1
|
Caneparo C, Carignan L, Lonina E, Goulet SM, Pellerin FA, Chabaud S, Bordeleau F, Bolduc S, Pelletier M. Impact of Endocrine Disruptors on the Genitourinary Tract. J Xenobiot 2024; 14:1849-1888. [PMID: 39728407 DOI: 10.3390/jox14040099] [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: 08/30/2024] [Revised: 11/04/2024] [Accepted: 11/26/2024] [Indexed: 12/28/2024] Open
Abstract
Over the last decades, the human species has seen an increase in the incidence of pathologies linked to the genitourinary tract. Observations in animals have allowed us to link these increases, at least in part, to changes in the environment and, in particular, to an increasing presence of endocrine disruptors. These can be physical agents, such as light or heat; natural products, such as phytoestrogens; or chemicals produced by humans. Endocrine disruptors may interfere with the signaling pathways mediated by the endocrine system, particularly those linked to sex hormones. These factors and their general effects are presented before focusing on the male and female genitourinary tracts by describing their anatomy, development, and pathologies, including bladder and prostate cancer.
Collapse
Affiliation(s)
- Christophe Caneparo
- Department of Pediatrics, Gynecology and Obstetrics, Faculty of Medicine, Geneva University Hospitals, University of Geneva, CH-1205 Geneva, Switzerland
| | - Laurence Carignan
- Oncology Division, CHU de Québec-Université Laval Research Center and Université Laval Cancer Research Center, Quebec, QC G1R 3S3, Canada
- Regenerative Medicine Division, Centre de Recherche en Organogénèse Expérimentale/LOEX, CHU de Québec-Université Laval Research Center, Université Laval, Quebec, QC G1J 5B3, Canada
| | - Elena Lonina
- Infectious and Immune Diseases Division, CHU de Québec-Université Laval Research Center, Quebec, QC G1V 4G2, Canada
- Intersectorial Centre for Endocrine Disruptors Analysis, Institut National de La Recherche Scientifique (INRS), Montreal, QC H4V 1B7, Canada
- Department of Microbiology-Infectious Diseases and Immunology, Faculty of Medicine, Laval University; ARThrite Research Center, Université Laval, Quebec, QC G1V 0A6, Canada
| | - Sarah-Maude Goulet
- Infectious and Immune Diseases Division, CHU de Québec-Université Laval Research Center, Quebec, QC G1V 4G2, Canada
- Intersectorial Centre for Endocrine Disruptors Analysis, Institut National de La Recherche Scientifique (INRS), Montreal, QC H4V 1B7, Canada
- Department of Microbiology-Infectious Diseases and Immunology, Faculty of Medicine, Laval University; ARThrite Research Center, Université Laval, Quebec, QC G1V 0A6, Canada
| | - Felix-Antoine Pellerin
- Oncology Division, CHU de Québec-Université Laval Research Center and Université Laval Cancer Research Center, Quebec, QC G1R 3S3, Canada
- Regenerative Medicine Division, Centre de Recherche en Organogénèse Expérimentale/LOEX, CHU de Québec-Université Laval Research Center, Université Laval, Quebec, QC G1J 5B3, Canada
| | - Stéphane Chabaud
- Regenerative Medicine Division, Centre de Recherche en Organogénèse Expérimentale/LOEX, CHU de Québec-Université Laval Research Center, Université Laval, Quebec, QC G1J 5B3, Canada
| | - François Bordeleau
- Oncology Division, CHU de Québec-Université Laval Research Center and Université Laval Cancer Research Center, Quebec, QC G1R 3S3, Canada
- Regenerative Medicine Division, Centre de Recherche en Organogénèse Expérimentale/LOEX, CHU de Québec-Université Laval Research Center, Université Laval, Quebec, QC G1J 5B3, Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology, Université Laval, Quebec, QC G1V 0A6, Canada
| | - Stéphane Bolduc
