1
|
Riaz MA, Kary FL, Jensen A, Zeppernick F, Meinhold-Heerlein I, Konrad L. Long-Term Maintenance of Viable Human Endometrial Epithelial Cells to Analyze Estrogen and Progestin Effects. Cells 2024; 13:811. [PMID: 38786035 PMCID: PMC11120542 DOI: 10.3390/cells13100811] [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: 03/20/2024] [Revised: 05/03/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024] Open
Abstract
There are fewer investigations conducted on human primary endometrial epithelial cells (HPEECs) compared to human primary endometrial stromal cells (HPESCs). One of the main reasons is the scarcity of protocols enabling prolonged epithelial cell culture. Even though it is possible to culture HPEECs in 3D over a longer period of time, it is technically demanding. In this study, we successfully established a highly pure, stable, and long-term viable human conditionally reprogrammed endometrial epithelial cell line, designated as eCRC560. These cells stained positive for epithelial markers, estrogen and progesterone receptors, and epithelial cell-cell contacts but negative for stromal and endothelial cell markers. Estradiol (ES) reduced the abundance of ZO-1 in a time- and dose-dependent manner, in contrast to the dose-dependent increase with the progestin dienogest (DNG) when co-cultured with HPESCs. Moreover, ES significantly increased cell viability, cell migration, and invasion of the eCRC560 cells; all these effects were inhibited by pretreatment with DNG. DNG withdrawal led to a significantly disrupted monolayer of eCRC560 cells in co-culture with HPESCs, yet it markedly increased the adhesion of eCRC560 to the human mesothelial MeT-5A cells. The long-term viable eCRC560 cells are suitable for in vitro analysis of HPEECs to study the epithelial compartment of the human endometrium and endometrial pathologies.
Collapse
Affiliation(s)
- Muhammad Assad Riaz
- Institute of Gynecology and Obstetrics, Faculty of Medicine, Justus Liebig University Giessen, 35392 Giessen, Germany; (M.A.R.); (F.L.K.); (F.Z.); (I.M.-H.)
| | - Franziska Louisa Kary
- Institute of Gynecology and Obstetrics, Faculty of Medicine, Justus Liebig University Giessen, 35392 Giessen, Germany; (M.A.R.); (F.L.K.); (F.Z.); (I.M.-H.)
| | - Alexandra Jensen
- Institute of Radiooncology and Radiotherapy, Clinic Fulda, 36043 Fulda, Germany;
| | - Felix Zeppernick
- Institute of Gynecology and Obstetrics, Faculty of Medicine, Justus Liebig University Giessen, 35392 Giessen, Germany; (M.A.R.); (F.L.K.); (F.Z.); (I.M.-H.)
| | - Ivo Meinhold-Heerlein
- Institute of Gynecology and Obstetrics, Faculty of Medicine, Justus Liebig University Giessen, 35392 Giessen, Germany; (M.A.R.); (F.L.K.); (F.Z.); (I.M.-H.)
| | - Lutz Konrad
- Institute of Gynecology and Obstetrics, Faculty of Medicine, Justus Liebig University Giessen, 35392 Giessen, Germany; (M.A.R.); (F.L.K.); (F.Z.); (I.M.-H.)
| |
Collapse
|
2
|
Harvey BJ, Harvey HM. Sex Differences in Colon Cancer: Genomic and Nongenomic Signalling of Oestrogen. Genes (Basel) 2023; 14:2225. [PMID: 38137047 PMCID: PMC10742859 DOI: 10.3390/genes14122225] [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: 11/22/2023] [Revised: 12/10/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023] Open
Abstract
Colon cancer (CRC) is a prevalent malignancy that exhibits distinct differences in incidence, prognosis, and treatment responses between males and females. These disparities have long been attributed to hormonal differences, particularly the influence of oestrogen signalling. This review aims to provide a comprehensive analysis of recent advances in our understanding of the molecular mechanisms underlying sex differences in colon cancer and the protective role of membrane and nuclear oestrogen signalling in CRC development, progression, and therapeutic interventions. We discuss the epidemiological and molecular evidence supporting sex differences in colon cancer, followed by an exploration of the impact of oestrogen in CRC through various genomic and nongenomic signalling pathways involving membrane and nuclear oestrogen receptors. Furthermore, we examine the interplay between oestrogen receptors and other signalling pathways, in particular the Wnt/β-catenin proliferative pathway and hypoxia in shaping biological sex differences and oestrogen protective actions in colon cancer. Lastly, we highlight the potential therapeutic implications of targeting oestrogen signalling in the management of colon cancer and propose future research directions to address the current gaps in our understanding of this complex phenomenon.
Collapse
Affiliation(s)
- Brian J. Harvey
- Faculty of Medicine, Royal College of Surgeons in Ireland, RCSI University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland
| | - Harry M. Harvey
- Princess Margaret Cancer Centre, Toronto, ON M5G 1Z5, Canada;
| |
Collapse
|
3
|
Jiménez-Salazar JE, Rivera-Escobar RM, Damián-Ferrara R, Maldonado-Cubas J, Rincón-Pérez C, Tarragó-Castellanos R, Damián-Matsumura P. Estradiol-Induced Epithelial to Mesenchymal Transition and Migration Are Inhibited by Blocking c-Src Kinase in Breast Cancer Cell Lines. J Breast Cancer 2023; 26:446-460. [PMID: 37704382 PMCID: PMC10625871 DOI: 10.4048/jbc.2023.26.e37] [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: 02/14/2023] [Revised: 06/18/2023] [Accepted: 07/19/2023] [Indexed: 09/15/2023] Open
Abstract
PURPOSE The epithelial-to-mesenchymal transition (EMT) is the main event that favors cell migration and metastasis in breast cancer. Previously, we demonstrated that 1 nM estradiol (E2) promotes EMT, induced by c-Src kinase, causing changes in the localization of proteins that compose the tight junction (TJ) and adherens junction (AJ). METHODS The present work highlights the central role of c-Src in the initiation of metastasis, induced by E2, through increasing the ability of MCF-7 and T47-D cells, which express estrogen receptor alpha (ERα), to migrate and invade before they become metastatic. RESULTS Treatment with E2 can activate two signaling pathways, the first one by the phosphorylated c-Src (p-Src) which forms the p-Src/E-cadherin complex. This phenomenon was completely prevented by incubation with a selective inhibitor of c-Src (5 µM PP2). p-Src then promotes the downregulation of E-cadherin and occludin, which are epithelial phenotype marker proteins of the AJ and TJ, respectively. In the second pathway, E2 binds to ERα, creating a complex that translocates to the nucleus, inducing the synthesis of SNAIL1 and N-cadherin proteins, markers of the mesenchymal phenotype. Both processes increased the migratory and invasive capacities of both cell lines. CONCLUSION The present study demonstrate that E2 enhance EMT and migration, through c-Src activation, in human breast cancer cells that express ERα and become potential therapeutic targets.
