1
|
Zhao B, Huo W, Yu X, Shi X, Lv L, Yang Y, Kang J, Li S, Wu H. USP13 promotes breast cancer metastasis through FBXL14-induced Twist1 ubiquitination. Cell Oncol (Dordr) 2023; 46:717-733. [PMID: 36732432 DOI: 10.1007/s13402-023-00779-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/23/2023] [Indexed: 02/04/2023] Open
Abstract
PURPOSE Epithelial-to-mesenchymal transition (EMT) is an important cause of high mortality in breast cancer. Twist1 is one of the EMT transcription factors (EMT-TFs) with a noticeably short half-life, which is regulated by proteasome degradation pathways. Recent studies have found that USP13 stabilizes several specific oncogenic proteins. As yet, however, the relationship between Twist1 and USP13 has not been investigated. METHODS Co-Immunoprecipitation, GST-pulldown, Western blot, qRT-PCR and immunofluorescence assays were used to investigate the role of USP13 in de-ubiquitination of Twist1. Chromatin immunoprecipitation and Luciferase reporter assays were used to investigate the role of Twist1 in inhibiting USP13 reporter transcription. Scratch wound healing, cell migration and invasion assays, and a mouse lung metastases assay were used to investigate the roles of USP13 and Twist1 in promoting breast cancer metastasis. RESULTS We found that Twist1 can be de-ubiquitinated by USP13. In addition, we found that the protein levels of Twist1 dose-dependently increased with USP13 overexpression, while USP13 knockdown resulted in a decreased expression of endogenous Twist1. We also found that USP13 can directly interact with Twist1 and specifically cleave the K48-linked polyubiquitin chains of Twist1 induced by FBXL14. We found that the effect of USP13 in promoting the migration and invasion capacities of breast cancer cells can at least partly be achieved through its regulation of Twist1, while Twist1 can inhibit the transcriptional activity of USP13. CONCLUSIONS Our data indicate that an interplay between Twist1 and USP13 can form a negative physiological feedback loop. Our findings show that USP13 may play an essential role in breast cancer metastasis by regulating Twist1 and, as such, provide a potential target for the clinical treatment of breast cancer.
Collapse
Affiliation(s)
- Binggong Zhao
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Liaoning Province, Dalian University of Technology, Dalian, China
| | - Wei Huo
- Central Hospital affiliated to Dalian University of Technology, Dalian, China
| | - Xiaomin Yu
- Central Hospital affiliated to Dalian University of Technology, Dalian, China
| | - Xiaoxia Shi
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Liaoning Province, Dalian University of Technology, Dalian, China
| | - Linlin Lv
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Liaoning Province, Dalian University of Technology, Dalian, China
| | - Yuxi Yang
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Liaoning Province, Dalian University of Technology, Dalian, China
| | - Jie Kang
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Liaoning Province, Dalian University of Technology, Dalian, China
| | - Shujing Li
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Liaoning Province, Dalian University of Technology, Dalian, China.
| | - Huijian Wu
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Liaoning Province, Dalian University of Technology, Dalian, China.
| |
Collapse
|
2
|
Pan B, Yin S, Peng F, Liu C, Liang H, Su J, Hsiao WLW, Cai Y, Luo D, Xia C. Vorinostat targets UBE2C to reverse epithelial-mesenchymal transition and control cervical cancer growth through the ubiquitination pathway. Eur J Pharmacol 2021; 908:174399. [PMID: 34331954 DOI: 10.1016/j.ejphar.2021.174399] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 07/19/2021] [Accepted: 07/28/2021] [Indexed: 01/16/2023]
Abstract
Vorinostat is a histone deacetylase inhibitor (HDACi) that was demonstrated in our previous study to inhibit the proliferation, migration, and invasion of cervical cancer cells by regulating the PI3K/Akt signaling pathway. However, the molecular mechanism of vorinostat in cervical cancer treatment remains to be further elucidated. A nude mouse xenograft model was established to analyze the antitumor effect of vorinostat in vivo. The combination of iTRAQ-based proteomics and parallel reaction monitoring (PRM) technology has proven to be an efficient and reliable method to identify potential targets for cancer chemotherapy. In this study, 254 differentially expressed proteins in vorinostat-treated cervical cancer cells, among which 180 were upregulated and 74 were downregulated, were identified by using an iTRAQ-based proteomic strategy. Subsequent bioinformatic and PRM analysis of these differentially expressed proteins indicated that UBE2C is a promising target of vorinostat in the inhibition of cervical cancer cell proliferation. We confirmed that the expression of endogenous UBE2C in cervical cancer cell lines was significantly higher than that in normal cervical epithelial cell lines. Additionally, we found that vorinostat downregulated the expression of UBE2C, SQSTM1/p62, N-cadherin, vimentin and upregulated E-cadherin in SiHa and HeLa cells. Our results also showed that vorinostat can downregulate the expression of SQSTM1/p62, N-cadherin, and vimentin during the treatment of cervical cancer cells by regulating UBE2C, while upregulating the expression of E-cadherin. In conclusion, vorinostat reverses epithelial-mesenchymal transition by targeting UBE2C and controls the proliferation of cervical cancer cells through the ubiquitination pathway. UBE2C can be used as a promising target for the development of vorinostat treatment strategies.
