatology and Immunology, Medical University of South Carolina, 96 Jonathan Lucas Street, Suite 822, MSC 637, Charleston, SC 29425, USA;
| |
Collapse
|
4
|
Wang X, Kong F, Liu Y, Lv S, Zhang K, Sun S, Liu J, Wang M, Cai X, Jin H, Yan S, Luo J. 17β-estradiol biosensors based on different bioreceptors and their applications. Front Bioeng Biotechnol 2024; 12:1347625. [PMID: 38357703 PMCID: PMC10864596 DOI: 10.3389/fbioe.2024.1347625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 01/16/2024] [Indexed: 02/16/2024] Open
Abstract
17β-Estradiol (E2) is a critical sex steroid hormone, which has significant effects on the endocrine systems of both humans and animals. E2 is also believed to play neurotrophic and neuroprotective roles in the brain. Biosensors present a powerful tool to detect E2 because of their small, efficient, and flexible design. Furthermore, Biosensors can quickly and accurately obtain detection results with only a small sampling amount, which greatly meets the detection of the environment, food safety, medicine safety, and human body. This review focuses on previous studies of biosensors for detecting E2 and divides them into non-biometric sensors, enzyme biosensors, antibody biosensors, and aptamer biosensors according to different bioreceptors. The advantages, disadvantages, and design points of various bioreceptors for E2 detection are analyzed and summarized. Additionally, applications of different bioreceptors of E2 detection are presented and highlight the field of environmental monitoring, food and medicine safety, and disease detection in recent years. Finally, the development of E2 detection by biosensor is prospected.
Collapse
Affiliation(s)
- Xinyi Wang
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Fanli Kong
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Yaoyao Liu
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Shiya Lv
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Kui Zhang
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Shutong Sun
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Juntao Liu
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Mixia Wang
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Xinxia Cai
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Hongyan Jin
- Obstetrics and Gynecology Department, Peking University First Hospital, Beijing, China
| | - Shi Yan
- Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jinping Luo
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
5
|
Baker Frost D, Savchenko A, Feghali-Bostwick C, Wolf B. The Relationship between Time, Race, and Estrogen Receptor Alpha in Estradiol-Induced Dermal Fibrosis. Biomedicines 2024; 12:182. [PMID: 38255287 PMCID: PMC10813671 DOI: 10.3390/biomedicines12010182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 12/08/2023] [Accepted: 12/14/2023] [Indexed: 01/24/2024] Open
Abstract
In the skin, estradiol (E2) promotes profibrotic and proinflammatory cytokines, contributing to extracellular matrix (ECM) deposition. However, the magnitude of the response differs. Using the human skin organ culture model, we evaluated donor characteristics and correlations that contribute to E2-induced interleukin-6 (IL-6), transforming growth factor beta 1 and 2 (TGFB1 and TGFB2), collagen IA2 (Col IA2), collagen IIIA1 (Col IIIA1), and fibronectin (FN) expressions. In vehicle- and E2-treated dermal skin tissue transcripts, we confirm differences in the magnitude; however, there were positive correlations between profibrotic mediators and ECM components 48 h after E2 treatment. Also, positive correlations exist between baseline and E2-induced TGFB1, IL-6, Col IIIA1, and FN transcripts. Since estrogen receptor alpha (ERA) can propagate E2's signal, we measured and detected differences in its baseline and fold change transcript levels, with a significant decline in baseline levels 48 h after incubation and an increase 48 h after E2 treatment. There was a trend to higher transcript levels in African American donors 24 h earlier. Finally, E2-induced ERA transcript levels negatively correlated with its own baseline levels and positively correlated with FN, TGFB1, and Col IA2 transcript levels. Therefore, our data suggest ERA, E2 exposure time, and race/ethnicity contribute to E2-induced dermal fibrosis.
Collapse
Affiliation(s)
- DeAnna Baker Frost
- Department of Medicine, Division of Rheumatology and Immunology, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC 29425, USA; (D.B.F.); (C.F.-B.)
| | - Alisa Savchenko
- Chobanian & Avedisian School of Medicine, Boston University, 72 E. Concord Street, Boston, MA 02118, USA;
| | - Carol Feghali-Bostwick
- Department of Medicine, Division of Rheumatology and Immunology, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC 29425, USA; (D.B.F.); (C.F.-B.)
| | - Bethany Wolf
- Department of Public Health Sciences, Medical University of South Carolina, 135 Cannon Street, Room 305F, Charleston, SC 29425, USA
| |
Collapse
|
6
|
Ge Y, Ni X, Li J, Ye M, Jin X. Roles of estrogen receptor α in endometrial carcinoma (Review). Oncol Lett 2023; 26:530. [PMID: 38020303 PMCID: PMC10644365 DOI: 10.3892/ol.2023.14117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/06/2023] [Indexed: 12/01/2023] Open
Abstract
Endometrial carcinoma (EC) is a group of endometrial epithelial malignancies, most of which are adenocarcinomas and occur in perimenopausal and postmenopausal women. It is one of the most common carcinomas of the female reproductive system. It has been shown that the occurrence and development of EC is closely associated with the interaction between estrogen (estradiol, E2) and estrogen receptors (ERs), particularly ERα. As a key nuclear transcription factor, ERα is a carcinogenic factor in EC. Its interactions with upstream and downstream effectors and co-regulators have important implications for the proliferation, metastasis, invasion and inhibition of apoptosis of EC. In the present review, the structure of ERα and the regulation of ERα in multiple dimensions are described. In addition, the classical E2/ERα signaling pathway and the crosstalk between ERα and other EC regulators are elucidated, as well as the therapeutic targeting of ERα, which may provide a new direction for clinical applications of ERα in the future.
Collapse
Affiliation(s)
- Yidong Ge
- Department of Medical Oncology, The First Hospital of Ningbo University, Ningbo University, Ningbo, Zhejiang 315020, P.R. China
- Zhejiang Key Laboratory of Pathophysiology, Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Xiaoqi Ni
- Department of Medical Oncology, The First Hospital of Ningbo University, Ningbo University, Ningbo, Zhejiang 315020, P.R. China
- Zhejiang Key Laboratory of Pathophysiology, Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Jingyun Li
- Department of Medical Oncology, The First Hospital of Ningbo University, Ningbo University, Ningbo, Zhejiang 315020, P.R. China
- Zhejiang Key Laboratory of Pathophysiology, Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Meng Ye
- Department of Medical Oncology, The First Hospital of Ningbo University, Ningbo University, Ningbo, Zhejiang 315020, P.R. China
- Zhejiang Key Laboratory of Pathophysiology, Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Xiaofeng Jin
- Department of Medical Oncology, The First Hospital of Ningbo University, Ningbo University, Ningbo, Zhejiang 315020, P.R. China
- Zhejiang Key Laboratory of Pathophysiology, Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| |
Collapse
|
7
|
Pacheco JHL, Elizondo G. Interplay between Estrogen, Kynurenine, and AHR Pathways: An immunosuppressive axis with therapeutic potential for breast cancer treatment. Biochem Pharmacol 2023; 217:115804. [PMID: 37716620 DOI: 10.1016/j.bcp.2023.115804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 09/11/2023] [Accepted: 09/11/2023] [Indexed: 09/18/2023]
Abstract
Breast cancer is one of the most common malignancies among women worldwide. Estrogen exposure via endogenous and exogenous sources during a lifetime, together with environmental exposure to estrogenic compounds, represent the most significant risk factor for breast cancer development. As breast tumors establish, multiple pathways are deregulated. Among them is the aryl hydrocarbon receptor (AHR) signaling pathway. AHR, a ligand-activated transcription factor associated with the metabolism of polycyclic aromatic hydrocarbons and estrogens, is overexpressed in breast cancer. Furthermore, AHR and estrogen receptor (ER) cross-talk pathways have been observed. Additionally, the Tryptophan (Trp) catabolizing enzymes indolamine-2,3-dioxygenase (IDO) and tryptophan-2,3-dioxygenase (TDO) are overexpressed in breast cancer. IDO/TDO catalyzes the formation of Kynurenine (KYN) and other tryptophan-derived metabolites, which are ligands of AHR. Once KYN activates AHR, it stimulates the expression of the IDO enzyme, increases the level of KYN, and activates non-canonical pathways to control inflammation and immunosuppression in breast tumors. The interplay between E2, AHR, and IDO/TDO/KYN pathways and their impact on the immune system represents an immunosuppressive axis on breast cancer. The potential modulation of the immunosuppressive E2-AHR-IDO/TDO/KYN axis has aroused great expectations in oncotherapy. The present article will review the mechanisms implicated in generating the immunosuppressive axis E2-AHR-IDO/TDO/KYN in breast cancer and the current state of knowledge as a potential therapeutic target.
Collapse
Affiliation(s)
| | - Guillermo Elizondo
- Departamento de Biología Celular, CINVESTAV-IPN, Av. IPN 2508, C.P. 07360 Ciudad de México, México.
| |
Collapse
|
8
|
Zhang Y, Hu Y, Yu J, Xie X, Jiang F, Wu C. Landscape of PCOS co-expression gene and its role in predicting prognosis and assisting immunotherapy in endometrial cancer. J Ovarian Res 2023; 16:129. [PMID: 37393293 DOI: 10.1186/s13048-023-01201-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 06/07/2023] [Indexed: 07/03/2023] Open
Abstract
BACKGROUND Endometrial carcinoma (EC) is the sixth most frequent malignancy in women and is often linked to high estrogen exposure. Polycystic ovarian syndrome (PCOS) is a known risk factor for EC, but the underlying mechanisms remain unclear. METHODS We investigated shared gene signals and potential biological pathways to identify effective therapy options for PCOS- and EC-related malignancies. Weighted gene expression network analysis (WGCNA) was used to identify genes associated with PCOS and EC using gene expression data from the Gene Expression Omnibus (GEO) and Cancer Genome Atlas (TCGA) datasets. Enrichment analysis using Cluego software revealed that the steroid hormone biosynthetic process was a critical feature in both PCOS and EC. A predictive signature encompassing genes involved in steroid hormone production was developed using multivariate and least absolute shrinkage and selection operator (LASSO) regression analysis to predict the prognosis of EC. Then, we conducted further experimental verification. RESULTS Patients in the TCGA cohort with high predictive scores had poorer outcomes than those with low scores. We also investigated the relationship between tumor microenvironment (TME) features and predictive risk rating and found that patients with low-risk scores had higher levels of inflammatory and inhibitory immune cells. Also, we found that immunotherapy against anti-CTLA4 and anti-PD-1/PD-L1 was successful in treating individuals with low risk. Low-risk individuals were more responsive to crizotinib therapy, according to further research performed using the "pRRophetic" R package. We further confirmed that IGF2 expression was associated with tumor cell migration, proliferation, and invasion in EC cells. CONCLUTIONS By uncovering the pathways and genes linking PCOS and EC, our findings may provide new therapeutic strategies for patients with PCOS-related EC.
Collapse
Affiliation(s)
- Yun Zhang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, No.300, Guangzhou Road, Nanjing, 210029, China
| | - Yifang Hu
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jian Yu
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, No.300, Guangzhou Road, Nanjing, 210029, China
| | - Xiaoyan Xie
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, No.300, Guangzhou Road, Nanjing, 210029, China
| | - Feng Jiang
- Department of Neonatology, Obstetrics and Gynecology Hospital of Fudan University, No.419, Fangxie Road, Shanghai, 200011, China.
| | - Chuyan Wu
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, No.300, Guangzhou Road, Nanjing, 210029, China.
| |
Collapse
|
9
|
Zang Y, Li H, Liu S, Zhao R, Zhang K, Zang Y, Wang Y, Xue F. The roles and clinical applications of interleukins in endometrial carcinoma. Front Oncol 2022; 12:1001693. [PMID: 36531027 PMCID: PMC9748080 DOI: 10.3389/fonc.2022.1001693] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 11/02/2022] [Indexed: 08/05/2023] Open
Abstract
As a common malignant tumor of the female reproductive system, endometrial carcinoma (EC) seriously endangers women's health with an increasing incidence. The oncogenesis and progression of cancer are closely linked with immune microenvironment, of which interleukins are the important components. In order to illustrate the roles and clinical applications of interleukins in EC, literature of interleukins and EC were reviewed. Based on the present studies, interleukins play crucial roles in the oncogenesis and development of EC via regulating the proliferation, migration, invasion, angiogenesis, apoptosis, pyroptosis and autophagy of EC as well as the immune function against EC. And some of the interleukins seems to have prospective clinical applications in EC, such as evaluating the risk of tumorigenesis, discriminating the malignancy from benign disorders or normal condition, indicating cancer aggressiveness, predicting the prognosis of patients and serving as the novel therapy. However, there is still a long way to go before the clinical applications of interleukins in EC come into reality. Nevertheless, it is certain that the exploration of interleukins will definitely be of great benefit to the screening, diagnosis and treatment of EC in the future.