- Regenerative Medicine Division, Centre de Recherche en Organogénèse Expérimentale/LOEX, CHU de Québec-Université Laval Research Center, Université Laval, Quebec, QC G1J 5B3, Canada
- Department of Surgery, Université Laval, Quebec, QC G1V 0A6, Canada
| | - Martin Pelletier
- Infectious and Immune Diseases Division, CHU de Québec-Université Laval Research Center, Quebec, QC G1V 4G2, Canada
- Intersectorial Centre for Endocrine Disruptors Analysis, Institut National de La Recherche Scientifique (INRS), Montreal, QC H4V 1B7, Canada
- Department of Microbiology-Infectious Diseases and Immunology, Faculty of Medicine, Laval University; ARThrite Research Center, Université Laval, Quebec, QC G1V 0A6, Canada
| |
Collapse
|
2
|
Lafront C, Germain L, Campolina-Silva GH, Weidmann C, Berthiaume L, Hovington H, Brisson H, Jobin C, Frégeau-Proulx L, Cotau R, Gonthier K, Lacouture A, Caron P, Ménard C, Atallah C, Riopel J, Latulippe É, Bergeron A, Toren P, Guillemette C, Pelletier M, Fradet Y, Belleannée C, Pouliot F, Lacombe L, Lévesque É, Audet-Walsh É. The estrogen signaling pathway reprograms prostate cancer cell metabolism and supports proliferation and disease progression. J Clin Invest 2024; 134:e170809. [PMID: 38625747 PMCID: PMC11142735 DOI: 10.1172/jci170809] [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: 03/24/2023] [Accepted: 04/09/2024] [Indexed: 04/17/2024] Open
Abstract
Just like the androgen receptor (AR), the estrogen receptor α (ERα) is expressed in the prostate and is thought to influence prostate cancer (PCa) biology. Yet the incomplete understanding of ERα functions in PCa hinders our ability to fully comprehend its clinical relevance and restricts the repurposing of estrogen-targeted therapies for the treatment of this disease. Using 2 human PCa tissue microarray cohorts, we first demonstrate that nuclear ERα expression was heterogeneous among patients, being detected in only half of the tumors. Positive nuclear ERα levels were correlated with disease recurrence, progression to metastatic PCa, and patient survival. Using in vitro and in vivo models of the normal prostate and PCa, bulk and single-cell RNA-Seq analyses revealed that estrogens partially mimicked the androgen transcriptional response and activated specific biological pathways linked to proliferation and metabolism. Bioenergetic flux assays and metabolomics confirmed the regulation of cancer metabolism by estrogens, supporting proliferation. Using cancer cell lines and patient-derived organoids, selective estrogen receptor modulators, a pure anti-estrogen, and genetic approaches impaired cancer cell proliferation and growth in an ERα-dependent manner. Overall, our study revealed that, when expressed, ERα functionally reprogrammed PCa metabolism, was associated with disease progression, and could be targeted for therapeutic purposes.
Collapse
Affiliation(s)
- Camille Lafront
- Department of Molecular Medicine, Université Laval, Quebec City, Québec, Canada
- Endocrinology and Nephrology Division, CHU de Québec – Université Laval Research Center (CRCHUQ-UL), Quebec City, Québec, Canada
- Cancer Research Center (CRC) of Université Laval, Quebec City, Québec, Canada
| | - Lucas Germain
- Department of Molecular Medicine, Université Laval, Quebec City, Québec, Canada
- Endocrinology and Nephrology Division, CHU de Québec – Université Laval Research Center (CRCHUQ-UL), Quebec City, Québec, Canada
- Cancer Research Center (CRC) of Université Laval, Quebec City, Québec, Canada
| | - Gabriel H. Campolina-Silva
- Department of Obstetrics, Gynecology and Reproduction, Université Laval, Quebec City, Québec, Canada
- Reproduction, Mother and Youth Health Division, CRCHUQ-UL, Quebec City, Québec, Canada
| | - Cindy Weidmann