Collapse
Affiliation(s)
- Javier E Jiménez-Salazar
- Department of Biology of Reproduction, Biological Sciences and Health Division (DCBS), Autonomous Metropolitan University (UAM), Mexico City, México
- Escuela Militar de Graduados de Sanidad, Secretaría de la Defensa Nacional (SEDENA), Mexico City, México
| | - Rene M Rivera-Escobar
- Department of Biology of Reproduction, Biological Sciences and Health Division (DCBS), Autonomous Metropolitan University (UAM), Mexico City, México
| | - Rebeca Damián-Ferrara
- Monterrey Institute of Technology and Higher Education (ITESM), School of Engineering and Sciences, Monterrey, México
| | | | - Catalina Rincón-Pérez
- Escuela Militar de Graduados de Sanidad, Secretaría de la Defensa Nacional (SEDENA), Mexico City, México
| | - Rosario Tarragó-Castellanos
- Department of Biology of Reproduction, Biological Sciences and Health Division (DCBS), Autonomous Metropolitan University (UAM), Mexico City, México
| | - Pablo Damián-Matsumura
- Department of Biology of Reproduction, Biological Sciences and Health Division (DCBS), Autonomous Metropolitan University (UAM), Mexico City, México.
| |
Collapse
|
4
|
Holmes J, Gaber M, Jenks MZ, Wilson A, Loy T, Lepetit C, Vitolins MZ, Herbert BS, Cook KL, Vidi PA. Reversion of breast epithelial polarity alterations caused by obesity. NPJ Breast Cancer 2023; 9:35. [PMID: 37160903 PMCID: PMC10170133 DOI: 10.1038/s41523-023-00539-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 04/21/2023] [Indexed: 05/11/2023] Open
Abstract
Molecular links between breast cancer risk factors and pro-oncogenic tissue alterations are poorly understood. The goal of this study was to characterize the impact of overweight and obesity on tissue markers of risk, using normal breast biopsies, a mouse model of diet-induced obesity, and cultured breast acini. Proliferation and alteration of epithelial polarity, both necessary for tumor initiation, were quantified by immunostaining. High BMI (>30) and elevated leptin were associated with compromised epithelial polarity whereas overweight was associated with a modest increase in proliferation in human and mice mammary glands. Human serum with unfavorable adipokine levels altered epithelial polarization of cultured acini, recapitulating the effect of leptin. Weight loss in mice led to metabolic improvements and restored epithelial polarity. In acini cultures, alteration of epithelial polarity was prevented by antioxidants and could be reverted by normalizing culture conditions. This study shows that obesity and/or dietary factors modulate tissue markers of risk. It provides a framework to set target values for metabolic improvements and to assess the efficacy of interventional studies aimed at reducing breast cancer risk.
Collapse
Affiliation(s)
- Julia Holmes
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Mohamed Gaber
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Mónica Z Jenks
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Adam Wilson
- Department of Surgery, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Tucker Loy
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | | | - Mara Z Vitolins
- Department of Epidemiology and Prevention, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Brittney-Shea Herbert
- Department of Medical & Molecular Genetics, IU School of Medicine, Indianapolis, IN, 46202, USA
| | - Katherine L Cook
- Department of Surgery, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
- Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC, USA
| | - Pierre-Alexandre Vidi
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA.
- Institut de Cancérologie de l'Ouest, Angers, 49055, France.
- Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC, USA.
| |
Collapse
|
5
|
Mittler F, Obeïd P, Haguet V, Allier C, Gerbaud S, Rulina AV, Gidrol X, Balakirev MY. Mechanical stress shapes the cancer cell response to neddylation inhibition. J Exp Clin Cancer Res 2022; 41:115. [PMID: 35354476 PMCID: PMC8966269 DOI: 10.1186/s13046-022-02328-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 03/13/2022] [Indexed: 12/28/2022] Open
Abstract
Background The inhibition of neddylation by the preclinical drug MLN4924 represents a new strategy to combat cancer. However, despite being effective against hematologic malignancies, its success in solid tumors, where cell–cell and cell-ECM interactions play essential roles, remains elusive. Methods Here, we studied the effects of MLN4924 on cell growth, migration and invasion in cultured prostate cancer cells and in disease-relevant prostate tumoroids. Using focused protein profiling, drug and RNAi screening, we analyzed cellular pathways activated by neddylation inhibition. Results We show that mechanical stress induced by MLN4924 in prostate cancer cells significantly affects the therapeutic outcome. The latter depends on the cell type and involves distinct Rho isoforms. In LNCaP and VCaP cells, the stimulation of RhoA and RhoB by MLN4924 markedly upregulates the level of tight junction proteins at cell–cell contacts, which augments the mechanical strain induced by Rho signaling. This “tight junction stress response” (TJSR) causes the collapse of cell monolayers and a characteristic rupture of cancer spheroids. Notably, TJSR is a major cause of drug-induced apoptosis in these cells. On the other hand, in PC3 cells that underwent partial epithelial-to-mesenchymal transition (EMT), the stimulation of RhoC induces an adverse effect by promoting amoeboid cell scattering and invasion. We identified complementary targets and drugs that allow for the induction of TJSR without stimulating RhoC. Conclusions Our finding that MLN4924 acts as a mechanotherapeutic opens new ways to improve the efficacy of neddylation inhibition as an anticancer approach. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s13046-022-02328-y.
Collapse
|
6
|
Huang T, Bao H, Meng YH, Zhu JL, Chu XD, Chu XL, Pan JH. Tumour budding is a novel marker in breast cancer: the clinical application and future prospects. Ann Med 2022; 54:1303-1312. [PMID: 35535687 PMCID: PMC9103277 DOI: 10.1080/07853890.2022.2070272] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/27/2022] [Accepted: 04/20/2022] [Indexed: 11/30/2022] Open
Abstract
Breast cancer (BC) is a group of markedly heterogeneous tumours. There are many subtypes with different biological behaviours and clinicopathological characteristics, leading to significantly different prognosis. Despite significant advances in the treatment of BC, early metastatic is a critical factor for poor prognosis in BC patients. Tumour budding (TB) is considered as the first step process of tumour metastasis and is related to the epithelial-mesenchymal transition (EMT). TB has been observed in a variety of cancers, such as colorectal and gastric cancer, and had been considered as a distinct clinicopathological characteristics for early metastasis. However, TB evaluation standards and clinical application are not uniform in BC, as well as its molecular mechanism is not fully understood. Here, we reviewed the interpretation criteria, mechanism, clinicopathological characteristics and clinical application prospects of TB in BC. Key messagesCurrently, tumour budding is a poor prognosis for various solid tumours, also in breast cancer.Tumour budding is based on epithelial-mesenchymal transition and tumour microenvironment factors and is presumed to be an early step in the metastatic process.Breast cancer tumour budding still needs multi-centre experiments. We summarize the current research on breast cancer tumour budding, analyse the method of discriminating breast cancer tumour budding and explore the prognostic role and mechanism in breast cancer.