Collapse
Affiliation(s)
- Yuqin Zang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| | - Huanrong Li
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| | - Shiqi Liu
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| | - Ruqian Zhao
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| | - Kaiwen Zhang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| | - Yuqi Zang
- Hangzhou College of Preschool Teacher Education, Zhejiang Normal University, Hangzhou, China
| | - Yingmei Wang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| | - Fengxia Xue
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
10
|
Lv M, Yu J, Huang Y, Ma J, Xiang J, Wang Y, Li L, Zhang Z, Liao H. Androgen Signaling in Uterine Diseases: New Insights and New Targets. Biomolecules 2022; 12:1624. [PMID: 36358974 PMCID: PMC9687413 DOI: 10.3390/biom12111624] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 10/30/2022] [Accepted: 10/31/2022] [Indexed: 12/06/2023] Open
Abstract
Common uterine diseases include endometriosis, uterine fibroids, endometrial polyps, endometrial hyperplasia, endometrial cancer, and endometrial dysfunction causing infertility. Patients with uterine diseases often suffer from abdominal pain, menorrhagia, infertility and other symptoms, which seriously impair their health and disturb their lives. Androgens play important roles in the normal physiological functions of the uterus and pathological progress of uterine diseases. Androgens in women are synthesized in the ovaries and adrenal glands. The action of androgens in the uterus is mainly mediated by its ligand androgen receptor (AR) that regulates transcription of the target genes. However, much less is known about the signaling pathways through which androgen functions in uterine diseases, and contradictory findings have been reported. This review summarizes and discusses the progress of research on androgens and the involvement of AR in uterine diseases. Future studies should focus on developing new therapeutic strategies that precisely target specific AR and their related signaling pathways in uterine diseases.
Collapse
Affiliation(s)
- Mu Lv
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Juanjuan Yu
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Yan Huang
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, China
| | - Jie Ma
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Jun Xiang
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Yanqiu Wang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Linxia Li
- Department of Obstetrics and Gynecology, Seventh People’s Hospital of Shanghai University of Traditional Chinese Medicine, 358 Datong Road, Shanghai 200137, China
| | - Zhenbo Zhang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China
| | - Hong Liao
- Department of Clinical Laboratory Medicine, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 200040, China
| |
Collapse
|
11
|
Gut and Endometrial Microbiome Dysbiosis: A New Emergent Risk Factor for Endometrial Cancer. J Pers Med 2021; 11:jpm11070659. [PMID: 34357126 PMCID: PMC8304951 DOI: 10.3390/jpm11070659] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/09/2021] [Accepted: 07/12/2021] [Indexed: 12/14/2022] Open
Abstract
Endometrial cancer is one of the most common gynaecological malignancies worldwide. Histologically, two types of endometrial cancer with morphological and molecular differences and also therapeutic implications have been identified. Type I endometrial cancer has an endometrioid morphology and is estrogen-dependent, while Type II appears with non-endometrioid differentiation and follows an estrogen-unrelated pathway. Understanding the molecular biology and genetics of endometrial cancer is crucial for its prognosis and the development of novel therapies for its treatment. However, until now, scant attention has been paid to environmental components like the microbiome. Recently, due to emerging evidence that the uterus is not a sterile cavity, some studies have begun to investigate the composition of the endometrial microbiome and its role in endometrial cancer. In this review, we summarize the current state of this line of investigation, focusing on the relationship between gut and endometrial microbiome and inflammation, estrogen metabolism, and different endometrial cancer therapies.
Collapse
|
12
|
Jeljeli M, Riccio LGC, Chouzenoux S, Moresi F, Toullec L, Doridot L, Nicco C, Bourdon M, Marcellin L, Santulli P, Abrão MS, Chapron C, Batteux F. Macrophage Immune Memory Controls Endometriosis in Mice and Humans. Cell Rep 2021; 33:108325. [PMID: 33147452 DOI: 10.1016/j.celrep.2020.108325] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 07/22/2020] [Accepted: 10/07/2020] [Indexed: 01/08/2023] Open
Abstract
Endometriosis is a frequent, chronic, inflammatory gynecological disease characterized by the presence of ectopic endometrial tissue causing pain and infertility. Macrophages have a central role in lesion establishment and maintenance by driving chronic inflammation and tissue remodeling. Macrophages can be reprogrammed to acquire memory-like characteristics after antigenic challenge to reinforce or inhibit a subsequent immune response, a phenomenon termed "trained immunity." Here, whereas bacille Calmette-Guérin (BCG) training enhances the lesion growth in a mice model of endometriosis, tolerization with repeated low doses of lipopolysaccharide (LPSlow) or adoptive transfer of LPSlow-tolerized macrophages elicits a suppressor effect. LPSlow-tolerized human macrophages mitigate the fibro-inflammatory phenotype of endometriotic cells in an interleukin-10 (IL-10)-dependent manner. A history of severe Gram-negative infection is associated with reduced infertility duration and alleviated symptoms, in contrast to patients with Gram-positive infection history. Thus, the manipulation of innate immune memory may be effective in dampening hyper-inflammatory conditions, opening the way to promising therapeutic approaches.
Collapse
Affiliation(s)
- Mohamed Jeljeli
- Département 3I, Infection, Immunité et Inflammation, Institut Cochin, INSERM U1016, Université de Paris, 75014 Paris, France; Université de Paris, Faculté de Médecine, AP-HP-Centre Université de Paris, Hôpital Cochin, Service d'immunologie biologique, 75014 Paris, France
| | - Luiza G C Riccio
- Département 3I, Infection, Immunité et Inflammation, Institut Cochin, INSERM U1016, Université de Paris, 75014 Paris, France; Disciplina de Ginecologia, Departamento de Obstetrícia e Ginecologia, Faculdade de Medicina FMUSP, Universidade de São Paulo, 01246903 São Paulo, Brasil
| | - Sandrine Chouzenoux
- Département 3I, Infection, Immunité et Inflammation, Institut Cochin, INSERM U1016, Université de Paris, 75014 Paris, France
| | - Fabiana Moresi
- Département 3I, Infection, Immunité et Inflammation, Institut Cochin, INSERM U1016, Université de Paris, 75014 Paris, France
| | - Laurie Toullec
- Département 3I, Infection, Immunité et Inflammation, Institut Cochin, INSERM U1016, Université de Paris, 75014 Paris, France
| | - Ludivine Doridot
- Département 3I, Infection, Immunité et Inflammation, Institut Cochin, INSERM U1016, Université de Paris, 75014 Paris, France
| | - Carole Nicco
- Département 3I, Infection, Immunité et Inflammation, Institut Cochin, INSERM U1016, Université de Paris, 75014 Paris, France
| | - Mathilde Bourdon
- Département 3I, Infection, Immunité et Inflammation, Institut Cochin, INSERM U1016, Université de Paris, 75014 Paris, France; Université de Paris, Faculté de Médecine, AP-HP-Centre Université de Paris, Hôpital Cochin, Département de Gynécologie Obstétrique II et Médecine de la Reproduction, 75014 Paris, France
| | - Louis Marcellin
- Département 3I, Infection, Immunité et Inflammation, Institut Cochin, INSERM U1016, Université de Paris, 75014 Paris, France; Université de Paris, Faculté de Médecine, AP-HP-Centre Université de Paris, Hôpital Cochin, Département de Gynécologie Obstétrique II et Médecine de la Reproduction, 75014 Paris, France
| | - Pietro Santulli
- Département 3I, Infection, Immunité et Inflammation, Institut Cochin, INSERM U1016, Université de Paris, 75014 Paris, France; Université de Paris, Faculté de Médecine, AP-HP-Centre Université de Paris, Hôpital Cochin, Département de Gynécologie Obstétrique II et Médecine de la Reproduction, 75014 Paris, France
| | - Mauricio S Abrão
- Disciplina de Ginecologia, Departamento de Obstetrícia e Ginecologia, Faculdade de Medicina FMUSP, Universidade de São Paulo, 01246903 São Paulo, Brasil
| | - Charles Chapron
- Département 3I, Infection, Immunité et Inflammation, Institut Cochin, INSERM U1016, Université de Paris, 75014 Paris, France; Université de Paris, Faculté de Médecine, AP-HP-Centre Université de Paris, Hôpital Cochin, Département de Gynécologie Obstétrique II et Médecine de la Reproduction, 75014 Paris, France
| | - Frédéric Batteux
- Département 3I, Infection, Immunité et Inflammation, Institut Cochin, INSERM U1016, Université de Paris, 75014 Paris, France; Université de Paris, Faculté de Médecine, AP-HP-Centre Université de Paris, Hôpital Cochin, Service d'immunologie biologique, 75014 Paris, France.
| |
Collapse
|
13
|
Fan Q, Huang T, Sun X, Wang YW, Wang J, Liu Y, Ni T, Gu SL, Li YH, Wang YD. HPV-16/18 E6-induced APOBEC3B expression associates with proliferation of cervical cancer cells and hypomethylation of Cyclin D1. Mol Carcinog 2021; 60:313-330. [PMID: 33631046 DOI: 10.1002/mc.23292] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 02/14/2021] [Accepted: 02/15/2021] [Indexed: 12/13/2022]
Abstract
Oncogenic high-risk human papillomavirus (HR-HPV) infection causes a majority of cases of cervical cancer and pre-cancerous cervical lesions. However, the mechanisms underlying the direct evolution from HPV-16/18-infected epithelium to cervical intraepithelial neoplasia (CIN) III, which can progress to cervical cancer, remain poorly identified. Here, we performed RNA-seq after laser capture microdissection, and found that APOBEC3B was highly expressed in cervical cancer specimens compared with CIN III with HPV-16/18 infection. Furthermore, immunohistochemical analysis confirmed that high levels of APOBEC3B were correlated with lymph node metastasis in cervical cancer. Subsequent experiments revealed that HPV-16 E6 could upregulate APOBEC3B through direct binding to the promoter of APOBEC3B in cervical cancer cells. Silencing of APOBEC3B by stable short hairpin RNA-mediated knockdown reduced the proliferative capacity of Caski and HeLa cells in vitro and in vivo, but had only a small effect on the migration and invasion of two cervical cancer cell lines. Finally, we identified the changes in gene expression following APOBEC3B silencing in Caski cells by microarray, demonstrating a biological link between APOBEC3B and CCND1 in cervical cancer cells. Importantly, through methyl-capture sequencing and pyrosequencing, APOBEC3B was found to affect the levels of the downstream protein Cyclin D1 (which is encoded by the CCND1 gene) through hypomethylation of the CCND1 promoter. In conclusion, our study supports HPV-16 E6-induced APOBEC3B expression associates with proliferation of cervical cancer cells and hypomethylation of Cyclin D1. Thus, APOBEC3B may be a potential therapeutic target in human cervical cancer.
Collapse
Affiliation(s)
- Qiong Fan
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Municipal Key Clinical Specialty, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Ting Huang
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Municipal Key Clinical Specialty, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Xiao Sun
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Municipal Key Clinical Specialty, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Yi-Wei Wang
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jing Wang
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yao Liu
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ting Ni
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Sheng-Lan Gu
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yu-Hong Li
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yu-Dong Wang
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Municipal Key Clinical Specialty, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| |
Collapse
|
14
|
Cheng R, Xue X, Liu X. Expression of IL17A in endometrial carcinoma and effects of IL17A on biological behaviour in Ishikawa cells. J Int Med Res 2020; 48:300060520950563. [PMID: 32878530 PMCID: PMC7780559 DOI: 10.1177/0300060520950563] [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] [Indexed: 11/26/2022] Open
Abstract
Objective A growing body of evidence suggests chronic inflammation triggers the process of endometrial carcinogenesis. Interleukin (IL) 17A is an important proinflammatory factor involved in the tumour angiogenesis processes of many solid tumours. This study aimed to characterize the function of IL17A in endometrioid-type endometrial carcinoma. Methods Levels of IL17A in human endometrial tissues were analysed by immunohistochemistry. In vitro proliferation and migration were analysed in Ishikawa cells treated with IL17A, using cell counting kit-8, wound healing and transwell assays. Western blots were used to analyse levels of oestrogen receptor (ER)α and ERβ proteins in Ishikawa cells treated with IL17A. Results IL17A levels were significantly higher in endometrial carcinoma tissues than in endometrial hyperplasic tissues. Significantly increased proliferation and migration was observed in Ishikawa cells treated with IL17A versus controls. Investigation of the molecular mechanism revealed that IL17A treatment upregulated the ERα/ERβ protein ratio in Ishikawa cells. Conclusions IL17A may be an important proinflammatory factor involved in promoting endometrial carcinogenesis.