Collapse
Affiliation(s)
- Botao Pan
- Foshan Maternal and Child Health Research Institute, South Medical University Affiliated Maternal & Child Health Hospital of Foshan, Foshan, 528000, China
| | - Shuanghong Yin
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Fang Peng
- Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
| | - Chang Liu
- Foshan Maternal and Child Health Research Institute, South Medical University Affiliated Maternal & Child Health Hospital of Foshan, Foshan, 528000, China
| | - Huiyi Liang
- Foshan Maternal and Child Health Research Institute, South Medical University Affiliated Maternal & Child Health Hospital of Foshan, Foshan, 528000, China
| | - Jiyan Su
- Foshan Maternal and Child Health Research Institute, South Medical University Affiliated Maternal & Child Health Hospital of Foshan, Foshan, 528000, China
| | - W L Wendy Hsiao
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science & Technology, Macau, 999078, China
| | - Yantao Cai
- Foshan Maternal and Child Health Research Institute, South Medical University Affiliated Maternal & Child Health Hospital of Foshan, Foshan, 528000, China.
| | - Dixian Luo
- Department of Laboratory Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital), Guangdong, 518000, China.
| | - Chenglai Xia
- Foshan Maternal and Child Health Research Institute, South Medical University Affiliated Maternal & Child Health Hospital of Foshan, Foshan, 528000, China; School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
3
|
Voutsadakis IA. Low-grade serous ovarian carcinoma: an evolution toward targeted therapy. Int J Gynecol Cancer 2020; 30:1619-1626. [PMID: 31780569 DOI: 10.1136/ijgc-2019-000832] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 09/16/2019] [Accepted: 09/24/2019] [Indexed: 11/04/2022] Open
Abstract
Low-grade serous ovarian carcinoma and its high-grade serous ovarian carcinoma counterpart differ in their precursor lesions, molecular profile, natural history, and response to therapies. As such, low-grade serous ovarian carcinoma needs to be studied separately from high-grade serous ovarian carcinoma, despite challenges stemming from its rarity. A deeper understanding of the pathogenesis of low-grade serous ovarian carcinoma and the most common molecular defects and pathways involved in the carcinogenesis of the ovarian epithelium from normal to serous borderline ovarian tumors to low-grade serous ovarian carcinoma will help develop better therapies. By adopting targeted approaches there may be an opportunity to integrate novel therapies without the need for robust numbers in clinical trials. This manuscript will discuss low-grade serous ovarian carcinoma and focus on the arising treatments being developed with an improved understanding of the pathogenesis of this disease.