Collapse
Affiliation(s)
- Ting Huang
- Department of Clinical Pathology, the First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Hui Bao
- Department of Plastic Surgery, the First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yu-hua Meng
- Department of Clinical Pathology, the First People's Hospital of Shunde, Foshan, China
| | - Jian-lin Zhu
- Department of General Surgery, the First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xiao-dong Chu
- Department of General Surgery, the First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xiao-li Chu
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong, China
- Guangdong Provincial Key laboratory of Chinese Medicine for Prevention and Treatment of Refractory Chronic Diseases, Guangzhou, China
| | - Jing-hua Pan
- Department of General Surgery, the First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
7
|
He X, Zhang L, Hu L, Liu S, Xiong A, Wang J, Xiong Y, Li G. PM2.5 Aggravated OVA-Induced Epithelial Tight Junction Disruption Through Fas Associated via Death Domain-Dependent Apoptosis in Asthmatic Mice. J Asthma Allergy 2021; 14:1411-1423. [PMID: 34848976 PMCID: PMC8612670 DOI: 10.2147/jaa.s335590] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 11/03/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Exposure to air pollutants cause exacerbation of asthma, but the experimental evidence and the mechanisms still need to be collected and addressed. METHODS Asthma model was constructed by ovalbumin (OVA) combined with or without airborne fine particulate matter 2.5 (PM2.5) exposure. Lung sections were stained by hematoxylin-eosin staining (H&E) and Masson's trichrome. RNA-seq and gene set enrichment analysis (GSEA) was performed to identify the key pathway. TdT mediated dUTP Nick End Labeling (TUNEL) assay, real-time qPCR, Western blot, immunofluorescence and lentivirus transfection were applied for mechanism discovery. RESULTS In this study, we found PM2.5 aggravated airway inflammation in OVA-induced asthmatic mice. RNA-seq analysis also showed that epithelial mesenchymal transition (EMT) was enhanced in OVA-induced mice exposed to PM2.5 compared with that in OVA-induced mice. In the meantime, we observed that apoptosis was significantly increased in asthmatic mice exposed to PM2.5 by using GSEA analysis, which was validated by TUNEL assay. By using bioinformatic analysis, Fas associated via death domain (FADD), a new actor in innate immunity and inflammation, was identified to be related to apoptosis, EMT and tight junction. Furthermore, we found that the transcript and protein levels of tight junction markers, E-cadherin, zonula occludens (ZO)-1 and Occludin, were decreased after PM2.5 exposure in vivo and in vitro by using RT-qPCR and immunofluorescence, with the increased expression of FADD. Moreover, down-regulation of FADD attenuated PM2.5-induced apoptosis and tight junction disruption in human airway epithelial cells. CONCLUSION Taken together, we demonstrated that PM2.5 aggravated epithelial tight junction disruption through apoptosis mediated by up-regulation of FADD in OVA-induced model.
Collapse
Affiliation(s)
- Xiang He
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, The Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, People’s Republic of China
- Department of Pulmonary and Critical Care Medicine, Chengdu Third People’s Hospital Branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu, 610031, People’s Republic of China
| | - Lei Zhang
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, The Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, People’s Republic of China
- Department of Pulmonary and Critical Care Medicine, Chengdu Third People’s Hospital Branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu, 610031, People’s Republic of China
| | - Lingjuan Hu
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, The Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, People’s Republic of China
- Department of Respiratory Disease, Renshou County People’s Hospital, Renshou, 620550, People’s Republic of China
| | - Shengbin Liu
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, The Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, People’s Republic of China
- Department of Pulmonary and Critical Care Medicine, Chengdu Third People’s Hospital Branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu, 610031, People’s Republic of China
| | - Anying Xiong
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, The Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, People’s Republic of China
- Department of Pulmonary and Critical Care Medicine, Chengdu Third People’s Hospital Branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu, 610031, People’s Republic of China
| | - Junyi Wang
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, The Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, People’s Republic of China
- Department of Pulmonary and Critical Care Medicine, Chengdu Third People’s Hospital Branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu, 610031, People’s Republic of China
| | - Ying Xiong
- Department of Pulmonary and Critical Care Medicine, Sichuan Friendship Hospital, Chengdu, 610000, People’s Republic of China
| | - Guoping Li
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, The Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, People’s Republic of China
- Department of Pulmonary and Critical Care Medicine, Chengdu Third People’s Hospital Branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu, 610031, People’s Republic of China
| |
Collapse
|
8
|
Jiménez-Salazar JE, Damian-Ferrara R, Arteaga M, Batina N, Damián-Matsumura P. Non-Genomic Actions of Estrogens on the DNA Repair Pathways Are Associated With Chemotherapy Resistance in Breast Cancer. Front Oncol 2021; 11:631007. [PMID: 33869016 PMCID: PMC8044931 DOI: 10.3389/fonc.2021.631007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 01/27/2021] [Indexed: 12/12/2022] Open
Abstract
Estrogens have been implicated in the etiology of breast cancer for a long time. It has been stated that long-term exposure to estrogens is associated with a higher incidence of breast cancer, since estradiol (E2) stimulates breast cell growth; however, its effect on DNA damage/repair is only starting to be investigated. Recent studies have documented that estrogens are able to modify the DNA damage response (DDR) and DNA repair mechanisms. On the other hand, it has been proposed that DDR machinery can be altered by estrogen signaling pathways, that can be related to cancer progression and chemoresistance. We have demonstrated that E2 promotes c-Src activation and breast cancer cell motility, through a non-genomic pathway. This review discusses scientific evidence supporting this non-genomic mechanism where estrogen modifies the DNA repair pathways, and its relationship to potential causes of chemoresistance.
Collapse
Affiliation(s)
- Javier E Jiménez-Salazar
- Department of Biology of Reproduction, Division of Biological Sciences and Health (DCBS), Autonomous Metropolitan University (UAM), Mexico City, Mexico.,School of Medicine, National Autonomous University of Mexico (UNAM), Mexico City, Mexico
| | - Rebeca Damian-Ferrara
- Monterrey Institute of Technology and Higher Education (ITESM), School of Engineering and Sciences, Monterrey, Mexico
| | - Marcela Arteaga
- Department of Biology of Reproduction, Division of Biological Sciences and Health (DCBS), Autonomous Metropolitan University (UAM), Mexico City, Mexico
| | - Nikola Batina
- Nanotechnology and Molecular Engineering Laboratory, Department of Chemistry, Division of Basic Science and Engineering (DCBI), Autonomous Metropolitan University (UAM), Mexico City, Mexico
| | - Pablo Damián-Matsumura
- Department of Biology of Reproduction, Division of Biological Sciences and Health (DCBS), Autonomous Metropolitan University (UAM), Mexico City, Mexico
| |
Collapse
|
9
|
Li Y, Li J, Shen Y, Xiong Y, Li X, Qin Z. Identification of estrogen receptor target genes involved in gonadal feminization caused by estrogen in Xenopus laevis. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2021; 232:105760. [PMID: 33515924 DOI: 10.1016/j.aquatox.2021.105760] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/10/2021] [Accepted: 01/15/2021] [Indexed: 06/12/2023]
Abstract
Estrogens and estrogenic endocrine disrupting chemicals can cause gonadal feminization in some vertebrates mainly through estrogen receptor (ER), but the underlying molecular mechanisms are unclear. The present study aimed to identify ER target genes involved in estrogen-caused gonadal feminization in Xenopus laevis. Based on our recent transcriptomic data that 10 nM 17β-estradiol (E2) altered gene transcription in feminizing gonads of male X. laevis at NF stages 48, 50, and 52, we searched estrogen response element (ERE) using the Dragon ERE Finder software in the promoter region of all the E2-regulated genes. As a result, 163 genes containing ERE sequence were identified as predicted ER target genes at NF stage 50 (on the 14th day postfertilization), a crucial stage for gonadal feminization. Then, some of these predicted ER target genes were further investigated, mainly including the genes that were suggested to be involved in E2-caused gonadal feminization and genes being dramatically up or down-regulated by E2. Fifteen genes were demonstrated to be responsive to E2, in turn ER antagonist blocked the E2-regulated transcription. Finally, we identified 10 genes that can bind to ERα by a chromatin immunoprecipitation-qPCR. Taken together, we identified the 10 genes that contain predicted ERE sequences, are responsive to estrogen and ER antagonist, and have ability to bind to ER as ER target genes, including pglyrp2, apoa1, fgb, tdo2, ca6, nags, cpb2, tmprss6, nudc, zwilch. Our results could help to improve the understanding of the molecular mechanisms for gonadal feminization caused by estrogenic endocrine disrupting chemicals in X. laevis, and even in other species.