Collapse
Affiliation(s)
- Ran Cheng
- Department of Gynaecology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoou Xue
- Department of Gynaecology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoli Liu
- Department of Gynaecology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
15
|
Chen Z, Yang HJ, Lin Q, Zhu MJ, Yu YY, He XY, Wan XP. Estrogen-ERα signaling and DNA hypomethylation co-regulate expression of stem cell protein PIWIL1 in ERα-positive endometrial cancer cells. Cell Commun Signal 2020; 18:84. [PMID: 32503542 PMCID: PMC7275358 DOI: 10.1186/s12964-020-00563-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 03/23/2020] [Indexed: 12/19/2022] Open
Abstract
Background We previously identified PIWIL1 as an oncogene involved in endometrial carcinogenesis. However, the mechanism of Piwil1 mediated regulation of tumorigenesis remains poorly understood. Methods The expression levels of target genes in endometrial cancer cells were detected by quantitative reverse transcription-PCR (RT-qPCR) and western blotting. Up- or down-regulation of ERα or PIWIL1 was achieved by transient transfection with expressing plasmids or short hairpin RNA (shRNA). Dual-luciferase reporter assays and chromatin immunoprecipitation (ChIP) were used to demonstrate the ERα bound to the half estrogen response element (half-ERE) located in PIWIL1 promoter. The expression of PIWIL1 and ERα in endometrial carcinoma tissues were investigated using immunohistochemistry and RT-qPCR. The proliferation ability of cancer cells were evaluated by MTT. Methylation status of the PIWIL1 promoter was detected by bisulfite sequencing PCR (BSP). Results In the present study, we found that PIWIL1 mediated E2-stimulated cancer cell proliferation. In ERα-positive endometrial cancer cells, we demonstrated that estrogen-ERα signaling significantly up-regulated the expression of PIWIL1, which was mediated by binding of the ERα onto the PIWIL1 promoter. Furthermore, we found that a half-ERE in the PIWIL1 promoter was essential for ERα binding. The PIWIL1 promoter was hypomethylated in ERα-positive endometrial cancer cells. Treatment with 5-aza-deoxycytidine (5-aza-dC) could up-regulate PIWIL1 expression. Conclusions These findings uncover a novel molecular mechanism by which estrogen-ERα signaling and DNA hypomethylation co-regulate PIWIL1 expression. These findings provide novel insights into the hormonal regulation of PIWIL1 in endometrial cancer and the PIWIL1’s role in estrogen-stimulated endometrial carcinogenesis. Video Abstract. (MP4 41319 kb)
Collapse
Affiliation(s)
- Zheng Chen
- Department of Obstetrics and Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, No.910, Hengshan Road, Shanghai, 200030, China.,Shanghai Municipal Key Clinical Specialty, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Hua-Jing Yang
- Department of Obstetrics and Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, No.910, Hengshan Road, Shanghai, 200030, China.,Shanghai Municipal Key Clinical Specialty, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Qin Lin
- Department of Obstetrics and Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, No.910, Hengshan Road, Shanghai, 200030, China.,Shanghai Municipal Key Clinical Specialty, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Min-Jiao Zhu
- Department of Obstetrics and Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, No.910, Hengshan Road, Shanghai, 200030, China.,Shanghai Municipal Key Clinical Specialty, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Ying-Ying Yu
- Department of Obstetrics and Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, No.910, Hengshan Road, Shanghai, 200030, China.,Shanghai Municipal Key Clinical Specialty, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Xiao-Ying He
- Department of Obstetrics and Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, No.910, Hengshan Road, Shanghai, 200030, China. .,Shanghai Municipal Key Clinical Specialty, Shanghai, China. .,Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China.
| | - Xiao-Ping Wan
- Department of Obstetrics and Gynecology, Shanghai First Maternity and Infant Hospital, Tong Ji University School of Medicine, No. 536, Changle Road, Shanghai, 200080, China.
| |
Collapse
|
16
|
Ulhaq ZS, Soraya GV, Zambrano LEA, Garcia CP. Sexual dimorphism in SARS-COV-2 infection. ACTA ENDOCRINOLOGICA-BUCHAREST 2020; 16:522-523. [PMID: 34084248 DOI: 10.4183/aeb.2020.522] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Previous studies have demonstrated that female patients with coronavirus disease 2019 (COVID-19) demonstrate more favorable prognosis relative to male patients. In this article, we elaborate the possible role of estrogen in the modulation of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection severity. The potential interplay between several factors, including inherently lower estradiol (E2) and slightly higher estrogen receptor β (ERβ) levels in males, with inflammatory mediators are described. Altogether, there seems to be a sexually dimorphic response towards SARS-CoV-2 infection, and a possibility that COVID-19 severity is dependent on both E2 levels and ERα:ERβ expression ratio in lymphoid and lung cells.
Collapse
Affiliation(s)
- Z S Ulhaq
- Maulana Malik Ibrahim State Islamic University of Malang - Faculty of Medicine and Health Sciences, Malang, East Java, Indonesia, United Kingdom of Great Britain and Northern Ireland
| | - G V Soraya
- Hasanuddin University - Faculty of Medicine, Makassar, South Sulawesi, Indonesia, United Kingdom of Great Britain and Northern Ireland
| | - L E A Zambrano
- Technical University of Manabi, Faculty of Health Sciences, Portoviejo, Manabí, Ecuador, United Kingdom of Great Britain and Northern Ireland
| | - C P Garcia
- Weston General Hospital, Weston-super-Mare, North Somerset, United Kingdom of Great Britain and Northern Ireland
| |
Collapse
|
17
|
Yang X, Wang J. The Role of Metabolic Syndrome in Endometrial Cancer: A Review. Front Oncol 2019; 9:744. [PMID: 31440472 PMCID: PMC6694738 DOI: 10.3389/fonc.2019.00744] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 07/24/2019] [Indexed: 12/12/2022] Open
Abstract
Endometrial cancer is one of the most common cancers of the female reproductive system. Although surgery, radiotherapy, chemotherapy, and hormone therapy can significantly improve the survival of patients, the treatment of patients with very early lesions and a strong desire to retain reproductive function or late recurrence is still in the early stages. Metabolic syndrome (MS) is a clustering of at least three of the five following medical conditions: central obesity, high blood pressure, high blood sugar, high serum triglycerides, and low serum high-density lipoprotein (HDL). Obesity, diabetes and hypertension often coexist in patients with endometrial cancer, which increases the risk of endometrial cancer, also known as the "triple syndrome of endometrial cancer." In recent years, epidemiological and clinical studies have found that MS associated with metabolic diseases is closely related to the incidence of endometrial cancer. However, the key molecular mechanisms underlying the induction of endometrial cancer by MS have not been elucidated to date. Characterizing the tumor metabolism microenvironment will be advantageous for achieving a comprehensive view of the molecular mechanism of metabolic syndrome associated with endometrial cancer and for providing a new target for the treatment of endometrial cancer. This review focuses on recent advances in determining the role of metabolic syndrome-related factors and mechanisms in the pathogenesis of endometrial cancer. We suggest that interfering with the tumor metabolic microenvironment-related molecular signals may inhibit the occurrence of endometrial cancer.
Collapse
Affiliation(s)
- Xiao Yang
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, China
| | - Jianliu Wang
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, China
| |
Collapse
|
18
|
Shiba S, Ikeda K, Suzuki T, Shintani D, Okamoto K, Horie-Inoue K, Hasegawa K, Inoue S. Hormonal Regulation of Patient-Derived Endometrial Cancer Stem-like Cells Generated by Three-Dimensional Culture. Endocrinology 2019; 160:1895-1906. [PMID: 31265065 DOI: 10.1210/en.2019-00362] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Accepted: 06/24/2019] [Indexed: 12/27/2022]
Abstract
Low-grade and early-stage endometrial cancer usually has a favorable prognosis, whereas recurrent or metastatic disease is often difficult to cure. Thus, the molecular mechanisms underlying advanced pathophysiology remain to be elucidated. From the perspective of the origin of advanced endometrial cancer, the characterization of cancer stem-like cells (CSCs) will be the first step toward the development of clinical management. We established long-term culturable patient-derived cancer cells (PDCs) from patient endometrial tumors by spheroid cell culture, which is favorable for the enrichment of CSCs. PDC-derived xenograft tumors were generated in immunodeficient NOD/Shi-scid, IL-2RγKO Jic mice. Morphologically, PDCs derived from three distinct patient samples and their xenograft tumors recapitulated the corresponding original patient tumors. Of note, CSC-related genes including ALDH1A1 were upregulated in all of these PDCs, and the therapeutic potentiality of aldehyde dehydrogenase inhibitors was demonstrated. In addition, these PDCs and their patient-derived xenograft (PDX) models exhibited distinct characteristics on the basis of their hormone responsiveness and metastatic features. Interestingly, genes associated with inflammation and tumor immunity were upregulated by 17β-estradiol in PDC lines with high estrogen receptor expression and were also overexpressed in secondary PDCs obtained from metastatic tumor models. These results suggest that PDC and PDX models from endometrial cancer specimens would be useful to elucidate CSC traits and to develop alternative diagnostic and therapeutic options for advanced disease.
Collapse
Affiliation(s)
- Sachiko Shiba
- Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan
| | - Kazuhiro Ikeda
- Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan
| | - Takashi Suzuki
- Department of Pathology and Histotechnology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Daisuke Shintani
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Saitama, Japan
| | - Koji Okamoto
- Division of Cancer Differentiation, National Cancer Center Research Institute, Tokyo, Japan
| | - Kuniko Horie-Inoue
- Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan
| | - Kosei Hasegawa
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Saitama, Japan
| | - Satoshi Inoue
- Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan
- Department of Functional Biogerontology, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| |
Collapse
|
19
|
Keshari S, Sipayung AD, Hsieh CC, Su LJ, Chiang YR, Chang HC, Yang WC, Chuang TH, Chen CL, Huang CM. IL-6/p-BTK/p-ERK signaling mediates calcium phosphate-induced pruritus. FASEB J 2019; 33:12036-12046. [PMID: 31365830 DOI: 10.1096/fj.201900016rr] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Uremic pruritus with elevated levels of calcium phosphate (CaP) in skin is a common symptom in patients with chronic kidney disease (CKD). In this study, we demonstrate that intradermal injection of CaP into mice triggered scratching by up-regulating the IL-6 in skin and phosphorylation of ERKs in dorsal root ganglion (DRG) in a dose-dependent manner. IL-6 is essential because the CaP-induced up-regulation of phosphorylated (p)-ERK in DRG was considerably reduced in the IL-6 knockout mice. Microarray analysis in conjunction with real-time PCR revealed a higher mRNA expression of Bruton's tyrosine kinase (BTK) gene in DRG after CaP injection. The inhibition of BTK by ibrutinib noticeably diminish the CaP-induced up-regulation of IL-6 and p-ERK in mice. A high amount of IL-6 was detected in itchy skin and blood of patients with CKD. The expressions of p-BTK and p-ERK in DRG primary cells reached maximum levels at 1 and 10 min, respectively, after treatment of recombinant IL-6 and were significantly reduced by treatment of IL-6 along with ibrutinib. The mechanism by which the CaP-induced pruritus mediated by the IL-6/p-BTK/p-ERK signaling was revealed.-Keshari, S., Sipayung, A. D., Hsieh, C.-C., Su, L.-J., Chiang, Y.-R., Chang, H.-C., Yang, W.-C., Chuang, T.-H., Chen, C.-L., Huang, C.-M. IL-6/p-BTK/p-ERK signaling mediates calcium phosphate-induced pruritus.
Collapse
Affiliation(s)
- Sunita Keshari
- Department of Life Sciences, National Central University, Taoyuan, Taiwan
| | | | - Ching-Chuan Hsieh
- Division of General Surgery, Department of Surgery, Chang Gung Memorial Hospital, Chia-Yi, Taiwan
| | - Li-Jen Su
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan, Taiwan
| | - Yun-Ru Chiang
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan, Taiwan
| | | | - Wu-Chang Yang
- Division of Nephrology, Landseed Hospital, Taoyuan, Taiwan
| | - Tsung-Hsien Chuang
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan
| | - Chien-Lung Chen
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan, Taiwan.,Division of Nephrology, Landseed Hospital, Taoyuan, Taiwan
| | - Chun-Ming Huang
- Department of Life Sciences, National Central University, Taoyuan, Taiwan.,Department of Biomedical Sciences and Engineering, National Central University, Taoyuan, Taiwan.,Department of Dermatology, University of California-San Diego, San Diego, California, USA
| |
Collapse
|
20
|
Che Q, Xiao X, Xu J, Liu M, Lu Y, Liu S, Dong X. 17β-Estradiol promotes endometrial cancer proliferation and invasion through IL-6 pathway. Endocr Connect 2019; 8:961-968. [PMID: 31189128 PMCID: PMC6612063 DOI: 10.1530/ec-19-0258] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 05/29/2019] [Indexed: 12/20/2022]
Abstract
Accumulating evidence revealed that the leading risk factor of endometrial cancer is exposure to endogenous and exogenous estrogens, while the exact mechanism underlying estrogen contribution to endometrial cancer progression has not been elucidated clearly. Interleukin (IL)-6 has been verified to be critical for tumor progression in several human cancers. In this study, we provided evidence that 17β-estradiol (E2) could significantly promote endometrial cancer cells viability, migration and invasion through activation of IL-6 pathway, which involved in its downstream pathway and target genes (p-Stat3, Bcl-2, Mcl-1, cyclin D1 and MMP2). Meanwhile, utilization of IL-6-neutralizing antibody could partially attenuate the increased cancer growth and invasion abilities in Ishikawa and RL95-2 endometrial cancer cell lines and an orthotopic endometrial cancer model. We established a causative link between estrogen and IL-6 signaling activation in the development of endometrial cancer. The molecular mechanism defined in this study provided the evidence that E2 promotes endometrial carcinoma progression via activating the IL-6 pathway, indicating that interruption of IL-6 might be an essential therapeutic strategy in estrogen-dependent endometrial cancer.
Collapse
Affiliation(s)
- Qi Che
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xirong Xiao
- Department of Obstetrics, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Jun Xu
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Miao Liu
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yongning Lu
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Suying Liu
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Correspondence should be addressed to S Liu or X Dong: or
| | - Xi Dong
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Correspondence should be addressed to S Liu or X Dong: or
| |
Collapse
|
21
|
Che Q, Xiao X, Liu M, Lu Y, Dong X, Liu S. IL-6 promotes endometrial cancer cells invasion and migration through signal transducers and activators of transcription 3 signaling pathway. Pathol Res Pract 2019; 215:152392. [PMID: 30922625 DOI: 10.1016/j.prp.2019.03.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 03/03/2019] [Accepted: 03/18/2019] [Indexed: 12/12/2022]
Abstract
Interleukin (IL)-6 is the most well-known traditional activator of activating signal transducers and activators of transcription 3 (Stat3). They have been proved to promote cancer progression in several human cancers. However, their exact roles in endometrial cancer have not been elucidated clearly. In this study, we aimed to investigate the role of IL-6/Stat3 signaling pathway in human endometrial cancer cells invasion and migration. We demonstrated that Stat3 is activated in endometrial cancer cell lines. To investigate the role of Stat3 in endometrial cancer invasive capacity, we used Stat3 inhibitor Stattic and found that Stattic significantly inhibited the migration and invasion of endometrial cancer cells elevated by IL-6. Furthermore, we showed that Stat3 inhibitor significantly decreased the expression of MMP2 enhanced by IL-6, indicating that IL-6 promoted endometrial cancer invasion and migration by Stat3-induced MMP2 upregulation. Taken together, our findings indicate that targeting IL-6/Stat3 pathway might be a potentially effective therapeutic strategy for treating endometrial cancer.