Collapse
Affiliation(s)
- Ioannis A Voutsadakis
- Section of Internal Medicine Division of Clinical Sciences, Northern Ontario School of Medicine, Sudbury, ON, Canada
| |
Collapse
|
4
|
King N, Kukreja K, Murzabdillaeva A, Ali Y, Willis J, Maiti A, Ma H, Bull J. A rare presentation of carcinosarcoma of the bone in a young female; response with gemcitabine and docetaxel. Clin Sarcoma Res 2019; 9:10. [PMID: 31304003 PMCID: PMC6604449 DOI: 10.1186/s13569-019-0120-7] [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: 03/27/2019] [Accepted: 06/25/2019] [Indexed: 11/17/2022] Open
Abstract
Background Sarcomatoid carcinoma, or carcinosarcoma, is a neoplasm that contains both sarcomatous and carcinomatous elements. It is an extremely rare cancer most often arising from visceral organs. Here we report the seventh documented de novo case of carcinosarcoma of the bone, in a young female who showed initial clinical improvement with gemcitabine and docetaxel. Case presentation A 36-year-old Caucasian female presented with diffuse musculoskeletal pain that had progressed from her shoulder to her back, arm, and knee over 6 months. Imaging revealed diffuse sclerotic lesions of bilateral humeral heads, iliac and ischial bones, and thoracic and lumbar spine. Histopathologic examination of biopsies from the T9 vertebra and left femur showed mainly sarcomatous spindle cells with focal osteoid production. Immunostaining showed the cells to be OSCAR cytokeratin, patchy positive for pankeratin, and negative for CK7, GATA3, S100, SOX10, CD99, EMA, AE1/AE3, and HMW keratin indicative of an epithelial origin. After thorough clinical correlation, sarcomatoid carcinoma of a visceral organ was excluded and the diagnosis of primary sarcomatoid carcinoma of the bone was ultimately favored. She received chemotherapy with gemcitabine and docetaxel, and showed improvement at 6 months but ultimately passed 1 year post diagnosis. Conclusions Primary carcinosarcoma of the bone is an extremely rare malignancy. Early diagnosis is crucial as localized disease may be curable with resection. As shown in this case, combination chemotherapy with gemcitabine and docetaxel is a potential option in patients with unresectable or metastatic disease.
Collapse
Affiliation(s)
- Nicholas King
- 1University of Texas Health Sciences Center at Houston, McGovern Medical School, Houston, TX USA
| | - Keshav Kukreja
- 1University of Texas Health Sciences Center at Houston, McGovern Medical School, Houston, TX USA
| | - Albina Murzabdillaeva
- 1University of Texas Health Sciences Center at Houston, McGovern Medical School, Houston, TX USA
| | - Yasir Ali
- 1University of Texas Health Sciences Center at Houston, McGovern Medical School, Houston, TX USA
| | - Jason Willis
- 2University of Texas Health Sciences Center MD Anderson Cancer Center, Houston, TX USA
| | - Abhishek Maiti
- 2University of Texas Health Sciences Center MD Anderson Cancer Center, Houston, TX USA
| | - Hilary Ma
- 2University of Texas Health Sciences Center MD Anderson Cancer Center, Houston, TX USA
| | - Joan Bull
- 1University of Texas Health Sciences Center at Houston, McGovern Medical School, Houston, TX USA
| |
Collapse
|
5
|
Yin J, Wang Y, Chang J, Li B, Zhang J, Liu Y, Lai S, Jiang Y, Li H, Zeng X. Apelin inhibited epithelial-mesenchymal transition of podocytes in diabetic mice through downregulating immunoproteasome subunits β5i. Cell Death Dis 2018; 9:1031. [PMID: 30301930 PMCID: PMC6178343 DOI: 10.1038/s41419-018-1098-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 09/09/2018] [Accepted: 09/12/2018] [Indexed: 01/12/2023]
Abstract
The epithelial−mesenchymal transition (EMT) of podocytes had been reported to be involved in the glomerular fibrosis in diabetic kidney diseases, which was regulated by TGFβ and NFκB pathways. And apelin, an adipokine which is upregulated in diabetic kidney diseases, was reported to be negatively correlated to TGFβ in polycystic kidney disease and attenuate EMT in renal tubular cells. Therefore, it is hypothesized that apelin might inhibit the EMT of podocytes through downregulating the expression and activation of TGFβ/Smad pathway in diabetic kidney diseases. The results showed that apelin in glomeruli of diabetic mice were increased and exogenous apelin inhibited the EMT of podocytes in diabetic mice, which were accompanied with the decreased expression of proteasome subunits β5i. The results from β5iKO mice confirmed that the inhibiting effects of apelin on EMT of podocytes in diabetic mice were dependent on β5i. The results from culture podocytes showed that apelin decreased the degradation of pIκB and promoted the translocation of IκB into nucleus through decreasing the expression of β5i, which would inhibit the promoting effects of NFκB on expression of TGFβ and followed by decreased activation of Smad pathway and EMT in podocytes. In conclusion, apelin might act as an EMT suppressor for podocytes to decrease the process of glomerular fibrosis in diabetic mice.