Collapse
Affiliation(s)
- Yuanyuan Li
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jinbo Li
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yanping Shen
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yiming Xiong
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xinghong Li
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhanfen Qin
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
10
|
Expression of Tight Junction Proteins Is Altered in Bladder Cancer. ACTA ACUST UNITED AC 2020; 2020:6341256. [PMID: 33282635 PMCID: PMC7685791 DOI: 10.1155/2020/6341256] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 11/03/2020] [Indexed: 11/17/2022]
Abstract
Bladder cancer (BC) is one of the tumors which occur most frequently in urological system, but less is known about the expression of tight junction proteins and its clinical significance in BC. In this study, expression of claudin-4, zonula occludens-1 (ZO-1) and zonula occludens-1 nucleic acid-binding protein (ZONAB), in BC tissues, adjacent nontumor tissue (ANTT), and BC cell lines was examined by Western blotting, semiquantitative RT-PCR, and immunohistochemistry, and then, the clinical significance of these proteins was investigated. The mRNA and protein expression of ZONAB were significantly upregulated, while those of ZO-1 was significantly downregulated in some BC cell lines and tissues in comparison with nontumor urothelial cell lines and ANTT. High expression rate of ZO-1 and ZONAB had negative correlation in BC tissues and was also correlated with muscle-invasive lesions in BC tissues. In conclusion, the expression of tight junction proteins is significantly altered in BC and ZO-1, and ZONAB interaction might be involved in BC development.
Collapse
|
11
|
Role of tight junctions in the epithelial-to-mesenchymal transition of cancer cells. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1863:183503. [PMID: 33189716 DOI: 10.1016/j.bbamem.2020.183503] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 12/15/2022]
Abstract
The epithelial-mesenchymal transition (EMT) is an essential step in cancer progression. Epithelial cells possess several types of cell-cell junctions, and tight junctions are known to play important roles in maintaining the epithelial program. EMT is characterized by a loss of epithelial markers, including E-cadherin and tight junction proteins. Somewhat surprisingly, the evidence is accumulating that upregulated expression of tight junction proteins plays an important role in the EMT of cancer cells. Tight junctions have distinct tissue-specific and cancer-specific regulatory mechanisms, enabling them to play different roles in EMT. Tight junctions and related signaling pathways are attractive targets for cancer treatments; signal transduction inhibitors and monoclonal antibodies for tight junction proteins may be used to suppress EMT, invasion, and metastasis. Here we review the role of bicellular and tricellular tight junction proteins during EMT. Further investigation of regulatory mechanisms of tight junctions during EMT in cancer cells will inform the development of biomarkers for predicting prognosis as well as novel therapies.
Collapse
|
12
|
González-Mariscal L, Miranda J, Gallego-Gutiérrez H, Cano-Cortina M, Amaya E. Relationship between apical junction proteins, gene expression and cancer. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183278. [PMID: 32240623 DOI: 10.1016/j.bbamem.2020.183278] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 01/09/2020] [Accepted: 03/06/2020] [Indexed: 12/11/2022]
Abstract
The apical junctional complex (AJC) is a cell-cell adhesion system present at the upper portion of the lateral membrane of epithelial cells integrated by the tight junction (TJ) and the adherens junction (AJ). This complex is crucial to initiate and stabilize cell-cell adhesion, to regulate the paracellular transit of ions and molecules and to maintain cell polarity. Moreover, we now consider the AJC as a hub of signal transduction that regulates cell-cell adhesion, gene transcription and cell proliferation and differentiation. The molecular components of the AJC are multiple and diverse and depending on the cellular context some of the proteins in this complex act as tumor suppressors or as promoters of cell transformation, migration and metastasis outgrowth. Here, we describe these new roles played by TJ and AJ proteins and their potential use in cancer diagnostics and as targets for therapeutic intervention.
Collapse
Affiliation(s)
- Lorenza González-Mariscal
- Department of Physiology, Biophysics and Neuroscience, Center of Research and Advanced Studies (Cinvestav), Mexico City, Mexico.
| | - Jael Miranda
- Department of Physiology, Biophysics and Neuroscience, Center of Research and Advanced Studies (Cinvestav), Mexico City, Mexico
| | - Helios Gallego-Gutiérrez
- Department of Physiology, Biophysics and Neuroscience, Center of Research and Advanced Studies (Cinvestav), Mexico City, Mexico
| | - Misael Cano-Cortina
- Department of Physiology, Biophysics and Neuroscience, Center of Research and Advanced Studies (Cinvestav), Mexico City, Mexico
| | - Elida Amaya
- Department of Physiology, Biophysics and Neuroscience, Center of Research and Advanced Studies (Cinvestav), Mexico City, Mexico
| |
Collapse
|
13
|
Lara-Cruz C, Jiménez-Salazar JE, Arteaga M, Arredondo M, Ramón-Gallegos E, Batina N, Damián-Matsumura P. Gold nanoparticle uptake is enhanced by estradiol in MCF-7 breast cancer cells. Int J Nanomedicine 2019; 14:2705-2718. [PMID: 31118607 PMCID: PMC6503330 DOI: 10.2147/ijn.s196683] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Accepted: 03/06/2019] [Indexed: 12/31/2022] Open
Abstract
Purpose: In the present study, we investigated the effects of 17β-estradiol (E2) on membrane roughness and gold nanoparticle (AuNP) uptake in MCF-7 breast cancer cells. Methods: Estrogen receptor (ER)-positive breast cancer cells (MCF-7) were exposed to bare 20 nm AuNPs in the presence and absence of 1×10-9 M E2 for different time intervals for up to 24 hrs. The effects of AuNP incorporation and E2 incubation on the MCF-7 cell surface roughness were measured using atomic force microscopy (AFM). Endocytic vesicle formation was studied using confocal laser scanning microscopy (CLSM). Finally, the results were confirmed by hyperspectral optical microscopy. Results: High-resolution AFM images of the surfaces of MCF-7 membranes (up to 250 nm2) were obtained. The incubation of cells for 12 hrs with AuNP and E2 increased the cell membrane roughness by 95% and 30% compared with the groups treated with vehicle (ethanol) or AuNPs only, respectively. This effect was blocked by an ER antagonist (7α,17β-[9-[(4,4,5,5,5-Pentafluoropentyl)sulfinyl]nonyl]estra-1,3,5(10)-triene-3,17-diol [ICI] 182,780). Higher amounts of AuNPs were localized inside MCF-7 cells around the nucleus, even after 6 hrs of E2 incubation, compared with vehicle-treated cells. Endolysosome formation was induced by E2, which may be associated with an increase in AuNP-uptake. Conclusions: E2 enhances AuNP incorporation in MCF-7 cells by modulating of plasma membrane roughness and inducing lysosomal endocytosis. These findings provide new insights into combined nanotherapies and hormone therapies for breast cancer.