Collapse
Affiliation(s)
- Qi Che
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xirong Xiao
- Department of Obstetrics, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
| | - Miao Liu
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yongning Lu
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xi Dong
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| | - Suying Liu
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| |
Collapse
|
22
|
Cai J, Cui K, Niu F, Jin T, Huang S, Zhang Y, Bao S. Genetics of IL6 polymorphisms: Case-control study of the risk of endometrial cancer. Mol Genet Genomic Med 2019; 7:e00600. [PMID: 30828987 PMCID: PMC6465666 DOI: 10.1002/mgg3.600] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 01/03/2019] [Accepted: 01/06/2019] [Indexed: 02/06/2023] Open
Abstract
Background Endometrial cancer is the most common gynaecological malignancy. Cytokines gene may be important in endometrial cancer development. This study sought to investigate whether the IL4, IL6 two gene genetic variants were associated with susceptibility to endometrial cancer (EC) in Hainan Chinese Han women by a hospital‐based study. Methods The genetic polymorphisms for IL4 and IL6 were analyzed by Agena MassARRAY method. Odds ratios (ORs) and 95% confidence intervals (CIs) were calculated by unconditional logistic regression. Results We observed a significant increase in risk of endometrial cancer of rs1524107 (IL6) (T/C, OR = 1.61, 95% CI = 1.09–2.37, p = 1.55 × 10−2), rs2066992 (IL 6) (OR = 3.09, 95% CI = 2.11–4.53, p = 3.13 × 10−9). However, for IL4 gene, no associations emerged the SNP and EC risk. Conclusion This study demonstrated that IL6 gene polymorphisms are significantly associated with increased EC susceptibility in Hainan Chinese Han women.
Collapse
Affiliation(s)
- Junhong Cai
- Key Laboratory of Cell and Molecular Genetic Translational Medicine in Hainan ProvinceHainanChina
| | - Kaiying Cui
- Department of Gynaecology and ObstetricsHainan General HospitalHainanChina
| | - Fanglin Niu
- Key Laboratory of Resource Biology and Biotechnology in Western China (Northwest University)Ministry of EducationXi’an, ShaanxiChina
- School of Life SciencesNorthwest UniversityXi’an, ShaanxiChina
| | - Tianbo Jin
- Key Laboratory of Resource Biology and Biotechnology in Western China (Northwest University)Ministry of EducationXi’an, ShaanxiChina
- School of Life SciencesNorthwest UniversityXi’an, ShaanxiChina
| | - Sizhe Huang
- Department of Gynaecology and ObstetricsHainan General HospitalHainanChina
| | - Ying Zhang
- Department of Gynaecology and ObstetricsHainan General HospitalHainanChina
| | - Shan Bao
- Department of Gynaecology and ObstetricsHainan General HospitalHainanChina
| |
Collapse
|
23
|
Lv Q, Xie L, Cheng Y, Shi Y, Shan W, Ning C, Xie B, Yang B, Luo X, He Q, Zhu Q, Zhang Y, Zhang Z, Wang C, Chen X, Xu C. A20-mediated deubiquitination of ERα in the microenvironment of CD163+ macrophages sensitizes endometrial cancer cells to estrogen. Cancer Lett 2019; 442:137-147. [DOI: 10.1016/j.canlet.2018.10.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 09/25/2018] [Accepted: 10/19/2018] [Indexed: 02/04/2023]
|
24
|
The Emerging Role of the Microenvironment in Endometrial Cancer. Cancers (Basel) 2018; 10:cancers10110408. [PMID: 30380719 PMCID: PMC6266917 DOI: 10.3390/cancers10110408] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 10/23/2018] [Accepted: 10/24/2018] [Indexed: 12/24/2022] Open
Abstract
Endometrial cancer (EC) is one of the most frequently diagnosed cancers in women, and despite recent therapeutic advances, in many cases, treatment failure results in cancer recurrence, metastasis, and death. Current research demonstrates that the interactive crosstalk between two discrete cell types (tumor and stroma) promotes tumor growth and investigations have uncovered the dual role of the stromal cells in the normal and cancerous state. In contrast to tumor cells, stromal cells within the tumor microenvironment (TME) are genetically stable. However, tumor cells modify adjacent stromal cells in the TME. The alteration in signaling cascades of TME from anti-tumorigenic to pro-tumorigenic enhances metastatic potential and/or confers therapeutic resistance. Therefore, the TME is a fertile ground for the development of novel therapies. Furthermore, disrupting cancer-promoting signals from the TME or re-educating stromal cells may be an effective strategy to impair metastatic progression. Here, we review the paradoxical role of different non-neoplastic stromal cells during specific stages of EC progression. We also suggest that the inhibition of microenvironment-derived signals may suppress metastatic EC progression and offer novel potential therapeutic interventions.
Collapse
|
25
|
Konings GF, Saarinen N, Delvoux B, Kooreman L, Koskimies P, Krakstad C, Fasmer KE, Haldorsen IS, Zaffagnini A, Häkkinen MR, Auriola S, Dubois L, Lieuwes N, Verhaegen F, Schyns LE, Kruitwagen RF, Xanthoulea S, Romano A. Development of an Image-Guided Orthotopic Xenograft Mouse Model of Endometrial Cancer with Controllable Estrogen Exposure. Int J Mol Sci 2018; 19:ijms19092547. [PMID: 30154339 PMCID: PMC6165149 DOI: 10.3390/ijms19092547] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 08/19/2018] [Accepted: 08/22/2018] [Indexed: 02/08/2023] Open
Abstract
Endometrial cancer (EC) is the most common gynaecological malignancy in Western society and the majority of cases are estrogen dependent. While endocrine drugs proved to be of insufficient therapeutic value in the past, recent clinical research shows promising results by using combinational regimens and pre-clinical studies and identified potential novel endocrine targets. Relevant pre-clinical models can accelerate research in this area. In the present study we describe an orthotopic and estrogen dependent xenograft mouse model of EC. Tumours were induced in one uterine horn of female athymic nude mice using the well-differentiated human endometrial adenocarcinoma Ishikawa cell line—modified to express the luciferase gene for bioluminescence imaging (BLI). BLI and contrast-enhanced computed-tomograph (CE-CT) were used to measure non-invasive tumour growth. Controlled estrogen exposure was achieved by the use of MedRod implants releasing 1.5 μg/d of 17β-estradiol (E2) in ovariectomized mice. Stable E2 serum concentration was demonstrated by LC-MS/MS. Induced tumours were E2 responsive as increased tumour growth was observed in the presence of E2 but not placebo, assessed by BLI, CE-CT, and tumour weight at sacrifice. Metastatic spread was assessed macroscopically by BLI and histology and was seen in the peritoneal cavity, in the lymphovascular space, and in the thoracic cavity. In conclusion, we developed an orthotopic xenograft mouse model of EC that exhibits the most relevant features of human disease, regarding metastatic spread and estrogen dependency. This model offers an easy to manipulate estrogen dosage (by simply adjusting the MedRod implant length), image-guided monitoring of tumour growth, and objectively measurable endpoints (including tumour weight). This is an excellent in vivo tool to further explore endocrine drug regimens and novel endocrine drug targets for EC.
Collapse
Affiliation(s)
- Gonda Fj Konings
- GROW-School for Oncology & Developmental Biology, Maastricht University, 6229HX Maastricht, The Netherlands.
- Department of Obstetrics and Gynaecology, Maastricht University Medical Centre, P. Debyelaan 25, 6229HX Maastricht, The Netherlands.
| | - Niina Saarinen
- Forendo Pharma Ltd., FI-20520 Turku, Finland.
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology and Turku Center for Disease Modeling (TCDM), University of Turku, FI-20520 Turku, Finland.
| | - Bert Delvoux
- GROW-School for Oncology & Developmental Biology, Maastricht University, 6229HX Maastricht, The Netherlands.
- Department of Obstetrics and Gynaecology, Maastricht University Medical Centre, P. Debyelaan 25, 6229HX Maastricht, The Netherlands.
| | - Loes Kooreman
- GROW-School for Oncology & Developmental Biology, Maastricht University, 6229HX Maastricht, The Netherlands.
- Department of Pathology, Maastricht University Medical Centre, 6229HX Maastricht, The Netherlands.
| | | | - Camilla Krakstad
- Department of Obstetrics and Gynaecology, Haukeland University Hospital, 5021 Bergen, Norway.
- Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, 5021 Bergen, Norway.
| | - Kristine E Fasmer
- Department of Radiology, Haukeland University Hospital, 5021 Bergen, Norway.
- Section for Radiology, Department of Clinical Medicine, University of Bergen, 5020 Bergen, Norway.
| | - Ingfrid S Haldorsen
- Department of Radiology, Haukeland University Hospital, 5021 Bergen, Norway.
- Section for Radiology, Department of Clinical Medicine, University of Bergen, 5020 Bergen, Norway.
| | - Amina Zaffagnini
- GROW-School for Oncology & Developmental Biology, Maastricht University, 6229HX Maastricht, The Netherlands.
- Department of Obstetrics and Gynaecology, Maastricht University Medical Centre, P. Debyelaan 25, 6229HX Maastricht, The Netherlands.
| | - Merja R Häkkinen
- School of Pharmacy, University of Eastern Finland, FI-80101 Kuopio, Finland.
| | - Seppo Auriola
- School of Pharmacy, University of Eastern Finland, FI-80101 Kuopio, Finland.
| | - Ludwig Dubois
- GROW-School for Oncology & Developmental Biology, Maastricht University, 6229HX Maastricht, The Netherlands.
- Department of Radiotherapy (MAASTRO), Maastricht University, 6229HX Maastricht, The Netherlands.
| | - Natasja Lieuwes
- GROW-School for Oncology & Developmental Biology, Maastricht University, 6229HX Maastricht, The Netherlands.
- Department of Radiotherapy (MAASTRO), Maastricht University, 6229HX Maastricht, The Netherlands.
| | - Frank Verhaegen
- GROW-School for Oncology & Developmental Biology, Maastricht University, 6229HX Maastricht, The Netherlands.
- Department of Radiotherapy (MAASTRO), Maastricht University, 6229HX Maastricht, The Netherlands.
| | - Lotte Ejr Schyns
- GROW-School for Oncology & Developmental Biology, Maastricht University, 6229HX Maastricht, The Netherlands.
- Department of Radiotherapy (MAASTRO), Maastricht University, 6229HX Maastricht, The Netherlands.
| | - Roy Fpm Kruitwagen
- GROW-School for Oncology & Developmental Biology, Maastricht University, 6229HX Maastricht, The Netherlands.
- Department of Obstetrics and Gynaecology, Maastricht University Medical Centre, P. Debyelaan 25, 6229HX Maastricht, The Netherlands.
| | - Sofia Xanthoulea
- GROW-School for Oncology & Developmental Biology, Maastricht University, 6229HX Maastricht, The Netherlands.
- Department of Obstetrics and Gynaecology, Maastricht University Medical Centre, P. Debyelaan 25, 6229HX Maastricht, The Netherlands.
| | - Andrea Romano
- GROW-School for Oncology & Developmental Biology, Maastricht University, 6229HX Maastricht, The Netherlands.
- Department of Obstetrics and Gynaecology, Maastricht University Medical Centre, P. Debyelaan 25, 6229HX Maastricht, The Netherlands.
| |
Collapse
|
26
|
Ke J, Shen Z, Li M, Peng C, Xu P, Wang M, Zhu Y, Zhang X, Wu D. Prostaglandin E2 triggers cytochrome P450 17α hydroxylase overexpression via signal transducer and activator of transcription 3 phosphorylation and promotes invasion in endometrial cancer. Oncol Lett 2018; 16:4577-4585. [PMID: 30214592 DOI: 10.3892/ol.2018.9165] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 09/22/2017] [Indexed: 12/28/2022] Open
Abstract
Prostaglandin E2 (PGE2) is the most common prostaglandin in the human body, meaning that its malfunction impacts on the development of numerous diseases. Prostaglandin E synthase 2 (PTGES2) is involved in the synthesis of PGE2. In the present study, immunohistochemistry of PTGES2 was performed in 152 patients with endometrial cancer and in 66 patients with normal endometria. The results indicate a notable association among increased expression of PTGES2 and age (P=0.0092) and the depth of myometrial invasion (P<0.0001). Reverse transcription-quantitative polymerase chain reaction and western blot analysis demonstrated that cytochrome P450 17α hydroxylase (CYP17), an enzyme for androgen synthesis, is overexpressed following PGE2 stimulation via signal transducer and activator of transcription 3 (STAT3) phosphorylation. ELISA also detected increased androgen (testosterone) secretion. Further invasion of endometrial cancer cells was induced at high androgen levels and when CYP17 was overexpressed. Furthermore, the present study observed that CYP17 is overexpressed via STAT3 phosphorylation in endometrial cancer cells, which grow at a high concentration of PGE2, resulting in increased androgen secretion. Concentrations of estrogen and progesterone were not elevated, while the concentration of androgens was. Overall, a high concentration of androgens caused increased invasion of endometrial cancer cells. A high concentration of androgens, which is initiated by a high expression of PTGES2 and a high concentration of PGE2, is an important promoter of myometrial invasion in endometrial cancer.