- Endocrinology and Nephrology Division, CHU de Québec – Université Laval Research Center (CRCHUQ-UL), Quebec City, Québec, Canada
- Cancer Research Center (CRC) of Université Laval, Quebec City, Québec, Canada
| | - Line Berthiaume
- Endocrinology and Nephrology Division, CHU de Québec – Université Laval Research Center (CRCHUQ-UL), Quebec City, Québec, Canada
- Cancer Research Center (CRC) of Université Laval, Quebec City, Québec, Canada
| | - Hélène Hovington
- Cancer Research Center (CRC) of Université Laval, Quebec City, Québec, Canada
- Department of Medicine, Université Laval, Quebec City, Québec, Canada
| | - Hervé Brisson
- Cancer Research Center (CRC) of Université Laval, Quebec City, Québec, Canada
- Department of Medicine, Université Laval, Quebec City, Québec, Canada
| | - Cynthia Jobin
- Department of Molecular Medicine, Université Laval, Quebec City, Québec, Canada
- Endocrinology and Nephrology Division, CHU de Québec – Université Laval Research Center (CRCHUQ-UL), Quebec City, Québec, Canada
- Cancer Research Center (CRC) of Université Laval, Quebec City, Québec, Canada
| | - Lilianne Frégeau-Proulx
- Department of Molecular Medicine, Université Laval, Quebec City, Québec, Canada
- Endocrinology and Nephrology Division, CHU de Québec – Université Laval Research Center (CRCHUQ-UL), Quebec City, Québec, Canada
- Cancer Research Center (CRC) of Université Laval, Quebec City, Québec, Canada
| | - Raul Cotau
- Endocrinology and Nephrology Division, CHU de Québec – Université Laval Research Center (CRCHUQ-UL), Quebec City, Québec, Canada
- Cancer Research Center (CRC) of Université Laval, Quebec City, Québec, Canada
- Oncology Research Division, CRCHUQ-UL, Quebec City, Québec, Canada
| | - Kevin Gonthier
- Department of Molecular Medicine, Université Laval, Quebec City, Québec, Canada
- Endocrinology and Nephrology Division, CHU de Québec – Université Laval Research Center (CRCHUQ-UL), Quebec City, Québec, Canada
- Cancer Research Center (CRC) of Université Laval, Quebec City, Québec, Canada
| | - Aurélie Lacouture
- Department of Molecular Medicine, Université Laval, Quebec City, Québec, Canada
- Endocrinology and Nephrology Division, CHU de Québec – Université Laval Research Center (CRCHUQ-UL), Quebec City, Québec, Canada
- Cancer Research Center (CRC) of Université Laval, Quebec City, Québec, Canada
| | - Patrick Caron
- Endocrinology and Nephrology Division, CHU de Québec – Université Laval Research Center (CRCHUQ-UL), Quebec City, Québec, Canada
- Cancer Research Center (CRC) of Université Laval, Quebec City, Québec, Canada
| | - Claire Ménard
- Department of Medicine, Université Laval, Quebec City, Québec, Canada
| | - Chantal Atallah
- Department of Medicine, Université Laval, Quebec City, Québec, Canada
- Department of Pathology, CHU de Québec-Université Laval, Quebec City, Québec, Canada
| | - Julie Riopel
- Endocrinology and Nephrology Division, CHU de Québec – Université Laval Research Center (CRCHUQ-UL), Quebec City, Québec, Canada
- Department of Pathology, CHU de Québec-Université Laval, Quebec City, Québec, Canada
| | - Éva Latulippe
- Department of Pathology, CHU de Québec-Université Laval, Quebec City, Québec, Canada
| | - Alain Bergeron
- Cancer Research Center (CRC) of Université Laval, Quebec City, Québec, Canada
- Oncology Research Division, CRCHUQ-UL, Quebec City, Québec, Canada
- Department of Surgery
| | - Paul Toren
- Cancer Research Center (CRC) of Université Laval, Quebec City, Québec, Canada
- Oncology Research Division, CRCHUQ-UL, Quebec City, Québec, Canada
- Department of Surgery
| | - Chantal Guillemette
- Endocrinology and Nephrology Division, CHU de Québec – Université Laval Research Center (CRCHUQ-UL), Quebec City, Québec, Canada