Collapse
Affiliation(s)
- Carlos Lara-Cruz
- Nanotechnology and Molecular Engineering Laboratory, Department of Chemistry, Division of Basic Science and Engineering (DCBI), Universidad Autónoma Metropolitana (UAM), Mexico City, Mexico
| | - Javier E Jiménez-Salazar
- Department of Biology of Reproduction, Division of Biological Sciences and Health (DCBS), Universidad Autónoma Metropolitana (UAM), Mexico City, Mexico
| | - Marcela Arteaga
- Department of Biology of Reproduction, Division of Biological Sciences and Health (DCBS), Universidad Autónoma Metropolitana (UAM), Mexico City, Mexico
| | - Michelle Arredondo
- Nanotechnology and Molecular Engineering Laboratory, Department of Chemistry, Division of Basic Science and Engineering (DCBI), Universidad Autónoma Metropolitana (UAM), Mexico City, Mexico
| | - Eva Ramón-Gallegos
- Department of Morphology, National School of Biological Sciences, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Nikola Batina
- Nanotechnology and Molecular Engineering Laboratory, Department of Chemistry, Division of Basic Science and Engineering (DCBI), Universidad Autónoma Metropolitana (UAM), Mexico City, Mexico
| | - Pablo Damián-Matsumura
- Department of Biology of Reproduction, Division of Biological Sciences and Health (DCBS), Universidad Autónoma Metropolitana (UAM), Mexico City, Mexico
| |
Collapse
|
14
|
Whitman NA, Lin ZW, DiProspero TJ, McIntosh JC, Lockett MR. Screening Estrogen Receptor Modulators in a Paper-Based Breast Cancer Model. Anal Chem 2018; 90:11981-11988. [PMID: 30226366 PMCID: PMC6200649 DOI: 10.1021/acs.analchem.8b02486] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The health risks associated with acute and prolonged exposure to estrogen receptor (ER) modulators has led to a concerted effort to identify and prioritize potential disruptors present in the environment. ER agonists and antagonists are identified with end-point assays, quantifying changes in cellular proliferation or gene transactivation in monolayers of estrogen receptor alpha expressing (ER+) cells upon exposure. While these monolayer cultures can be prepared, dosed, and analyzed in a highly parallelized manner, they are unable to predict the potencies of ER modulators in vivo accurately. Physiologically relevant model systems that better predict tissue- or organ-level responses are needed. To address this need, we describe here a screening platform capable of quantitatively assessing ER modulators in 96 chemically isolated 3D cultures. These cultures are supported in wax-patterned paper scaffolds whose design has improved performance and throughput over previously described paper-based setups. To highlight the potential of paper-based cultures for toxicity screens, we measured the potency of known ER modulators with a luciferase-based reporter assay. We also quantified the proliferation and invasion of two ER+ cell lines in the presence of estradiol. Despite the inability of the current setup to better predict in vivo potencies of ER modulators than monolayer cultures, the results demonstrate the potential of this platform to support increasingly complex and physiologically relevant tissue-like structures for environmental chemical risk assessment.
Collapse
Affiliation(s)
- Nathan A. Whitman
- Department of Chemistry, Kenan and Caudill Laboratories, University of North Carolina at Chapel Hill, 125 South Road, Chapel Hill, North Carolina 27599-3290, United States
| | - Zhi-Wei Lin
- Department of Chemistry, Kenan and Caudill Laboratories, University of North Carolina at Chapel Hill, 125 South Road, Chapel Hill, North Carolina 27599-3290, United States
| | - Thomas J. DiProspero
- Department of Chemistry, Kenan and Caudill Laboratories, University of North Carolina at Chapel Hill, 125 South Road, Chapel Hill, North Carolina 27599-3290, United States
| | - Julie C. McIntosh
- Department of Chemistry, Kenan and Caudill Laboratories, University of North Carolina at Chapel Hill, 125 South Road, Chapel Hill, North Carolina 27599-3290, United States
| | - Matthew R. Lockett
- Department of Chemistry, Kenan and Caudill Laboratories, University of North Carolina at Chapel Hill, 125 South Road, Chapel Hill, North Carolina 27599-3290, United States
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, 450 West Drive, Chapel Hill, North Carolina 27599-7295, United States
| |
Collapse
|
15
|
Zhang J, Wang Y, Jiang X, Chan HC. Cystic fibrosis transmembrane conductance regulator-emerging regulator of cancer. Cell Mol Life Sci 2018; 75:1737-1756. [PMID: 29411041 PMCID: PMC11105598 DOI: 10.1007/s00018-018-2755-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 12/27/2017] [Accepted: 01/17/2018] [Indexed: 12/11/2022]
Abstract
Mutations of cystic fibrosis transmembrane conductance regulator (CFTR) cause cystic fibrosis, the most common life-limiting recessive genetic disease among Caucasians. CFTR mutations have also been linked to increased risk of various cancers but remained controversial for a long time. Recent studies have begun to reveal that CFTR is not merely an ion channel but also an important regulator of cancer development and progression with multiple signaling pathways identified. In this review, we will first present clinical findings showing the correlation of genetic mutations or aberrant expression of CFTR with cancer incidence in multiple cancers. We will then focus on the roles of CFTR in fundamental cellular processes including transformation, survival, proliferation, migration, invasion and epithelial-mesenchymal transition in cancer cells, highlighting the signaling pathways involved. Finally, the association of CFTR expression levels with patient prognosis, and the potential of CFTR as a cancer prognosis indicator in human malignancies will be discussed.
Collapse
Affiliation(s)
- Jieting Zhang
- Faculty of Medicine, Epithelial Cell Biology Research Center, School of Biomedical Sciences, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, People's Republic of China
- Key Laboratory for Regenerative Medicine of the Ministry of Education of China, Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, People's Republic of China
- School of Biomedical Sciences Core Laboratory, Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, People's Republic of China
| | - Yan Wang
- Faculty of Medicine, Epithelial Cell Biology Research Center, School of Biomedical Sciences, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, People's Republic of China
- Key Laboratory for Regenerative Medicine of the Ministry of Education of China, Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, People's Republic of China
- School of Biomedical Sciences Core Laboratory, Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, People's Republic of China
| | - Xiaohua Jiang
- Faculty of Medicine, Epithelial Cell Biology Research Center, School of Biomedical Sciences, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, People's Republic of China.
- Key Laboratory for Regenerative Medicine of the Ministry of Education of China, Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, People's Republic of China.
- School of Biomedical Sciences Core Laboratory, Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, People's Republic of China.
| | - Hsiao Chang Chan
- Faculty of Medicine, Epithelial Cell Biology Research Center, School of Biomedical Sciences, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, People's Republic of China.
- Key Laboratory for Regenerative Medicine of the Ministry of Education of China, Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, People's Republic of China.
- School of Biomedical Sciences Core Laboratory, Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, People's Republic of China.
- Sichuan University-The Chinese University of Hong Kong Joint Laboratory for Reproductive Medicine, West China Second University Hospital, Chengdu, People's Republic of China.