Collapse
Affiliation(s)
- Jieqi Ke
- Department of Obstetrics and Gynecology, Anhui Provincial Hospital, Hefei, Anhui 230001, P.R. China
| | - Zhen Shen
- Department of Obstetrics and Gynecology, Anhui Provincial Hospital, Hefei, Anhui 230001, P.R. China
| | - Min Li
- Department of Obstetrics and Gynecology, Anhui Provincial Hospital, Hefei, Anhui 230001, P.R. China
| | - Cheng Peng
- Department of Obstetrics and Gynecology, Anhui Provincial Hospital, Hefei, Anhui 230001, P.R. China
| | - Ping Xu
- Department of Obstetrics and Gynecology, Anhui Provincial Hospital, Hefei, Anhui 230001, P.R. China
| | - Meimei Wang
- Department of Obstetrics and Gynecology, Anhui Provincial Hospital, Hefei, Anhui 230001, P.R. China
| | - Yi Zhu
- Department of Obstetrics and Gynecology, Anhui Provincial Hospital, Hefei, Anhui 230001, P.R. China
| | - Xuefen Zhang
- Department of Obstetrics and Gynecology, Anhui Provincial Hospital, Hefei, Anhui 230001, P.R. China
| | - Dabao Wu
- Department of Obstetrics and Gynecology, Anhui Provincial Hospital, Hefei, Anhui 230001, P.R. China
| |
Collapse
|
27
|
Huang Q, Zhang Z, Liao Y, Liu C, Fan S, Wei X, Ai B, Xiong J. 17β-estradiol upregulates IL6 expression through the ERβ pathway to promote lung adenocarcinoma progression. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:133. [PMID: 29970138 PMCID: PMC6029357 DOI: 10.1186/s13046-018-0804-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Accepted: 06/21/2018] [Indexed: 01/06/2023]
Abstract
Background In non-small cell lung cancer (NSCLC), estrogen (E2) significantly promotes NSCLC cell growth via estrogen receptor beta (ERβ). Discovery and elucidation of the mechanism underlying estrogen-promoted NSCLC progression is critical for effective preventive interventions. IL6 has been demonstrated to be involved in the development, progression and metastasis in several cancers and IL6 overexpression is associated with poor prognosis in NSCLC. However, the exact role played by IL6 in estrogen-promoted NSCLC progress remain unknown. Here, we evaluated the expression and biological effects of IL6 in NSCLC cells when treated with E2 and explored the underlying mechanism of IL6 in E2-promoted NSCLC progression. Methods Expression of ERβ/IL6 in 289 lung cancer samples was assessed by immunohistochemistry. Matched samples of metastatic lymph node and primary tumor tissues were used to quantify the expression of ERβ/IL6 by western blot. Expression levels of IL6 in NSCLC cells were quantified by western blotting, ELISA, and immunofluorescence staining. The effects of IL6 stimulated by E2 on cell malignancy were evaluated using CCK8, colony formation, wound healing and transwell. Furthermore, overexpression and knockdown ERβ constructs were constructed to measure the expression of IL6. The effects of IL6 stimulated by E2 on tumor growth were evaluated using a urethane-induced adenocarcinoma model. In addition, a xenograft mouse model was used to observe differences in ERβ subtype tumor growth with respect to IL6 expression. Results IL6/ERβ expression were significantly increased in lung cancer. Higher IL6/ERβ expression was associated with decreased differentiation or increased metastasis. IL6 was an independent prognostic factor for overall survival (OS), higher IL6 expression was associated with decreased OS. Furthermore, ERβ regulates IL6 expression via MAPK/ERK and PI3K/AKT pathways when stimulated by E2 and promotes cell malignancy in vitro and induced tumor growth in vivo. Finally we confirm that ERβ isolation 1/5 is essential for E2 promotion of IL6 expression, while ERβ2 not. Conclusions Our findings demonstrate that E2 stimulates IL6 expression to promote lung adenocarcinoma progression through the ERβ pathway. We also clarify the difference in each ERβ subtype for E2 promoting IL6 expression, suggesting that ERβ/IL6 might be potential targets for prognostic assessment and therapeutic intervention in lung cancer. Electronic supplementary material The online version of this article (10.1186/s13046-018-0804-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Quanfu Huang
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zheng Zhang
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yongde Liao
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Changyu Liu
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sheng Fan
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao Wei
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Ai
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Xiong
- Department of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
28
|
Giles ED, Jindal S, Wellberg EA, Schedin T, Anderson SM, Thor AD, Edwards DP, MacLean PS, Schedin P. Metformin inhibits stromal aromatase expression and tumor progression in a rodent model of postmenopausal breast cancer. Breast Cancer Res 2018; 20:50. [PMID: 29898754 PMCID: PMC6000949 DOI: 10.1186/s13058-018-0974-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 04/30/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Obesity and type II diabetes are linked to increased breast cancer risk in postmenopausal women. Patients treated with the antidiabetic drug metformin for diabetes or metabolic syndrome have reduced breast cancer risk, a greater pathologic complete response to neoadjuvant therapy, and improved breast cancer survival. We hypothesized that metformin may be especially effective when targeted to the menopausal transition, as this is a lifecycle window when weight gain and metabolic syndrome increase, and is also when the risk for obesity-related breast cancer increases. METHODS Here, we used an 1-methyl-1-nitrosourea (MNU)-induced mammary tumor rat model of estrogen receptor (ER)-positive postmenopausal breast cancer to evaluate the long-term effects of metformin administration on metabolic and tumor endpoints. In this model, ovariectomy (OVX) induces rapid weight gain, and an impaired whole-body response to excess calories contributes to increased tumor glucose uptake and increased tumor proliferation. Metformin treatment was initiated in tumor-bearing animals immediately prior to OVX and maintained for the duration of the study. RESULTS Metformin decreased the size of existing mammary tumors and inhibited new tumor formation without changing body weight or adiposity. Decreased lipid accumulation in the livers of metformin-treated animals supports the ability of metformin to improve overall metabolic health. We also found a decrease in the number of aromatase-positive, CD68-positive macrophages within the tumor microenvironment, suggesting that metformin targets the immune microenvironment in addition to improving whole-body metabolism. CONCLUSIONS These findings suggest that peri-menopause/menopause represents a unique window of time during which metformin may be highly effective in women with established, or at high risk for developing, breast cancer.
Collapse
Affiliation(s)
- Erin D Giles
- Department of Nutrition & Food Science, Texas A&M University, 373 Olsen Blvd; 2253 TAMU, College Station, TX, 77843, USA.
| | - Sonali Jindal
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, 3181 S.W. Sam Jackson Park Rd, Mailing Code: L215, Portland, OR, 97239, USA
| | - Elizabeth A Wellberg
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Troy Schedin
- Department of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Steven M Anderson
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Ann D Thor
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Dean P Edwards
- Departments of Molecular & Cellular Biology and Pathology Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Paul S MacLean
- Anschutz Health & Wellness Center, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.,Department of Medicine, Divisions of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.,Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Pepper Schedin
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, 3181 S.W. Sam Jackson Park Rd, Mailing Code: L215, Portland, OR, 97239, USA.,Knight Cancer Institute, Oregon Health & Science University, 1130 NW 22nd Ave #100, Portland, OR, 97239, USA
| |
Collapse
|
29
|
Pedersen AL, Brownrout JL, Saldanha CJ. Neuroinflammation and neurosteroidogenesis: Reciprocal modulation during injury to the adult zebra finch brain. Physiol Behav 2018; 187:51-56. [DOI: 10.1016/j.physbeh.2017.10.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 10/11/2017] [Accepted: 10/11/2017] [Indexed: 01/10/2023]
|
30
|
Filardo EJ. A role for G-protein coupled estrogen receptor (GPER) in estrogen-induced carcinogenesis: Dysregulated glandular homeostasis, survival and metastasis. J Steroid Biochem Mol Biol 2018; 176:38-48. [PMID: 28595943 DOI: 10.1016/j.jsbmb.2017.05.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 05/09/2017] [Accepted: 05/16/2017] [Indexed: 12/18/2022]
Abstract
Mechanisms of carcinogenesis by estrogen center on its mitogenic and genotoxic potential on tumor target cells. These models suggest that estrogen receptor (ER) signaling promotes expansion of the transformed population and that subsequent accumulation of somatic mutations that drive cancer progression occur via metabolic activation of cathecol estrogens or by epigenetic mechanisms. Recent findings that GPER is linked to obesity, vascular pathology and immunosuppression, key events in the development of metabolic syndrome and intra-tissular estrogen synthesis, provides an alternate view of estrogen-induced carcinogenesis. Consistent with this concept, GPER is directly associated with clinicopathological indices that predict cancer progression and poor survival in breast and gynecological cancers. Moreover, GPER manifests cell biological responses and a microenvironment conducive for tumor development and cancer progression, regulating cellular responses associated with glandular homeostasis and survival, invading surrounding tissue and attracting a vascular supply. Thus, the cellular actions attributed to GPER fit well with the known molecular mechanisms of G-protein coupled receptors, GPCRs, namely, their ability to transactivate integrins and EGF receptors and alter the interaction between glandular epithelia and their extracellular environment, affecting epithelial-to-mesenchymal transition (EMT) and allowing for tumor cell survival and dissemination. This perspective reviews the molecular and cellular responses manifested by GPER and evaluates its contribution to female reproductive cancers as diseases that progress as a result of dysregulated glandular homeostasis resulting in chronic inflammation and metastasis. This review is organized in sections as follows: I) a brief synopsis of the current state of knowledge regarding estrogen-induced carcinogenesis, II) a review of evidence from clinical and animal-based studies that support a role for GPER in cancer progression, and III) a mechanistic framework describing how GPER-mediated estrogen action may influence the tumor and its microenvironment.
Collapse
Affiliation(s)
- Edward J Filardo
- Division of Hematology & Oncology, The Warren Alpert School of Medicine, Brown University, Providence, RI 02818, United States.
| |
Collapse
|
31
|
Li J, Zhu Q, Yang B, Ning C, Liu X, Luo X, Chen X. Risk factors for ovarian involvement in young and premenopausal endometrioid endometrial cancer patients. Eur J Obstet Gynecol Reprod Biol 2018; 222:151-154. [PMID: 29408747 DOI: 10.1016/j.ejogrb.2018.01.030] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Revised: 01/21/2018] [Accepted: 01/25/2018] [Indexed: 11/18/2022]
Abstract
OBJECTIVE This study aimed to investigate the incidence of ovarian malignant involvement in young and premenopausal endometrioid endometrial cancer and study the possible risk factors. METHODS Premenopausal patients 45 years of age or younger with endometrioid endometrial cancer treated at the OB/GYN Hospital of Fudan University between 2009 and 2013 were identified. The incidence of ovarian malignant involvement in young and premenopausal endometrioid endometrial cancer patients were calculated and the possible risk factors were investigated. RESULTS A total of 144 younger (age ≤ 45, premenopausal) patients with endometrioid endometrial cancer were identified and coexisting malignant ovarian neoplasms were detected in 6 patients. Univariate analysis revealed that deeper myometrial invasion, positive lymphonode metastasis, positive LVSI, and high histologic grade (G2-G3) were associated with ovarian involvement in younger endometrial cancer patients. However, multivariate analysis revealed that only deep myometrial invasion was an independent risk factors for ovarian involvement (OR = 12.81, P = 0.046). CONCLUSION In conclusions, the incidence of coexisting ovarian malignant neoplasms in young and premenopausal patients with endometrioid endometrial cancer is low, and these findings may facilitate preoperative counseling of patients and decision making at the time of surgery.
Collapse
Affiliation(s)
- Jun Li
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China; Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| | - Qin Zhu
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China
| | - Bingyi Yang
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China; Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| | - Chengcheng Ning
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China; Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| | - Xiaoxia Liu
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China.
| | - Xuezhen Luo
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China; Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China.
| | - Xiaojun Chen
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China; Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China.
| |
Collapse
|
32
|
Cheng Y, Lv Q, Xie B, Yang B, Shan W, Ning C, Li B, Xie L, Gu C, Luo X, Chen X, Zhu Q. Estrogen and high-fat diet induced alterations in C57BL/6 mice endometrial transcriptome profile. Endocr Connect 2018; 7:36-46. [PMID: 29133384 PMCID: PMC5744625 DOI: 10.1530/ec-17-0315] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 11/10/2017] [Indexed: 12/31/2022]
Abstract
Unopposed estrogen stimulation and insulin resistance are known to play important roles in endometrial cancer (EC), but the interaction between these two factors and how they contribute to endometrial lesions are not completely elucidated. To investigate the endometrial transcriptome profile and the associated molecular pathway alterations, we established an ovariectomized C57BL/6 mouse model treated with subcutaneous implantation of 17-β estradiol (E2) pellet and/or high-fat diet (HFD) for 12 weeks to mimic sustained estrogen stimulation and insulin resistance. Histomorphologically, we found that both E2 and E2 + HFD groups showed markedly enlarged uterus and increased number of endometrial glands. The endometrium samples were collected for microarray assay. GO and KEGG analysis showed that genes regulated by E2 and/or HFD are mainly responsible for immune response, inflammatory response and metabolic pathways. Further IPA analysis demonstrated that the acute phase response signaling, NF-κB signaling, leukocyte extravasation signaling, PPAR signaling and LXR/RXR activation pathways are mainly involved in the pathways above. In addition, the genes modulated reciprocally by E2 and/or HFD were also analyzed, and their crosstalk mainly focuses on enhancing one another's activity. The combination analysis of microarray data and TCGA database provided potential diagnostic or therapeutic targets for EC. Further validation was performed in mice endometrium and human EC cell lines. In conclusion, this study unraveled the endometrial transcriptome profile alterations affected by E2 and/or HFD that may disturb endometrial homeostasis and contribute to the development of endometrial hyperplasia.