- Cancer Research Center (CRC) of Université Laval, Quebec City, Québec, Canada
- Faculty of Pharmacy, and
| | - Martin Pelletier
- Department of Microbiology-Infectious Diseases and Immunology, Université Laval, Quebec City, Québec, Canada
- Infectious and Immune Diseases Research Division, CRCHUQ-UL, Quebec City, Québec, Canada
- ARThrite Research Center, Université Laval, Quebec City, Québec, Canada
| | - Yves Fradet
- Cancer Research Center (CRC) of Université Laval, Quebec City, Québec, Canada
- Oncology Research Division, CRCHUQ-UL, Quebec City, Québec, Canada
- Department of Surgery
| | - Clémence Belleannée
- Department of Obstetrics, Gynecology and Reproduction, Université Laval, Quebec City, Québec, Canada
- Reproduction, Mother and Youth Health Division, CRCHUQ-UL, Quebec City, Québec, Canada
| | - Frédéric Pouliot
- Cancer Research Center (CRC) of Université Laval, Quebec City, Québec, Canada
- Oncology Research Division, CRCHUQ-UL, Quebec City, Québec, Canada
- Department of Surgery
| | - Louis Lacombe
- Cancer Research Center (CRC) of Université Laval, Quebec City, Québec, Canada
- Oncology Research Division, CRCHUQ-UL, Quebec City, Québec, Canada
- Department of Surgery
| | - Éric Lévesque
- Endocrinology and Nephrology Division, CHU de Québec – Université Laval Research Center (CRCHUQ-UL), Quebec City, Québec, Canada
- Cancer Research Center (CRC) of Université Laval, Quebec City, Québec, Canada
- Department of Medicine, Université Laval, Quebec City, Québec, Canada
| | - Étienne Audet-Walsh
- Department of Molecular Medicine, Université Laval, Quebec City, Québec, Canada
- Endocrinology and Nephrology Division, CHU de Québec – Université Laval Research Center (CRCHUQ-UL), Quebec City, Québec, Canada
- Cancer Research Center (CRC) of Université Laval, Quebec City, Québec, Canada
| |
Collapse
|
3
|
Yang Y, Sun X, Luo L, Peng R, Yang R, Cheng Z, Lv Y, Li H, Tang Q, Zhu W, Qiao D, Xu S. Discovery of novel potent PI3K/mTOR dual-target inhibitors based on scaffold hopping: Design, synthesis, and antiproliferative activity. Arch Pharm (Weinheim) 2023; 356:e2300403. [PMID: 37840368 DOI: 10.1002/ardp.202300403] [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: 07/24/2023] [Revised: 09/21/2023] [Accepted: 09/22/2023] [Indexed: 10/17/2023]
Abstract
The PI3K/AKT/mTOR pathway is one of the most common dysregulated signaling cascade responses in human cancers, playing a crucial role in cell proliferation and angiogenesis. Therefore, the development of anticancer drugs targeting the PI3K and mTOR pathways has become a research hotspot in cancer treatment. In this study, the PI3K selective inhibitor GDC-0941 was selected as a lead compound, and 28 thiophenyl-triazine derivatives with aromatic urea structures were synthesized based on scaffold hopping, serving as a novel class of PI3K/mTOR dual inhibitors. The most promising compound Y-2 was obtained through antiproliferative activity evaluation, kinase inhibition, and toxicity assays. The results showed that Y-2 demonstrated potential inhibitory effects on both PI3K kinase and mTOR kinase, with IC50 values of 171.4 and 10.2 nM, respectively. The inhibitory effect of Y-2 on mTOR kinase was 52 times greater than that of the positive drug GDC-0941. Subsequently, the antitumor activity of Y-2 was verified through pharmacological experiments such as AO staining, cell apoptosis, scratch assays, and cell colony formation. The antitumor mechanism of Y-2 was further investigated through JC-1 experiments, real-time quantitative PCR, and Western blot analysis. Based on the above experiments, Y-2 can be identified as a potent PI3K/mTOR dual inhibitor for cancer treatment.