| |
Collapse
|
16
|
Silva RDS, Lombardi APG, de Souza DS, Vicente CM, Porto CS. Activation of estrogen receptor beta (ERβ) regulates the expression of N-cadherin, E-cadherin and β-catenin in androgen-independent prostate cancer cells. Int J Biochem Cell Biol 2018; 96:40-50. [PMID: 29341930 DOI: 10.1016/j.biocel.2018.01.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 01/10/2018] [Accepted: 01/12/2018] [Indexed: 12/11/2022]
Abstract
The aim of the present study was to investigate the impact of the activation of estrogen receptors on expression and localization of N-cadherin, E-cadherin and non-phosphorylated β-catenin in androgen-independent prostate cancer cells (PC-3 and DU-145) and in human post pubertal prostate epithelial cells (PNT1A). Expression of N-cadherin was detected in PNT1A and PC-3 cells, but not in DU-145 cells. E-cadherin was detected only in DU-145 cells and β-catenin was detected in all cells studied. N-cadherin and β-catenin were located preferentially in the cellular membrane of PNT1A cells and in the cytoplasm of PC-3 cells. E-cadherin and β-catenin were located preferentially in the cellular membrane of DU-145 cells. 17β-estradiol (E2) or the ERα-selective agonist PPT did not affect the content and localization of N-cadherin in PC-3 and PNT1A cells or E-cadherin in DU-145 cells. In PC-3 cells, ERβ-selective agonist DPN decreased the expression of N-cadherin. DPN-induced downregulation of N-cadherin was blocked by pretreatment with the ERβ-selective antagonist (PHTPP), indicating that ERβ1 is the upstream receptor regulating the expression of N-cadherin. In DU-145 cells, the activation of ERβ1 by DPN increased the expression of E-cadherin. Taken together, these results suggest that activation of ERβ1 is required to maintain an epithelial phenotype in PC-3 and DU-145 cells. The activation of ERβ1 also increased the expression of β-catenin in cytoplasm of PC-3 and in the cellular membrane of DU-145 cells. In conclusion, our results indicate differential expression and localization of N-cadherin, E-cadherin and β-catenin in androgen-independent prostate cancer cells. The reduction of N-cadherin content by activation of ERβ, exclusively observed in androgen-independent prostate cancer cells (PC-3), may be related to the activation of signaling pathways, such as the release of β-catenin into the cytoplasm, translocation of β-catenin to the nucleus and activation of gene transcription.
Collapse
Affiliation(s)
- Rafael de Souza Silva
- Section of Experimental Endocrinology, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Pedro de Toledo 669, Vila Clementino, São Paulo, SP, 04039-032, Brazil
| | - Ana Paola G Lombardi
- Section of Experimental Endocrinology, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Pedro de Toledo 669, Vila Clementino, São Paulo, SP, 04039-032, Brazil
| | - Deborah Simão de Souza
- Section of Experimental Endocrinology, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Pedro de Toledo 669, Vila Clementino, São Paulo, SP, 04039-032, Brazil
| | - Carolina M Vicente
- Section of Experimental Endocrinology, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Pedro de Toledo 669, Vila Clementino, São Paulo, SP, 04039-032, Brazil
| | - Catarina S Porto
- Section of Experimental Endocrinology, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Pedro de Toledo 669, Vila Clementino, São Paulo, SP, 04039-032, Brazil.
| |
Collapse
|
17
|
Laedrach C, Salhia B, Cihoric N, Zlobec I, Tapia C. Immunophenotypic profile of tumor buds in breast cancer. Pathol Res Pract 2017; 214:25-29. [PMID: 29254793 PMCID: PMC7474545 DOI: 10.1016/j.prp.2017.11.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2017] [Revised: 11/20/2017] [Accepted: 11/28/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND Tumor buds are associated with lympho-vascular invasion and lymph node metastases leading to the assumption that they are involved in the early metastatic process. Hence, it would be important to know if tumor buds can be targeted with the most widely used targeted therapies in breast cancer (BC) and if changes in hormone and Her2 status occur. The aim of this study was to answer these questions by determining whether hormone receptor (HR) and Her2 status are expressed in the tumor buds of a large cohort of BCs. DESIGN We constructed a tumor bud next-generation tissue microarray (ngTMA) consisting of n = 199 BCs of non-special type. Generally, two 1 mm punches were taken from the tumor bud areas in the periphery (PTB) and within the tumor center (ITB). HR and Her2 status was assessed using immunohistochemistry and fluorescence in situ hybridization, respectively. HR status was positive if ≥1% of tumor bud cells were positive. Her2 status was considered positive if bud cells showed strong complete membranous Her2 over-expression or Her2 amplification. RESULTS Most tumor buds were positive for estrogen (ER) (PTB: 86%; ITB: 88.3) and progesterone receptor (PgR) (PTB: 72%; ITB: 72.8%) and Her2 was positive in: PTB 11.5% and ITB 11%. A difference between the main tumor mass and tumor buds (PTB and ITB) was seen for PgR in 3.5% of cases (n = 7). No differences were seen for ER and Her2 between tumor buds and main tumor mass. CONCLUSION Most tumor buds (96.5%) share the same HR and Her2 expression profile of the main tumor mass, implying that tumor buds relay on the same pathways as the main tumor mass and might be equally responsive to targeted therapies.
Collapse
Affiliation(s)
- C Laedrach
- Intstitute of Pathology, University of Bern, Bern, Switzerland
| | - B Salhia
- Department of Translational Genomics, Keck School of Medicine of USC, Los Angeles, CA, USA
| | - N Cihoric
- Department of Radio-Oncology, Inselspital, Bern, Switzerland; Breast Center, Inselspital, Bern, Switzerland
| | - I Zlobec
- Intstitute of Pathology, University of Bern, Bern, Switzerland
| | - C Tapia
- Intstitute of Pathology, University of Bern, Bern, Switzerland; Breast Center, Inselspital, Bern, Switzerland; Department of Molecular Pathology, UT MD Anderson Cancer Center, Houston, TX, USA; Department of Investigational Cancer Therapeutics, UT MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
18
|
Paramita P, Louisa M, Nafrialdi N. Increased vimentin mRNA expression in MCF-7 breast cancer cell line after repeated endoxifen-treatment. MEDICAL JOURNAL OF INDONESIA 2017. [DOI: 10.13181/mji.v25i4.1397] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Background: Epithelial mesenchymal transition (EMT) plays a significant role in the development of cancer cell resistance to drugs. Vimentin, a type III intermediate filament protein, is a marker of EMT. Vimentin's over-expression in cancer correlates well with increased tumor growth, change in cell shape and poor prognosis. Endoxifen is an active metabolite of tamoxifen and has become a new potent agent in the treatment of breast cancer. This is a study that aimed to investigate the effect of endoxifen exposure with or without estradiol on cell viability, cell morphology and EMT progression through the analysis of vimentin mRNA expression after 4-week treatment.
Methods: Endoxifen, 100 nM or 1,000 nM, with or without beta-estradiol were given repeatedly to MCF-7 cells. Cells treated with dimethyl sulfoxide (DMSO) 0.001% were used as control. After 2- and 4-week exposure, the cells were counted, analyzed for mRNA vimentin expression, and observed for morphological changes.
Results: Compared to control, there were significant decreases in vimentin mRNA expressions in endoxifen and endoxifen+β-estradiol treated cells after 2-weeks, which then significantly increased after 4-week compared with the 2-week exposure. We found no change in morphology of MCF-7 cells.
Conclusion: Repeated exposure of endoxifen might induce EMT progression through increased expression of vimentin in MCF-7 breast cancer cell line.
Collapse
|
19
|
Gonzalez-Mariscal L, Miranda J, Ortega-Olvera JM, Gallego-Gutierrez H, Raya-Sandino A, Vargas-Sierra O. Zonula Occludens Proteins in Cancer. CURRENT PATHOBIOLOGY REPORTS 2016. [DOI: 10.1007/s40139-016-0109-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
20
|
The complex nature of oestrogen signalling in breast cancer: enemy or ally? Biosci Rep 2016; 36:BSR20160017. [PMID: 27160081 PMCID: PMC5293589 DOI: 10.1042/bsr20160017] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 05/09/2016] [Indexed: 02/07/2023] Open
Abstract
The pleiotropic nature of oestradiol, the main oestrogen found in women, has been well described in the literature. Oestradiol is positioned to play a unique role since it can respond to environmental, genetic and non-genetic cues to affect genetic expression and cellular signalling. In breast cancer, oestradiol signalling has a dual effect, promoting or inhibiting cancer growth. The potential impact of oestradiol on tumorigenesis depends on the molecular and cellular characteristics of the breast cancer cell. In this review, we provide a broad survey discussing the cellular and molecular consequences of oestrogen signalling in breast cancer. First, we review the structure of the classical oestrogen receptors and resultant transcriptional (genomic) and non-transcriptional (non-genomic) signalling. We then discuss the nature of oestradiol signalling in breast cancer including the specific receptors that initiate these signalling cascades as well as potential outcomes, such as cancer growth, proliferation and angiogenesis. Finally, we examine cellular and molecular mechanisms underlying the dimorphic effect of oestrogen signalling in breast cancer.