Collapse
Affiliation(s)
- Yali Cheng
- Department of GynecologyObstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Qiaoying Lv
- Department of GynecologyObstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Bingying Xie
- Department of GynecologyObstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Bingyi Yang
- Department of GynecologyObstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Weiwei Shan
- Department of GynecologyObstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Chengcheng Ning
- Department of GynecologyObstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Bing Li
- Department of GynecologyObstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Liying Xie
- Department of GynecologyObstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Chao Gu
- Department of GynecologyObstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Xuezhen Luo
- Department of GynecologyObstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Xiaojun Chen
- Department of GynecologyObstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Qin Zhu
- Department of PathologyObstetrics and Gynecology, Hospital of Fudan University, Shanghai, China
| |
Collapse
|
33
|
Ding DC, Chu TY, Liu HW. Reciprocal crosstalk between endometrial carcinoma and mesenchymal stem cells via transforming growth factor-β/transforming growth factor receptor and C-X-C motif chemokine ligand 12/C-X-C chemokine receptor type 4 aggravates malignant phenotypes. Oncotarget 2017; 8:115202-115214. [PMID: 29383153 PMCID: PMC5777765 DOI: 10.18632/oncotarget.23212] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 08/06/2017] [Indexed: 01/02/2023] Open
Abstract
Designated for cyclic shedding, the endometrial stroma is rich in endometrial mesenchymal stem cells (EMSCs) and may play an important role in the development of endometrial carcinoma (EC). This study characterized the crosstalk of EC cells with EMSCs and the resultant effects on malignant phenotypes. The cultured EMSCs expressed CD73, CD90, and CD105, but not CD14, CD19, CD34, CD45, or human leukocyte antigen—antigen D related markers. These EMSCs also showed osteogenic, adipogenic, and chondrogenic differentiation ability. Transforming growth factor (TGF)-β1 and C–X–C motif chemokine ligand 12 (CXCL12) secretion or expression were reciprocally enhanced in EC cells and EMSCs, as well as in their tissues. By acting on the receptors expressed in their mutual target cells, the interaction between TGF-β and CXCL12 results in the enhanced migration, invasion, tumorigenesis, and epithelial–mesenchymal transition of EC cells, which can be blocked by neutralizing the antibody of either CXCL12 or C–X–C chemokine receptor type 4. The study revealed unprecedented paracrine interactions between EC cells and EMSCs that resulted in the enhancement of transformation phenotypes. Thus, the blocking of TGF-β or CXCL12 signaling can be a therapeutic target for EC.
Collapse
Affiliation(s)
- Dah-Ching Ding
- Department of Obstetrics and Gynecology, Buddhist Tzu-Chi General Hospital, Hualien, Taiwan.,Institute of Medical Sciences, Tzu Chi University; Hualien, Taiwan
| | - Tang-Yuan Chu
- Department of Obstetrics and Gynecology, Buddhist Tzu-Chi General Hospital, Hualien, Taiwan.,Institute of Medical Sciences, Tzu Chi University; Hualien, Taiwan.,Cervical Cancer Prevention Center, Department of Research, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Hwan-Wun Liu
- Institute of Medical Sciences, Tzu Chi University; Hualien, Taiwan.,Department of Occupational Medicine, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| |
Collapse
|
34
|
Yang T, Zhang H, Qiu H, Li B, Wang J, Du G, Ren C, Wan X. EFEMP1 is repressed by estrogen and inhibits the epithelial-mesenchymal transition via Wnt/β-catenin signaling in endometrial carcinoma. Oncotarget 2017; 7:25712-25. [PMID: 27015552 PMCID: PMC5041938 DOI: 10.18632/oncotarget.8263] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 03/02/2016] [Indexed: 01/06/2023] Open
Abstract
Epidermal growth factor-containing fibulin-like extracellular matrix protein 1 (EFEMP1) acted as a tumor suppressor in endometrial carcinoma (EC). However, the correlation between EFEMP1 and estrogen is unknown. Here, we reported that the expression of EFEMP1 was conversely associated with ERα in endometrial carcinoma tissues. In endometrial carcinoma cells, estrogen/ERα signaling significantly suppressed the expression of EFEMP1. Moreover, chromatin immunoprecipitation (CHIP) and dual-luciferase reporter assays demonstrate that estrogen/ERα bound to the estrogen response element (ERE) located in EFEMP1 promoter and repressed its expression. Besides, in vitro and in vivo, EFEMP1 could remarkably suppress the expression of epithelial-mesenchymal transition (EMT) markers such as Vimentin, Snail and the Wnt/β-catenin target genes like Cyclin-D1 and c-Myc, which could be restored when EFEMP1 was silenced. In addition, XAV93920 (the inhibitor of the Wnt/β-catenin pathway) blocked and LiCl (the activator of the Wnt/β-catenin pathway) enhanced the effect of EFEMP1 on EMT. In conclusion, we demonstrated that estrogen/ERα signal suppresses EFEMP1. Besides, EFEMP1 inhibits EMT via interfering the Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Tingting Yang
- Department of Obstetrics and Gynecology, Shanghai First People's Hospital Affiliated to Nanjing Medical University, Nanjing, China.,Center for Reproductive Medicine, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Huilin Zhang
- Department of Obstetrics and Gynecology, Shanghai First People's Hospital Affiliated to Nanjing Medical University, Nanjing, China.,Department of Gynecology and Obstetrics, Nanjing Maternal and Children Care Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Haifeng Qiu
- Department of Obstetrics and Gynecology, Shanghai First People's Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Bilan Li
- Department of Obstetrics and Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jingyun Wang
- Department of Obstetrics and Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Guiqiang Du
- Department of Obstetrics and Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chune Ren
- Center for Reproductive Medicine, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Xiaoping Wan
- Department of Obstetrics and Gynecology, Shanghai First People's Hospital Affiliated to Nanjing Medical University, Nanjing, China.,Department of Obstetrics and Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
35
|
Navarro FC, Watkins SK. Estrogen Stimulation Differentially Impacts Human Male and Female Antigen-Specific T Cell Anti-Tumor Function and Polyfunctionality. GENDER AND THE GENOME 2017. [DOI: 10.1089/gg.2017.0014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Sex-specific differences exist in innate and adaptive immune responses and are mediated by hormone signaling. Estrogen is able to differentially modulate the development and differentiation of immune cells, including T cells. However, the effect of estrogen on T cell function, especially at concentrations other than physiological, remains controversial and incompletely understood. Immunotherapy is one of the most promising cancer treatments to date with a high probability of future enhancements. The adoptive transfer of genetically modified T cells can mediate tumor regression but there are still many hurdles to enhancing the proficiency of this treatment. This study demonstrates for the first time that one major aspect to consider for designing potent immunotherapies for cancer is the impact of the patient's sex. Herein, using two different Ag-specific T cell groups, we investigated the effect of sex and estrogen in antitumor effector responses, T helper cytokine secretion, and, importantly, on T cell whole polyfunctionality important for memory T cell development and survival. Major differences were observed in T cell function and polyfunctionality between sexes and on E2 treatment. The findings of this study may be critical to understand the results of immunotherapy on different patients and for the enhancement of immunotherapy for cancer.
Collapse
Affiliation(s)
- Flor C. Navarro
- Department of Surgery, Cardinal Bernardin Cancer Center, Loyola University Chicago, Maywood, Illinois
| | - Stephanie K. Watkins
- Department of Surgery, Cardinal Bernardin Cancer Center, Loyola University Chicago, Maywood, Illinois
| |
Collapse
|
36
|
Aspirin use and endometrial cancer risk and survival. Gynecol Oncol 2017; 148:222-232. [PMID: 29132875 DOI: 10.1016/j.ygyno.2017.10.026] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 10/20/2017] [Accepted: 10/23/2017] [Indexed: 02/06/2023]
Abstract
The role of acetylsalicylic acid (aspirin) as a chemo-preventive and adjuvant therapeutic agent for cancers is generating attention. Mounting evidence indicates that aspirin reduces the incidence and mortality of certain obesity-related cancers, particularly colorectal cancer. In endometrial cancer, previous studies examining the effect of aspirin remain inconsistent as to the reduction in the risk of endometrial cancer. While some evidence indicates protective effects in obese women, other studies have showed a potential deleterious effect of these medications on endometrial cancer outcomes. However, exposure measurement across studies has been inconsistent in recording dose, duration, and frequency of use; thus making comparisons difficult. In this article, we review the evidence for the association between endometrial cancer and obesity, the pharmacological differences between regular- and low-dose aspirin, as well as the potential anti-tumor mechanism of aspirin, supporting a possible therapeutic effect on endometrial cancer. A proposed mechanism behind decreased cancer mortality in endometrial cancer may be a result of inhibition of metastasis via platelet inactivation and possible prostaglandin E2 suppression by aspirin. Additionally, aspirin use in particular may have a secondary benefit for obesity-related comorbidities including cardiovascular disease in women with endometrial cancer. Although aspirin-related bleeding needs to be considered as a possible adverse effect, the benefits of aspirin therapy may exceed the potential risk in women with endometrial cancer. The current evidence reviewed herein has resulted in conflicting findings regarding the potential effect on endometrial cancer outcomes, thus indicating that future studies in this area are needed to resolve the effects of aspirin on endometrial cancer survival, particularly to identify specific populations that might benefit from aspirin use.
Collapse
|
37
|
Su T, Qu JJ, Wang K, Li BL, Zhao D, Zhu YP, Ye L, Lu W, Wan XP. Cross-talk between p21-activated kinase 4 and ERα signaling triggers endometrial cancer cell proliferation. Oncotarget 2017; 8:68083-68094. [PMID: 28978098 PMCID: PMC5620238 DOI: 10.18632/oncotarget.19188] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Accepted: 06/13/2017] [Indexed: 01/16/2023] Open
Abstract
Cross-talk between estrogen receptor alpha (ERα) and signal transduction pathways plays an important role in the progression of endometrial cancer (EC). Here, we show that 17β-estradiol (E2) stimulation increases p21-activated kinase 4 (Pak4) expression and activation in ER-positive EC cells. The estrogen-induced Pak4 activation is mediated via the PI3K/AKT pathway. Estrogen increases Pak4 and phosphorylated-Pak4 (p-Pak4) nuclear accumulation, and Pak4 in turn enhances ERα trans-activation. Depletion or functional inhibition of Pak4 abrogates EC cell proliferation induced by E2, whereas overexpression of Pak4 rescues cell proliferation decreased by inhibiting the estrogen pathway. Pak4 knockdown decreases cyclin D1 expression and induces G1-S arrest. Importantly, Pak4 suppression inhibits E2 induced EC tumor growth in vivo, in a mouse xenograft model. These data demonstrate that estrogen stimulation increases Pak4 expression and activation, which in turn enhances ERα transcriptional activity and ERα-dependent gene expression, resulting in increased proliferation of EC cells. Thus inhibition of Pak4-ERα signaling may represent a novel therapeutic strategy against endometrial carcinoma.
Collapse
Affiliation(s)
- Tao Su
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, P.R. China.,Department of Gynecology, The International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Jun-Jie Qu
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, P.R. China
| | - Kai Wang
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, P.R. China
| | - Bi-Lan Li
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, P.R. China
| | - Dong Zhao
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, P.R. China
| | - Yi-Ping Zhu
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, P.R. China
| | - Lei Ye
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, P.R. China
| | - Wen Lu
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, P.R. China
| | - Xiao-Ping Wan
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, P.R. China
| |
Collapse
|
38
|
STAT5 deletion in macrophages alters ductal elongation and branching during mammary gland development. Dev Biol 2017; 428:232-244. [PMID: 28606561 PMCID: PMC5621646 DOI: 10.1016/j.ydbio.2017.06.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 04/04/2017] [Accepted: 06/06/2017] [Indexed: 12/31/2022]
Abstract
Macrophages are required for proper mammary gland development and maintaining tissue homeostasis. However, the mechanisms by which macrophages regulate this process remain unclear. Here, we identify STAT5 as an important regulator of macrophage function in the developing mammary gland. Analysis of mammary glands from mice with STAT5-deficient macrophages demonstrates delayed ductal elongation, enhanced ductal branching and increased epithelial proliferation. Further analysis reveals that STAT5 deletion in macrophages leads to enhanced expression of proliferative factors such as Cyp19a1/aromatase and IL-6. Mechanistic studies demonstrate that STAT5 binds directly to the Cyp19a1 promoter in macrophages to suppress gene expression and that loss of STAT5 results in enhanced stromal expression of aromatase. Finally, we demonstrate that STAT5 deletion in macrophages cooperates with oncogenic initiation in mammary epithelium to accelerate the formation of estrogen receptor (ER)-positive hyperplasias. These studies establish the importance of STAT5 in macrophages during ductal morphogenesis in the mammary gland and demonstrate that altering STAT5 function in macrophages can affect the development of tissue-specific disease.