Collapse
Affiliation(s)
- Jiming Yin
- Beijing You An Hospital, Capital Medical University, 100069, Beijing, China.,Beijing Institute of Hepatology, 100069, Beijing, China
| | - Yangjia Wang
- Department of Pathology and Pathophysiology, Capital Medical University, 100069, Beijing, China
| | - Jing Chang
- Beijing You An Hospital, Capital Medical University, 100069, Beijing, China
| | - Bin Li
- Department of Pathology and Pathophysiology, Capital Medical University, 100069, Beijing, China
| | - Jia Zhang
- Department of Pathology and Pathophysiology, Capital Medical University, 100069, Beijing, China
| | - Yu Liu
- Department of Pathology and Pathophysiology, Capital Medical University, 100069, Beijing, China
| | - Song Lai
- Department of Cardiology. Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, No.193, Lianhe Road, Xigang District, 116011, Dalian, China
| | - Ying Jiang
- Department of Pathology and Pathophysiology, Capital Medical University, 100069, Beijing, China
| | - Huihua Li
- Department of Cardiology. Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, No.193, Lianhe Road, Xigang District, 116011, Dalian, China
| | - Xiangjun Zeng
- Department of Pathology and Pathophysiology, Capital Medical University, 100069, Beijing, China.
| |
Collapse
|
6
|
Survey of gynecological carcinosarcomas in families with breast and ovarian cancer predisposition. Cancer Genet 2018; 221:38-45. [DOI: 10.1016/j.cancergen.2017.12.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 12/13/2017] [Indexed: 12/21/2022]
|
7
|
Menderes G, Bonazzoli E, Bellone S, Black J, Predolini F, Pettinella F, Masserdotti A, Zammataro L, Altwerger G, Buza N, Hui P, Wong S, Litkouhi B, Ratner E, Silasi DA, Azodi M, Schwartz PE, Santin AD. SYD985, a Novel Duocarmycin-Based HER2-Targeting Antibody-Drug Conjugate, Shows Antitumor Activity in Uterine and Ovarian Carcinosarcoma with HER2/Neu Expression. Clin Cancer Res 2017; 23:5836-5845. [PMID: 28679774 DOI: 10.1158/1078-0432.ccr-16-2862] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 04/25/2017] [Accepted: 06/28/2017] [Indexed: 12/24/2022]
Abstract
Purpose: Carcinosarcomas (CS) are highly aggressive gynecologic malignancies containing both carcinomatous and sarcomatous elements with heterogeneous HER2/neu expression. We compared the efficacy of SYD985 (Synthon Biopharmaceuticals BV), a novel HER2-targeting antibody-drug conjugate (ADC), to trastuzumab emtansine (T-DM1, Genentech-Roche) against primary uterine and ovarian CS.Experimental Design: Eight primary CS cell lines were evaluated for HER2/neu surface expression by IHC and gene amplification by FISH assays. The in vitro experiments included cytotoxicity, antibody-dependent cellular cytotoxicity (ADCC), proliferation, viability, and bystander killing. In vivo activity was studied in mouse xenograft and patient-derived xenograft (PDX) models.Results: SYD985 and T-DM1 induced similar levels of ADCC against CS cell lines with low and high HER2/neu expression when challanged in the presence of effector cells. In contrast, SYD985 was 7- to 54-fold more potent than T-DM1 in the absence of effector cells. SYD985, unlike T-DM1, was active against CS demonstrating low or heterogeneous HER2/neu expression. Specifically, the mean IC50 values were 0.060 μg/mL and 3.221 μg/mL (P < 0.0001) against HER2/neu 0/1+ cell lines and 0.013 μg/mL and 0.096 μg/mL (P < 0.0001) against HER2/neu 3+ cell lines for SYD985 versus T-DM1, respectively. Importantly, unlike T-DM1, SYD985 induced efficient bystander killing of HER2/neu 0/1+ tumor cells admixed with HER2/neu 3+ cells. In vivo studies confirmed that SYD985 is more active than T-DM1 in CS and highly effective against HER2/neu expressing xenografts and PDX.Conclusions: SYD985 may represent a novel and highly effective ADC against HER2-expressing CS. Clinical studies with SYD985 in patients harboring chemotherapy-resistant CS with low/moderate and high HER2 expression are warranted. Clin Cancer Res; 23(19); 5836-45. ©2017 AACR.