Collapse
|
21
|
Vantangoli MM, Wilson S, Madnick SJ, Huse SM, Boekelheide K. Morphologic effects of estrogen stimulation on 3D MCF-7 microtissues. Toxicol Lett 2016; 248:1-8. [PMID: 26921789 DOI: 10.1016/j.toxlet.2016.02.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 01/29/2016] [Accepted: 02/22/2016] [Indexed: 12/16/2022]
Abstract
In the development of human cell-based assays, 3-dimensional (3D) cell culture models are intriguing as they are able to bridge the gap between animal models and traditional two-dimensional (2D) cell culture. Previous work has demonstrated that MCF-7 human breast carcinoma cells cultured in a 3D scaffold-free culture system self-assemble and develop into differentiated microtissues that possess a luminal space. Exposure to estradiol for 7 days decreased lumen formation in MCF-7 microtissues, altered microtissue morphology and altered expression of genes involved in estrogen signaling, cell adhesion and cell cycle regulation. Exposure to receptor-specific agonists for estrogen receptor alpha, estrogen receptor beta and g-protein coupled estrogen receptor resulted in unique, receptor-specific phenotypes and gene expression signatures. The use of a differentiated scaffold-free 3D culture system offers a unique opportunity to study the phenotypic and molecular changes associated with exposure to estrogenic compounds.
Collapse
Affiliation(s)
- Marguerite M Vantangoli
- Department of Pathology and Laboratory Medicine, 70 Ship Street, Brown University, Providence, RI 02903, USA
| | - Shelby Wilson
- Department of Pathology and Laboratory Medicine, 70 Ship Street, Brown University, Providence, RI 02903, USA
| | - Samantha J Madnick
- Department of Pathology and Laboratory Medicine, 70 Ship Street, Brown University, Providence, RI 02903, USA
| | - Susan M Huse
- Department of Pathology and Laboratory Medicine, 70 Ship Street, Brown University, Providence, RI 02903, USA
| | - Kim Boekelheide
- Department of Pathology and Laboratory Medicine, 70 Ship Street, Brown University, Providence, RI 02903, USA.
| |
Collapse
|
22
|
Role of Epithelial-Mesenchyme Transition in Chlamydia Pathogenesis. PLoS One 2015; 10:e0145198. [PMID: 26681200 PMCID: PMC4683008 DOI: 10.1371/journal.pone.0145198] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 12/01/2015] [Indexed: 12/15/2022] Open
Abstract
Chlamydia trachomatis genital infection in women causes serious adverse reproductive complications, and is a strong co-factor for human papilloma virus (HPV)-associated cervical epithelial carcinoma. We tested the hypothesis that Chlamydia induces epithelial-mesenchyme transition (EMT) involving T cell-derived TNF-alpha signaling, caspase activation, cleavage inactivation of dicer and dysregulation of micro-RNA (miRNA) in the reproductive epithelium; the pathologic process of EMT causes fibrosis and fertility-related epithelial dysfunction, and also provides the co-factor function for HPV-related cervical epithelial carcinoma. Using a combination of microarrays, immunohistochemistry and proteomics, we showed that chlamydia altered the expression of crucial miRNAs that control EMT, fibrosis and tumorigenesis; specifically, miR-15a, miR-29b, miR-382 and MiR-429 that maintain epithelial integrity were down-regulated, while miR-9, mi-R-19a, miR-22 and miR-205 that promote EMT, fibrosis and tumorigenesis were up-regulated. Chlamydia induced EMT in vitro and in vivo, marked by the suppression of normal epithelial cell markers especially E-cadherin but up-regulation of mesenchymal markers of pathological EMT, including T-cadherin, MMP9, and fibronectin. Also, Chlamydia upregulated pro-EMT regulators, including the zinc finger E-box binding homeobox protein, ZEB1, Snail1/2, and thrombospondin1 (Thbs1), but down-regulated anti-EMT and fertility promoting proteins (i.e., the major gap junction protein connexin 43 (Cx43), Mets1, Add1Scarb1 and MARCKSL1). T cell-derived TNF-alpha signaling was required for chlamydial-induced infertility and caspase inhibitors prevented both infertility and EMT. Thus, chlamydial-induced T cell-derived TNF-alpha activated caspases that inactivated dicer, causing alteration in the expression of reproductive epithelial miRNAs and induction of EMT. EMT causes epithelial malfunction, fibrosis, infertility, and the enhancement of tumorigenesis of HPV oncogene-transformed epithelial cells. These findings provide a novel understanding of the molecular pathogenesis of chlamydia-associated diseases, which may guide a rational prevention strategy.
Collapse
|
23
|
Yokdang N, Hatakeyama J, Wald JH, Simion C, Tellez JD, Chang DZ, Swamynathan MM, Chen M, Murphy WJ, Carraway Iii KL, Sweeney C. LRIG1 opposes epithelial-to-mesenchymal transition and inhibits invasion of basal-like breast cancer cells. Oncogene 2015; 35:2932-47. [PMID: 26387542 PMCID: PMC4805527 DOI: 10.1038/onc.2015.345] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Revised: 06/24/2015] [Accepted: 08/04/2015] [Indexed: 01/04/2023]
Abstract
LRIG1, a member of the LRIG family of transmembrane leucine rich repeat-containing proteins, is a negative regulator of receptor tyrosine kinase signaling and a tumor suppressor. LRIG1 expression is broadly decreased in human cancer and in breast cancer, low expression of LRIG1 has been linked to decreased relapse-free survival. Recently, low expression of LRIG1 was revealed to be an independent risk factor for breast cancer metastasis and death. These findings suggest that LRIG1 may oppose breast cancer cell motility and invasion, cellular processes which are fundamental to metastasis. However, very little is known of LRIG1 function in this regard. In this study, we demonstrate that LRIG1 is down-regulated during epithelial to mesenchymal transition (EMT) of human mammary epithelial cells, suggesting that LRIG1 expression may represent a barrier to EMT. Indeed, depletion of endogenous LRIG1 in human mammary epithelial cells expands the stem cell population, augments mammosphere formation and accelerates EMT. Conversely, expression of LRIG1 in highly invasive Basal B breast cancer cells provokes a mesenchymal to epithelial transition accompanied by a dramatic suppression of tumorsphere formation and a striking loss of invasive growth in three-dimensional culture. LRIG1 expression perturbs multiple signaling pathways and represses markers and effectors of the mesenchymal state. Furthermore, LRIG1 expression in MDA-MB-231 breast cancer cells significantly slows their growth as tumors, providing the first in vivo evidence that LRIG1 functions as a growth suppressor in breast cancer.