Collapse
|
39
|
Uzan J, Laas E, Alsamad IA, Skalli D, Mansouri D, Haddad B, Touboul C. Supervised Clustering of Adipokines and Hormonal Receptors Predict Prognosis in a Population of Obese Women with Type 1 Endometrial Cancer. Int J Mol Sci 2017; 18:ijms18051055. [PMID: 28505082 PMCID: PMC5454967 DOI: 10.3390/ijms18051055] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 05/06/2017] [Accepted: 05/10/2017] [Indexed: 12/29/2022] Open
Abstract
Obesity is a major risk factor for endometrial cancer (EC). Yet, its impact on prognosis is controversial. Obesity is associated with metabolic and hormonal dysregulation as well as adipokines increase. The aim of this study was to characterize the expression of biological factors related to obesity within the tumor and evaluate their impact on prognosis. One hundred and thirty-six patients, including 55 obese patients, with endometrioid type I EC operated by total hysterectomy were included in this retrospective study conducted in a Tertiary teaching hospital between 2000 and 2013. Immunohistochemistry (IHC) study was performed on type I EC tumor samples using five adipokines (SPARC, RBP4 (Retinol Binding Protein 4), adiponectin, TNF α, IL-6) and hormonal receptors (estrogen receptor and progesterone receptor). Supervised clustering of immunohistochemical markers was performed to identify clusters that could be associated with prognostic groups. The prognosis of the obese population was not different from the prognosis of the general population. Adipokine expression within tumors was not different in these two populations. In obese population, we found three clusters where co-expression was associated with a recurrence group in comparison with a non-recurrence group and four clusters where co-expression was associated with the high risk FIGO (International Federation of Gynecology and Obstetrics) stage I group in comparison of low risk FIGO stage I group. While obesity does not appear as a prognostic factor in endometrioid type I EC, the co-expression of biological factors in IHC on hysterectomy specimens allowed to distinguish two prognostic groups in obese population.
Collapse
Affiliation(s)
- Jennifer Uzan
- Department of Obstetrics and Gynecology, Centre Hospitalier Intercommunal de Créteil, 40 Avenue de Verdun, 94000 Créteil, France.
- Department of Pathology, Centre Hospitalier Intercommunal de Créteil, 40 Avenue de Verdun, 94000 Créteil, France.
| | - Enora Laas
- Department of Surgery, Institut Curie, 26 rue d'Ulm, 75005 Paris, France.
| | - Issam Abd Alsamad
- Department of Pathology, Centre Hospitalier Intercommunal de Créteil, 40 Avenue de Verdun, 94000 Créteil, France.
| | - Dounia Skalli
- Department of Obstetrics and Gynecology, Centre Hospitalier Intercommunal de Créteil, 40 Avenue de Verdun, 94000 Créteil, France.
| | - Dhouha Mansouri
- Department of Pathology, Centre Hospitalier Intercommunal de Créteil, 40 Avenue de Verdun, 94000 Créteil, France.
| | - Bassam Haddad
- Department of Obstetrics and Gynecology, Centre Hospitalier Intercommunal de Créteil, 40 Avenue de Verdun, 94000 Créteil, France.
| | - Cyril Touboul
- Department of Obstetrics and Gynecology, Centre Hospitalier Intercommunal de Créteil, 40 Avenue de Verdun, 94000 Créteil, France.
- INSERM/Paris 7 U965 "Carcinose, Angiogénèse-Recherche Translationnelle", Centre Hospitalier Universitaire Lariboisière, Assistance Publique des Hôpitaux de Paris (AP-HP), 2 rue Ambroise Paré, 75010 Paris, France.
| |
Collapse
|
40
|
Thompson MG, Peiffer DS, Larson M, Navarro F, Watkins SK. FOXO3, estrogen receptor alpha, and androgen receptor impact tumor growth rate and infiltration of dendritic cell subsets differentially between male and female mice. Cancer Immunol Immunother 2017; 66:615-625. [PMID: 28229217 PMCID: PMC11028910 DOI: 10.1007/s00262-017-1972-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 02/07/2017] [Indexed: 01/18/2023]
Abstract
Tumors evade immune recognition and destruction in many ways including the creation of an immune-suppressive tumor microenvironment (TME). Dendritic cells (DC) that infiltrate the TME are tolerogenic, and suppress effector T cells and anti-tumor activity. Previous reports demonstrated that a key regulator of tolerance in DC is the transcription factor FOXO3. Gender disparity has been studied in cancer in relation to incidence, aggressiveness, and prognosis. Few studies have touched on the importance in relation to impact on the immune system. In the current study, we show that there are significant differences in tumor growth between males and females. Additionally, frequencies and the function of FOXO3 expressed by DC subsets that infiltrate tumors vary between genders. Our results show for the first time that DC FOXO3 expression and function is altered in females. In vitro results indicate that these differences may be the result of exposure to estrogen. These differences may be critical considerations for the enhancement of immunotherapy for cancer.
Collapse
Affiliation(s)
- Matthew G Thompson
- Department of Surgery, Cardinal Bernardin Cancer Center, Loyola University Chicago, 2160 S. 1st Ave. Building 112, Maywood, IL, 60153, USA
| | - Daniel S Peiffer
- Department of Surgery, Cardinal Bernardin Cancer Center, Loyola University Chicago, 2160 S. 1st Ave. Building 112, Maywood, IL, 60153, USA
| | - Michelle Larson
- Department of Surgery, Cardinal Bernardin Cancer Center, Loyola University Chicago, 2160 S. 1st Ave. Building 112, Maywood, IL, 60153, USA
| | - Flor Navarro
- Department of Surgery, Cardinal Bernardin Cancer Center, Loyola University Chicago, 2160 S. 1st Ave. Building 112, Maywood, IL, 60153, USA
| | - Stephanie K Watkins
- Department of Surgery, Cardinal Bernardin Cancer Center, Loyola University Chicago, 2160 S. 1st Ave. Building 112, Maywood, IL, 60153, USA.
| |
Collapse
|
41
|
Lee J, Hong BS, Ryu HS, Lee HB, Lee M, Park IA, Kim J, Han W, Noh DY, Moon HG. Transition into inflammatory cancer-associated adipocytes in breast cancer microenvironment requires microRNA regulatory mechanism. PLoS One 2017; 12:e0174126. [PMID: 28333977 PMCID: PMC5363867 DOI: 10.1371/journal.pone.0174126] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Accepted: 03/05/2017] [Indexed: 01/25/2023] Open
Abstract
The role of adipocytes in cancer microenvironment has gained focus during the recent years. However, the characteristics of the cancer-associated adipocytes (CAA) in human breast cancer tissues and the underlying regulatory mechanism are not clearly understood. We reviewed pathology specimens of breast cancer patients to understand the morphologic characteristics of CAA, and profiled the mRNA and miRNA expression of CAA by using indirect co-culture system in vitro. The CAAs in human breast cancers showed heterogeneous topographic relationship with breast cancer cells within the breast microenvironment. The CAAs exhibited the characteristics of de-differentiation determined by their microscopic appearance and the expression levels of adipogenic markers. Additionally, the 3T3-L1 adipocytes indirectly co-cultured with breast cancer cells showed up-regulation of inflammation-related genes including Il6 and Ptx3. The up-regulation of IL6 in CAA was further observed in human breast cancer tissues. miRNA array of indirectly co-cultured 3T3-L1 cells showed increased expression of mmu-miR-5112 which may target Cpeb1. Cpeb1 is a negative regulator of Il6. The suppressive role of mmu-miR-5112 was confirmed by dual luciferase reporter assay, and mmu-miR-5112-treated adipocytes showed up-regulation of Il6. The transition of adipocytes into more inflammatory CAA resulted in proliferation-promoting effect in ER positive breast cancer cells such as MCF7 and ZR-75-1 but not in ER negative cells. In this study, we have determined the de-differentiated and inflammatory natures of CAA in breast cancer microenvironment. Additionally, we propose a miRNA-based regulatory mechanism underlying the process of acquiring inflammatory phenotypes in CAA.
Collapse
Affiliation(s)
- Jiwoo Lee
- Center for Medical Innovation, Seoul National University Hospital, Seoul, Korea
| | - Bok Sil Hong
- Center for Medical Innovation, Seoul National University Hospital, Seoul, Korea
| | - Han Suk Ryu
- Department of Pathology, Seoul National University Hospital, Seoul, Korea
| | - Han-Byoel Lee
- Center for Medical Innovation, Seoul National University Hospital, Seoul, Korea
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Minju Lee
- Center for Medical Innovation, Seoul National University Hospital, Seoul, Korea
| | - In Ae Park
- Department of Pathology, Seoul National University Hospital, Seoul, Korea
| | - Jisun Kim
- Center for Medical Innovation, Seoul National University Hospital, Seoul, Korea
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Wonshik Han
- Center for Medical Innovation, Seoul National University Hospital, Seoul, Korea
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Dong-Young Noh
- Center for Medical Innovation, Seoul National University Hospital, Seoul, Korea
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Hyeong-Gon Moon
- Center for Medical Innovation, Seoul National University Hospital, Seoul, Korea
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
- * E-mail:
| |
Collapse
|
42
|
Lv QY, Xie BY, Yang BY, Ning CC, Shan WW, Gu C, Luo XZ, Chen XJ, Zhang ZB, Feng YJ. Increased TET1 Expression in Inflammatory Microenvironment of Hyperinsulinemia Enhances the Response of Endometrial Cancer to Estrogen by Epigenetic Modulation of GPER. J Cancer 2017; 8:894-902. [PMID: 28382153 PMCID: PMC5381179 DOI: 10.7150/jca.17064] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 12/09/2016] [Indexed: 12/27/2022] Open
Abstract
Background: Insulin resistance (IR) has been well studied in the initiation and development of endometrial endometrioid carcinoma (EEC). As yet, it has been largely neglected for estrogen sensitivity in local endometrium in hyperinsulinemia-induced systemic microenvironment. The aim of this study was to investigate the role of insulin in regulating estrogen sensitivity and explore the potential mechanisms in insulin-driven inflammatory microenvironment. Methods: We first investigated the effect of insulin on estradiol-driven endometrial cancer cells proliferation in vitro to address the roles of insulin in modulating estrogen sensitivity. Then GPER, ERα and TET1 in EEC samples with or without insulin resistance were screened by immunohistochemistry to confirm whether insulin resistance regulates estrogen receptors. Further mechanism analysis was carried out to address whether TET1 was mediated epigenetic modulation of GPER in insulin-induced microenvironment. Results: Insulin enhanced estradiol-driven endometrial cancer cells proliferation by up-regulating G-protein-coupled estrogen receptor (GPER) expression, but not ERα or ERβ. Immunohistochemistry of EEC tissues showed that GPER expression was greatly increased in endometrial tissues from EEC subjects with insulin resistance and was positively correlated with Ten-eleven-translocation 1 (TET1) expression. Mechanistically, insulin up-regulates TET1 expression, and the latter, an important DNA hydroxymethylase, could up-regulate GPER expression through epigenetic modulation. Conclusion: This study identified TET1 as the upstream regulator of GPER expression and provides a possible mechanism that insulin-induced positive regulation of estrogen sensitivity in endometrial cancer cells. Increasing expression of GPER through TET1-mediated epigenetic modulation may emerge as the main regulator to enhance the response of endometrial cancer to estrogen in insulin-driven inflammatory microenvironment.
Collapse
Affiliation(s)
- Qiao-Ying Lv
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200011, China
| | - Bing-Ying Xie
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200011, China
| | - Bing-Yi Yang
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200011, China
| | - Cheng-Cheng Ning
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200011, China
| | - Wei-Wei Shan
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200011, China
| | - Chao Gu
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200011, China
| | - Xue-Zhen Luo
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200011, China
| | - Xiao-Jun Chen
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200011, China
| | - Zhen-Bo Zhang
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University school of medicine, Shanghai, 201620, China
| | - You-Ji Feng
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University school of medicine, Shanghai, 201620, China
| |
Collapse
|
43
|
Manna PR, Molehin D, Ahmed AU. Dysregulation of Aromatase in Breast, Endometrial, and Ovarian Cancers: An Overview of Therapeutic Strategies. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 144:487-537. [PMID: 27865465 DOI: 10.1016/bs.pmbts.2016.10.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Aromatase is the rate-limiting enzyme in the biosynthesis of estrogens, which play crucial roles on a spectrum of developmental and physiological processes. The biological actions of estrogens are classically mediated by binding to two estrogen receptors (ERs), ERα and ERβ. Encoded by the cytochrome P450, family 19, subfamily A, polypeptide 1 (CYP19A1) gene, aromatase is expressed in a wide variety of tissues, as well as benign and malignant tumors, and is regulated in a pathway- and tissue-specific manner. Overexpression of aromatase, leading to elevated systemic levels of estrogen, is unequivocally linked to the pathogenesis and growth of a number malignancies, including breast, endometrium, and ovarian cancers. Aromatase inhibitors (AIs) are routinely used to treat estrogen-dependent breast cancers in postmenopausal women; however, their roles in endometrial and ovarian cancers remain obscure. While AI therapy is effective in hormone sensitive cancers, they diminish estrogen production throughout the body and, thus, generate undesirable side effects. Despite the effectiveness of AI therapy, resistance to endocrine therapy remains a major concern and is the leading cause of cancer death. Considerable advances, toward mitigating these issues, have evolved in conjunction with a number of histone deacetylase (HDAC) inhibitors for countering an assortment of diseases and cancers, including the aforesaid malignancies. HDACs are a family of enzymes that are frequently dysregulated in human tumors. This chapter will discuss the current understanding of aberrant regulation and expression of aromatase in breast, endometrial, and ovarian cancers, and potential therapeutic strategies for prevention and treatment of these life-threatening diseases.