Collapse
Affiliation(s)
- Gulden Menderes
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, Connecticut
| | - Elena Bonazzoli
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, Connecticut
| | - Stefania Bellone
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, Connecticut
| | - Jonathan Black
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, Connecticut
| | - Federica Predolini
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, Connecticut
| | - Francesca Pettinella
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, Connecticut
| | - Alice Masserdotti
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, Connecticut
| | - Luca Zammataro
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, Connecticut
| | - Gary Altwerger
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, Connecticut
| | - Natalia Buza
- Department of Pathology, Yale University School of Medicine, Connecticut
| | - Pei Hui
- Department of Pathology, Yale University School of Medicine, Connecticut
| | - Serena Wong
- Department of Pathology, Yale University School of Medicine, Connecticut
| | - Babak Litkouhi
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, Connecticut
| | - Elena Ratner
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, Connecticut
| | - Dan-Arin Silasi
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, Connecticut
| | - Masoud Azodi
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, Connecticut
| | - Peter E Schwartz
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, Connecticut
| | - Alessandro D Santin
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, Connecticut.
| |
Collapse
|
8
|
Schwab CL, English DP, Black J, Bellone S, Lopez S, Cocco E, Bonazzoli E, Bussi B, Predolini F, Ferrari F, Ratner E, Silasi DA, Azodi M, Rutherford T, Schwartz PE, Santin AD. Neratinib shows efficacy in the treatment of HER2 amplified carcinosarcoma in vitro and in vivo. Gynecol Oncol 2015; 139:112-7. [PMID: 26260909 PMCID: PMC4587290 DOI: 10.1016/j.ygyno.2015.08.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 08/03/2015] [Accepted: 08/05/2015] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Carcinosarcoma is a deadly gynecologic malignancy with few effective treatment options. The study of new therapies is difficult because of its rarity. The objective of this study was to determine the efficacy of neratinib in the treatment of HER2 amplified carcinosarcoma. METHODS The efficacy of neratinib in the treatment of HER2 amplified carcinosarcoma was determined in vitro using seven primary carcinosarcoma cell lines with differential expression of HER2/neu. Data regarding IC50, cell cycle distribution, and cell signaling changes were assessed by flow cytometry. The efficacy of neratinib was determined in treating mice harboring HER2 amplified carcinosarcoma xenografts. RESULTS Two of seven (28.5%) carcinosarcoma cell lines were HER2/neu amplified. HER2/neu amplified cell lines SARARK6 and SARARK9 were significantly more sensitive to neratinib than the five non-HER2/neu amplified carcinosarcoma cell lines (mean±SEM IC50:0.014μM±0.004vs.0.164μM±0.019 p=0.0003). Neratinib treatment caused a significant build up in G0/G1 phase of the cell cycle, arrest auto phosphorylation of HER2/neu and activation of S6. Neratinib inhibited tumor growth (p=0.012) and prolonged survival in mice harboring HER2 amplified carcinosarcoma xenografts (p=0.0039). CONCLUSIONS Neratinib inhibits HER2 amplified carcinosarcoma proliferation, signaling, cell cycle progression and tumor growth in vitro. Neratinib inhibits HER2/neu amplified xenograft growth and improves overall survival. Clinical trials are warranted.