Collapse
Affiliation(s)
- N Yokdang
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Sacramento, CA, USA
| | - J Hatakeyama
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Sacramento, CA, USA
| | - J H Wald
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Sacramento, CA, USA
| | - C Simion
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Sacramento, CA, USA
| | - J D Tellez
- Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, CA, USA
| | - D Z Chang
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Sacramento, CA, USA
| | - M M Swamynathan
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Sacramento, CA, USA
| | - M Chen
- Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, CA, USA
| | - W J Murphy
- Department of Dermatology, University of California, Davis, Sacramento, CA, USA
| | - K L Carraway Iii
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Sacramento, CA, USA
| | - C Sweeney
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Sacramento, CA, USA
| |
Collapse
|
24
|
The relationship between tumour budding, the tumour microenvironment and survival in patients with invasive ductal breast cancer. Br J Cancer 2015; 113:1066-74. [PMID: 26263482 PMCID: PMC4651125 DOI: 10.1038/bjc.2015.287] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 07/08/2015] [Accepted: 07/14/2015] [Indexed: 12/17/2022] Open
Abstract
Background: Tumour budding has previously been reported to predict survival in several solid organ tumours, including breast; however, whether this is independent of other aspects of the tumour microenvironment is unknown. In the present study, the relationship between tumour budding, the tumour microenvironment and survival was examined in patients with invasive ductal breast cancer. Methods: Patients presenting between 1995 and 1998 were studied (n=474). Using routine pathological sections, tumour budding was measured at the invasive margin and its association with clinicopathological characteristics and cancer-specific survival (CSS) was examined. Results: Tumour budding was associated with several adverse pathological characteristics, including lymph node involvement, lymph vessel invasion (LVI), increased tumour stroma percentage (TSP) and weaker local inflammatory infiltrative. Tumour budding was associated with reduced CSS (hazard ratio (HR) 2.08, 95% confidence interval (CI) 1.14–3.09, P=0.004), independent of nodal status, molecular subtypes, tumour necrosis, CD8+, CD138+, LVI, blood vessel invasion and TSP. Further, tumour budding was independently associated with reduced CSS in node-negative patients (HR 2.63, 95% CI 1.16–5.92, P=0.020) and those who have low TSP (HR 1.98, 95% CI 1.09–3.57, P=0.024) and high-grade local inflammatory infiltrative (HR 2.27, 95% CI 1.35–5.36, P=0.014). Conclusions: Tumour budding was a significant predictor of survival in patients with invasive ductal breast cancer, independent of adverse pathological characteristics and components of tumour microenvironment. The present study further confirms the clinical utility of both tumour and host-based factors of tumour microenvironment.
Collapse
|
25
|
Liu LB, Liu XB, Ma J, Liu YH, Li ZQ, Ma T, Zhao XH, Xi Z, Xue YX. Bradykinin increased the permeability of BTB via NOS/NO/ZONAB-mediating down-regulation of claudin-5 and occludin. Biochem Biophys Res Commun 2015; 464:118-25. [PMID: 26106824 DOI: 10.1016/j.bbrc.2015.06.082] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 06/11/2015] [Indexed: 12/12/2022]
Abstract
After demonstrating bradykinin (BK) could increase the permeability of blood-tumor barrier (BTB) via opening the tight junction (TJ), and that the possible mechanism is unclear, we demonstrated that BK could increase the expressions of eNOS and nNOS and promote ZONAB translocation into nucleus. NOS inhibitors l-NAME and 7-NI could effectively block the effect of BK on increasing BTB permeability, decreasing the expressions of claudin-5 and occludin and promoting the translocation of ZONAB. Overexpression of ZONAB could significantly enhance BK-mediating BTB permeability. Meanwhile, chromatin immunoprecipitation verified ZONAB interacted with the promoter of claudin-5 and occludin respectively. This study indicated NOS/NO/ZONAB pathway might be involved in BK's increasing the permeability of BTB.
Collapse
Affiliation(s)
- Li-bo Liu
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang 110122, PR China
| | - Xiao-bai Liu
- The 96th Class, 7-Year Program, China Medical University, Shenyang 110122, PR China
| | - Jun Ma
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang 110122, PR China
| | - Yun-hui Liu
- Department of Neurosurgery, Shengjing Hospital, China Medical University, Shenyang 110004, PR China
| | - Zhi-qing Li
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang 110122, PR China
| | - Teng Ma
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang 110122, PR China
| | - Xi-he Zhao
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang 110122, PR China
| | - Zhuo Xi
- Department of Neurosurgery, Shengjing Hospital, China Medical University, Shenyang 110004, PR China
| | - Yi-xue Xue
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang 110122, PR China.
| |
Collapse
|
26
|
Salhia B, Trippel M, Pfaltz K, Cihoric N, Grogg A, Lädrach C, Zlobec I, Tapia C. High tumor budding stratifies breast cancer with metastatic properties. Breast Cancer Res Treat 2015; 150:363-71. [PMID: 25779101 PMCID: PMC4368849 DOI: 10.1007/s10549-015-3333-3] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Accepted: 03/04/2015] [Indexed: 12/20/2022]
Abstract
Tumor budding refers to single or small cluster of tumor cells detached from the main tumor mass. In colon cancer high tumor budding is associated with positive lymph nodes and worse prognosis. Therefore, we investigated the value of tumor budding as a predictive feature of lymph node status in breast cancer (BC). Whole tissue sections from 148 surgical resection specimens (SRS) and 99 matched preoperative core biopsies (CB) with invasive BC of no special type were analyzed on one slide stained with pan-cytokeratin. In SRS, the total number of intratumoral (ITB) and peripheral tumor buds (PTB) in ten high-power fields (HPF) were counted. A bud was defined as a single tumor cell or a cluster of up to five tumor cells. High tumor budding equated to scores averaging >4 tumor buds across 10HPFs. In CB high tumor budding was defined as ≥10 buds/HPF. The results were correlated with pathological parameters. In SRS high PTB stratified BC with lymph node metastases (p ≤ 0.03) and lymphatic invasion (p ≤ 0.015). In CB high tumor budding was significantly (p = 0.0063) associated with venous invasion. Pathologists are able, based on morphology, to categorize BC into a high and low risk groups based in part on lymph node status. This risk assessment can be easily performed during routine diagnostics and it is time and cost effective. These results suggest that high PTB is associated with loco-regional metastasis, highlighting the possibility that this tumor feature may help in therapeutic decision-making.
Collapse
Affiliation(s)
- Bodour Salhia
- Integrated Cancer Genomics Division, Translational Genomics Research Institute, 445 N. Fifth Street, Phoenix, AZ 85004 USA
| | - Mafalda Trippel
- Division of Clinical Pathology, Institute of Pathology, University of Bern, Murtenstrasse 31, 3010 Bern, Switzerland
| | - Katrin Pfaltz
- Division of Clinical Pathology, Institute of Pathology, University of Bern, Murtenstrasse 31, 3010 Bern, Switzerland
| | - Nikola Cihoric
- Department of Radiation Oncology, Bern University Hospital, and University of Bern, Bern, Switzerland
- Breast Center, University Hospital Bern, Bern, Switzerland
| | - André Grogg
- Division of Clinical Pathology, Institute of Pathology, University of Bern, Murtenstrasse 31, 3010 Bern, Switzerland
| | - Claudia Lädrach
- Division of Clinical Pathology, Institute of Pathology, University of Bern, Murtenstrasse 31, 3010 Bern, Switzerland
| | - Inti Zlobec
- Translational Research Unit (TRU), Institute of Pathology, University of Bern, Bern, Switzerland
| | - Coya Tapia
- Division of Clinical Pathology, Institute of Pathology, University of Bern, Murtenstrasse 31, 3010 Bern, Switzerland
- Breast Center, University Hospital Bern, Bern, Switzerland
| |
Collapse
|