Collapse
Affiliation(s)
- P R Manna
- Texas Tech University Health Sciences Center School of Medicine, Lubbock, TX, United States.
| | - D Molehin
- Texas Tech University Health Sciences Center School of Medicine, Lubbock, TX, United States
| | - A U Ahmed
- Texas Tech University Health Sciences Center School of Medicine, Lubbock, TX, United States
| |
Collapse
|
44
|
Pekson R, Poltoratsky V, Gorasiya S, Sundaram S, Ashby CR, Vancurova I, Reznik SE. N,N-Dimethylacetamide Significantly Attenuates LPS- and TNFα-Induced Proinflammatory Responses Via Inhibition of the Nuclear Factor Kappa B Pathway. Mol Med 2016; 22:747-758. [PMID: 27782292 DOI: 10.2119/molmed.2016.00017] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Accepted: 10/18/2016] [Indexed: 12/25/2022] Open
Abstract
Previously, we have shown that N,N-dimethylacetamide (DMA) prevents inflammation-induced preterm birth in a murine model, inhibits LPS-induced increases in placental pro-inflammatory cytokines and up-regulates the anti-inflammatory cytokine Interleukin-10 (IL-10). However, DMA's mechanism of action remains to be elucidated. In the current study we investigate how DMA produces its anti-inflammatory effect. Using in vitro and ex vivo models, we show that DMA suppresses secretion of pro-inflammatory cytokines in lipopolysaccharide (LPS)-induced RAW 264.7 cells, TNFα-challenged JEG-3 cells and LPS-stimulated human placental explants. DMA significantly attenuated the secretion of TNFα, IL-6, IL-10, and granulocyte macrophage colony stimulating factor (GM-CSF) from LPS-stimulated RAW 264.7 cells, IL-6 secretion from TNFα-stimulated JEG-3 cells and TNFα, IL-6, IL-10, GM-CSF and Interleukin-8 (IL-8) from LPS-stimulated human placental explants. We further investigated if DMA's effect on cytokine expression involves the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and mitogen-activated protein kinase (MAPK) pathways. DMA (10 mM) significantly inhibited nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor, alpha (IκBα) degradation in LPS-stimulated RAW 264.7 cells, but there was no significant change in the expression of phosphorylated or native forms of downstream proteins in the MAPK pathway. In addition, DMA significantly attenuated luciferase activity in cells co-transfected with NF-κB-Luc reporter plasmid, but not with AP-1-Luc or CEBP-Luc reporters. Overall, our findings suggest that the anti-inflammatory activity of DMA is mediated by inhibition of the NF-κB pathway via decreased IκBα degradation.
Collapse
Affiliation(s)
- Ryan Pekson
- Dept of Pharmaceutical Sciences, St. John's University
| | | | | | | | | | | | - Sandra E Reznik
- Dept of Pharmaceutical Sciences, St. John's University.,Depts of Pathology and Obstetrics and Gynecology and Women's Health, Albert Einstein College of Medicine
| |
Collapse
|
45
|
Ito K, Miki Y, Suzuki T, McNamara KM, Sasano H. In situ androgen and estrogen biosynthesis in endometrial cancer: focus on androgen actions and intratumoral production. Endocr Relat Cancer 2016; 23:R323-35. [PMID: 27287451 DOI: 10.1530/erc-15-0470] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 06/10/2016] [Indexed: 01/20/2023]
Abstract
In situ estrogen biosynthesis is considered to play pivotal roles in the development and progression of human endometrial carcinoma. However, the biological roles of androgen have remained virtually unknown. Various epidemiological studies have revealed that elevated serum androgen levels are generally associated with an increased risk of developing endometrial carcinoma; however, studies directly examining androgens in carcinoma tissues are relatively rare and reviews summarizing this information are scarce. Therefore, we summarized recent studies on androgens in endometrial carcinoma, especially focusing androgen actions and in situ androgen biosynthesis. Among the enzymes required for local biosynthesis of androgen, 17β-hydroxysteroid dehydrogenase type 5 (conversion from androstenedione to testosterone) and 5α-reductase (reduction of testosterone to dihydrotestosterone (DHT)) are the principal enzymes involved in the formation of biologically most potent androgen, DHT. Both enzymes and androgen receptor were expressed in endometrial carcinoma tissues, and in situ production of DHT has been reported to exist in endometrial carcinoma tissues. However, testosterone is not only a precursor of DHT production, but also a precursor of estradiol synthesis, as a substrate of the aromatase enzyme. Therefore, aromatase could be another key enzyme serving as a negative regulator for in situ production of DHT by reducing amounts of the precursor. In an in vitro study, DHT was reported to exert antiproliferative effects on endometrial carcinoma cells. Intracrine mechanisms of androgens, the downstream signals of AR, which are directly related to anticancer progression, and the clinical significance of DHT-AR pathway in the patients with endometrial carcinoma have, however, not been fully elucidated.
Collapse
Affiliation(s)
- Kiyoshi Ito
- Department of Disaster Obstetrics and GynecologyInternational Research Institute of Disaster Science (IRIDeS), Tohoku University, Sendai, Japan Department of Disaster Obstetrics and GynecologyTohoku University Graduate School of Medicine, Sendai, Japan
| | - Yasuhiro Miki
- Department of Disaster Obstetrics and GynecologyInternational Research Institute of Disaster Science (IRIDeS), Tohoku University, Sendai, Japan
| | - Takashi Suzuki
- Department of Pathology and HistotechnologyTohoku University Graduate School of Medicine, Sendai, Japan
| | - Keely May McNamara
- Department of PathologyTohoku University Graduate School of Medicine, Sendai, Japan
| | - Hironobu Sasano
- Department of PathologyTohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
46
|
Ke J, Yang Y, Che Q, Jiang F, Wang H, Chen Z, Zhu M, Tong H, Zhang H, Yan X, Wang X, Wang F, Liu Y, Dai C, Wan X. Prostaglandin E2 (PGE2) promotes proliferation and invasion by enhancing SUMO-1 activity via EP4 receptor in endometrial cancer. Tumour Biol 2016; 37:12203-12211. [PMID: 27230680 PMCID: PMC5080328 DOI: 10.1007/s13277-016-5087-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 05/15/2016] [Indexed: 11/26/2022] Open
Abstract
Prostaglandin E2 (PGE2), a derivative of arachidonic acid, has been identified as a tumorigenic factor in many cancers in recent studies. Prostaglandin E synthase 2 (PTGES2) is an enzyme that in humans is encoded by the PTGES2 gene located on chromosome 9, and it synthesizes PGE2 in human cells. In our study, we selected 119 samples from endometrial cancer patients, with 50 normal endometrium tissue samples as controls, in which we examined the expression of PTGES2. Both immunohistochemistry (IHC) and Western blot analyses demonstrated that synthase PTGES2, which is required for PGE2 synthesis, was highly expressed in endometrium cancer tissues compared with normal endometrium. Stable PTGES2-shRNA transfectants were generated in Ishikawa and Hec-1B endometrial cancer cell lines, and transfection efficiencies were confirmed by RT-PCR and Western blot analyses. We found that PGE2 promoted proliferation and invasion of cells in Ishikawa and Hec-1B cells by cell counting kit-8 tests (CCK8) and transwell assays, respectively. PGE2 stimulation enhanced the expression of SUMO-1, via PGE2 receptor subtype 4 (EP4). Further analysis implicated the Wnt/β-catenin signaling pathway function as the major mediator of EP4 and SUMO-1. The increase in SUMO-1 activity prompted the SUMOlyation of target proteins which may be involved in proliferation and invasion. These findings suggest SUMO-1 and EP4 as two potential targets for new therapeutic or prevention strategies for endometrial cancers.
Collapse
Affiliation(s)
- Jieqi Ke
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yixia Yang
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qi Che
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feizhou Jiang
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huihui Wang
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zheng Chen
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Minjiao Zhu
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huan Tong
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huilin Zhang
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaofang Yan
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaojun Wang
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fangyuan Wang
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuan Liu
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chenyun Dai
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoping Wan
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Department of Obstetrics and Gynecology, Shanghai First Maternity and Infant Hospital affiliated with Tong Ji University, No. 536, Changle Road, Jing'an District, Shanghai, China.
| |
Collapse
|
47
|
Tong H, Ke JQ, Jiang FZ, Wang XJ, Wang FY, Li YR, Lu W, Wan XP. Tumor-associated macrophage-derived CXCL8 could induce ERα suppression via HOXB13 in endometrial cancer. Cancer Lett 2016; 376:127-36. [PMID: 27018308 DOI: 10.1016/j.canlet.2016.03.036] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 03/21/2016] [Accepted: 03/21/2016] [Indexed: 01/05/2023]
Abstract
PURPOSE To elucidate the role of tumor-associated macrophage (TAM) in the loss of ERα in endometrial cancer (EC) and the underlying mechanism. MATERIALS AND METHODS Tissue microarrays and immunohistochemistry assays were performed using endometrial cancer tissue along with coculture, immunofluorescence, invasion assays and ChIP-qPCR using a human endometrial cancer cell line. RESULTS Compared with normal tissue, an increased number of TAM was found in EC tissue (34.0 ± 2.6 vs. 8.3 ± 1.1, respectively; p < 0.001), which may downregulate ERα (27.4%, p < 0.05 for HEC-1A and 16.9%, p < 0.05 for Ishikawa) and promote EC cell invasion (1.8-fold, p < 0.001 for HEC-1A and 2.0-fold, p < 0.001 for Ishikawa). Furthermore, we found that TAM-derived CXCL8 mediated the loss of ERα and cancer invasion via HOXB13. HOXB13 was highly expressed in the ERα-negative subtype (r = -0.204, p = 0.002) and low expression of ESR1 was associated with a poor prognosis for EC patients (log-rank p < 0.05). CONCLUSION TAM-secreted CXCL8 downregulated the ERα expression of EC cells via HOXB13, which may be associated with cancer invasion, metastasis and poor prognosis.
Collapse
Affiliation(s)
- Huan Tong
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie-Qi Ke
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fei-Zhou Jiang
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao-Jun Wang
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fang-Yuan Wang
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi-Ran Li
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wen Lu
- Department of Obstetrics and Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiao-Ping Wan
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Obstetrics and Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
48
|
Bharti R, Dey G, Mandal M. Cancer development, chemoresistance, epithelial to mesenchymal transition and stem cells: A snapshot of IL-6 mediated involvement. Cancer Lett 2016; 375:51-61. [PMID: 26945971 DOI: 10.1016/j.canlet.2016.02.048] [Citation(s) in RCA: 174] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Revised: 02/24/2016] [Accepted: 02/24/2016] [Indexed: 12/12/2022]
Abstract
Interleukin-6 (IL-6) is a cytokine present in tumor microenvironment. Elevated level of IL-6 is associated with cancer cell proliferation, angiogenesis and metastasis through fueling STAT3, MAPK and Akt signaling. It promotes epithelial to mesenchymal transition (EMT) through altered expression of N-cadherin, vimentin, snail, twist and E-cadherin leading to cancer metastasis. IL-6 boosts mammosphere formation, self-renewal of stem cells, stemness properties of cancer cells and recruitment of mesenchymal stem cells. IL-6 is also a contributing factor for multidrug resistance in cancer due to gp130/MAPK/STAT3 mediated activation of transcription factors C/EBPβ/δ, overexpression of p-glycoprotein, EMT transition and expansion of stem cells. The in-depth investigation of IL-6 mediated cellular effects and its signaling pathway can provide the new window for future research and clinical development of IL-6 targeted therapy in cancer. Thus, an overview is delivered in this review deciphering the emerging aspect of the predominant influence of IL-6 in malignant transformation, EMT, cancer-associated stem cells and chemoresistance.
Collapse
Affiliation(s)
- Rashmi Bharti
- School of Medical Science & Technology, Indian Institute of Technology Kharagpur, Kharagpur 721302, West Bengal, India
| | - Goutam Dey
- School of Medical Science & Technology, Indian Institute of Technology Kharagpur, Kharagpur 721302, West Bengal, India
| | - Mahitosh Mandal
- School of Medical Science & Technology, Indian Institute of Technology Kharagpur, Kharagpur 721302, West Bengal, India.
| |
Collapse
|
49
|
Macrophages: Regulators of the Inflammatory Microenvironment during Mammary Gland Development and Breast Cancer. Mediators Inflamm 2016; 2016:4549676. [PMID: 26884646 PMCID: PMC4739263 DOI: 10.1155/2016/4549676] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 12/21/2015] [Indexed: 12/22/2022] Open
Abstract
Macrophages are critical mediators of inflammation and important regulators of developmental processes. As a key phagocytic cell type, macrophages evolved as part of the innate immune system to engulf and process cell debris and pathogens. Macrophages produce factors that act directly on their microenvironment and also bridge innate immune responses to the adaptive immune system. Resident macrophages are important for acting as sensors for tissue damage and maintaining tissue homeostasis. It is now well-established that macrophages are an integral component of the breast tumor microenvironment, where they contribute to tumor growth and progression, likely through many of the mechanisms that are utilized during normal wound healing responses. Because macrophages contribute to normal mammary gland development and breast cancer growth and progression, this review will discuss both resident mammary gland macrophages and tumor-associated macrophages with an emphasis on describing how macrophages interact with their surrounding environment during normal development and in the context of cancer.
Collapse
|
50
|
Ho LJ, Luo SF, Lai JH. Biological effects of interleukin-6: Clinical applications in autoimmune diseases and cancers. Biochem Pharmacol 2015; 97:16-26. [DOI: 10.1016/j.bcp.2015.06.009] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2015] [Accepted: 06/09/2015] [Indexed: 01/13/2023]
|