Collapse
Affiliation(s)
- Carlton L Schwab
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, 333 Cedar Street New Haven, CT 06520, United States
| | - Diana P English
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, 333 Cedar Street New Haven, CT 06520, United States
| | - Jonathan Black
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, 333 Cedar Street New Haven, CT 06520, United States
| | - Stefania Bellone
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, 333 Cedar Street New Haven, CT 06520, United States
| | - Salvatore Lopez
- Division of Gynecologic Oncology, University Campus Biomedico of Roma, Via Álvaro del Portillo, 21-00128 Rome, Italy
| | - Emiliano Cocco
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, 333 Cedar Street New Haven, CT 06520, United States
| | - Elena Bonazzoli
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, 333 Cedar Street New Haven, CT 06520, United States
| | - Beatrice Bussi
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, 333 Cedar Street New Haven, CT 06520, United States
| | - Federica Predolini
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, 333 Cedar Street New Haven, CT 06520, United States
| | - Francesca Ferrari
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, 333 Cedar Street New Haven, CT 06520, United States
| | - Elena Ratner
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, 333 Cedar Street New Haven, CT 06520, United States
| | - Dan-Arin Silasi
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, 333 Cedar Street New Haven, CT 06520, United States
| | - Masoud Azodi
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, 333 Cedar Street New Haven, CT 06520, United States
| | - Thomas Rutherford
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, 333 Cedar Street New Haven, CT 06520, United States
| | - Peter E Schwartz
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, 333 Cedar Street New Haven, CT 06520, United States
| | - Alessandro D Santin
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, 333 Cedar Street New Haven, CT 06520, United States.
| |
Collapse
|
9
|
Carrarelli P, Yen CF, Arcuri F, Funghi L, Tosti C, Wang TH, Huang JS, Petraglia F. Myostatin, follistatin and activin type II receptors are highly expressed in adenomyosis. Fertil Steril 2015; 104:744-52.e1. [DOI: 10.1016/j.fertnstert.2015.05.032] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 05/13/2015] [Accepted: 05/24/2015] [Indexed: 10/23/2022]
|
10
|
Dunnick JK, Sanders JM, Kissling GE, Johnson CL, Boyle MH, Elmore SA. Environmental chemical exposure may contribute to uterine cancer development: studies with tetrabromobisphenol A. Toxicol Pathol 2015; 43:464-73. [PMID: 25476797 PMCID: PMC6706771 DOI: 10.1177/0192623314557335] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Tetrabromobisphenol A (TBBPA), a widely used flame retardant, caused uterine tumors in rats. In this study, TBBPA was administered to male and female Wistar Han rats and B6C3F1/N mice by oral gavage in corn oil for 2 years at doses up to 1,000 mg/kg. TBBPA induced uterine epithelial tumors including adenomas, adenocarcinomas, and malignant mixed Müllerian tumors (MMMTs). In addition, endometrial epithelial atypical hyperplasia occurred in TBBPA-treated rats. Also found to be related to TBBPA treatment, but at lower incidence and at a lower statistical significance, were testicular tumors in rats, and hepatic tumors, hemangiosarcomas (all organs), and intestinal tumors in male mice. It is hypothesized that the TBBPA uterine tumor carcinogenic mechanisms involve altered estrogen levels and/or oxidative damage. TBBPA treatment may affect hydroxysteroid-dehydrogenase-17β (HSD17β) and/or sulfotransferases, enzymes involved in estrogen homeostasis. Metabolism of TBBPA may also result in the formation of free radicals. The finding of TBBPA-mediated uterine cancer in rats is of concern because TBBPA exposure is widespread and endometrial tumors are a common malignancy in women. Further work is needed to understand TBBPA cancer mechanisms.
Collapse
Affiliation(s)
- J K Dunnick
- National Institute of Environmental Health Sciences, National Toxicology Program, Research Triangle Park, North Carolina, USA
| | - J M Sanders
- National Cancer Institute, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - G E Kissling
- National Institute of Environmental Health Sciences, National Toxicology Program, Research Triangle Park, North Carolina, USA
| | - C L Johnson
- Charles River Laboratories, Pathology Associates, Research Triangle Park, North Carolina, USA
| | - M H Boyle
- Integrated Laboratory Systems, Research Triangle Park, North Carolina, USA
| | - S A Elmore
- National Institute of Environmental Health Sciences, National Toxicology Program, Research Triangle Park, North Carolina, USA
| |
Collapse
|
11
|
|
12
|
Miura Y, Hagiwara N, Radisky DC, Hirai Y. CCAAT/enhancer binding protein beta (C/EBPβ) isoform balance as a regulator of epithelial-mesenchymal transition in mouse mammary epithelial cells. Exp Cell Res 2014; 327:146-55. [PMID: 24881817 DOI: 10.1016/j.yexcr.2014.05.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 05/03/2014] [Accepted: 05/21/2014] [Indexed: 12/22/2022]
Abstract
Activation of the epithelial-mesenchymal transition (EMT) program promotes cell invasion and metastasis, and is reversed through mesenchymal-epithelial transition (MET) after formation of distant metastases. Here, we show that an imbalance of gene products encoded by the transcriptional factor C/EBPβ, LAP (liver-enriched activating protein) and LIP (liver-enriched inhibitory protein), can regulate both EMT- and MET-like phenotypic changes in mouse mammary epithelial cells. By using tetracycline repressive LIP expression constructs, we found that SCp2 cells, a clonal epithelial line of COMMA1-D cells, expressed EMT markers, lost the ability to undergo alveolar-like morphogenesis in 3D Matrigel, and acquired properties of benign adenoma cells. Conversely, we found that inducible expression of LAP in SCg6 cells, a clonal fibroblastic line of COMMA1-D cells, began to express epithelial keratins with suppression of proliferation. The overexpression of the C/EBPβ gene products in these COMMA1-D derivatives was suppressed by long-term cultivation on tissue culture plastic, but gene expression was maintained in cells grown on Matrigel or exposed to proteasome inhibitors. Thus, imbalances of C/EBPβ gene products in mouse mammary epithelial cells, which are affected by contact with basement membrane, are defined as a potential regulator of metastatic potential.
Collapse
Affiliation(s)
- Yuka Miura
- Department of Bioscience, Graduate School of Science and Technology, Kwansei Gakuin University, Hyogo, 2-1 Gakuen, Sanda 669-1337 Japan
| | - Natsumi Hagiwara
- Department of Bioscience, Graduate School of Science and Technology, Kwansei Gakuin University, Hyogo, 2-1 Gakuen, Sanda 669-1337 Japan
| | - Derek C Radisky
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32225 USA
| | - Yohei Hirai
- Department of Bioscience, Graduate School of Science and Technology, Kwansei Gakuin University, Hyogo, 2-1 Gakuen, Sanda 669-1337 Japan.
| |
Collapse
|
13
|
Voutsadakis IA. Large cell neuroendocrine carcinoma of the ovary: A pathologic entity in search of clinical identity. World J Clin Oncol 2014; 5:36-38. [PMID: 24829849 PMCID: PMC4014794 DOI: 10.5306/wjco.v5.i2.36] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 02/12/2014] [Accepted: 04/16/2014] [Indexed: 02/06/2023] Open
Abstract
Large cell neuroendocrine carcinoma (LCNEC) of the ovary is a rare diagnosis and only a few dozen cases have been reported in the literature. It is characterized by large pleiomorphic cells with large round or oval nuclei, presence of mitoses and staining for neuroendocrine (NE) markers such as chromogranin A, synaptophysin, neuron specific enolase. This editorial gives a brief overview of this histologic type of ovarian carcinomas. LCNEC of the ovary is a pathologic entity that may not be diagnosed purely on clinical grounds due to the similarity of its clinical features with those of the more common epithelial ovarian cancers. Nevertheless the diagnosis is worth-making from a practical point of view in order to consider treatments tailored towards the NE component if it is dominant or it becomes dominant during the natural evolution of the disease. Establishment of an international tumor registry with an accompanying tumor tissue bank of ovarian LCNEC could be a means of obtaining further knowledge on clinical characteristics and advance research on this rare entity. This will further inform on treatment strategies and could identify future molecular treatment targets.
Collapse
|
14
|
Semczuk A, Ignatov A, Obrzut B, Reventos J, Rechberger T. Role of p53 Pathway Alterations in Uterine Carcinosarcomas (Malignant Mixed Müllerian Tumors). Oncology 2014; 87:193-204. [DOI: 10.1159/000363574] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2013] [Accepted: 05/07/2014] [Indexed: 01/10/2023]
|