Collapse
Affiliation(s)
- Yang Yang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, China
| | - Xin Sun
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, China
| | - Leixuan Luo
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, China
| | - Rujue Peng
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, China
| | - Ruiqing Yang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, China
| | - Zhenjie Cheng
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, China
| | - Yao Lv
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, China
| | - Hongfeng Li
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, China
| | - Qidong Tang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, China
| | - Wufu Zhu
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, China
| | - Dan Qiao
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, China
| | - Shan Xu
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, China
| |
Collapse
|
4
|
Dahut M, Fousek K, Horn LA, Angstadt S, Qin H, Hamilton DH, Schlom J, Palena C. Fulvestrant increases the susceptibility of enzalutamide-resistant prostate cancer cells to NK-mediated lysis. J Immunother Cancer 2023; 11:e007386. [PMID: 37678915 PMCID: PMC10496692 DOI: 10.1136/jitc-2023-007386] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/08/2023] [Indexed: 09/09/2023] Open
Abstract
BACKGROUND Enzalutamide, a next-generation antiandrogen agent, is approved for the treatment of metastatic castration-resistant prostate cancer (CRPC). While enzalutamide has been shown to improve time to progression and extend overall survival in men with CRPC, the majority of patients ultimately develop resistance to treatment. Immunotherapy approaches have shown limited clinical benefit in this patient population; understanding resistance mechanisms could help develop novel and more effective treatments for CRPC. One of the mechanisms involved in tumor resistance to various therapeutics is tumor phenotypic plasticity, whereby carcinoma cells acquire mesenchymal features with or without the loss of classical epithelial characteristics. This work investigated a potential link between enzalutamide resistance, tumor phenotypic plasticity, and resistance to immune-mediated lysis in prostate cancer. METHODS Models of prostate cancer resistant to enzalutamide were established by long-term exposure of human prostate cancer cell lines to the drug in culture. Tumor cells were evaluated for phenotypic features in vitro and in vivo, as well as for sensitivity to immune effector cell-mediated cytotoxicity. RESULTS Resistance to enzalutamide was associated with gain of mesenchymal tumor features, upregulation of estrogen receptor expression, and significantly reduced tumor susceptibility to natural killer (NK)-mediated lysis, an effect that was associated with decreased tumor/NK cell conjugate formation with enzalutamide-resistant cells. Fulvestrant, a selective estrogen receptor degrader, restored the formation of target/NK cell conjugates and increased susceptibility to NK cell lysis in vitro. In vivo, fulvestrant demonstrated antitumor activity against enzalutamide-resistant cells, an effect that was associated with activation of NK cells. CONCLUSION NK cells are emerging as a promising therapeutic approach in prostate cancer. Modifying tumor plasticity via blockade of estrogen receptor with fulvestrant may offer an opportunity for immune intervention via NK cell-based approaches in enzalutamide-resistant CRPC.
Collapse
Affiliation(s)
- Madeline Dahut
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Kristen Fousek
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Lucas A Horn
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Shantel Angstadt
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Haiyan Qin
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Duane H Hamilton
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Jeffrey Schlom
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Claudia Palena
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
5
|
Ramesh S, Selvakumar P, Ameer MY, Lian S, Abdullah Alzarooni AIM, Ojha S, Mishra A, Tiwari A, Kaushik A, Jung YD, Chouaib S, Lakshmanan VK. State-of-the-art therapeutic strategies for targeting cancer stem cells in prostate cancer. Front Oncol 2023; 13:1059441. [PMID: 36969009 PMCID: PMC10035756 DOI: 10.3389/fonc.2023.1059441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 01/30/2023] [Indexed: 03/11/2023] Open
Abstract
The development of new therapeutic strategies is on the increase for prostate cancer stem cells, owing to current standardized therapies for prostate cancer, including chemotherapy, androgen deprivation therapy (ADT), radiotherapy, and surgery, often failing because of tumor relapse ability. Ultimately, tumor relapse develops into advanced castration-resistant prostate cancer (CRPC), which becomes an irreversible and systemic disease. Hence, early identification of the intracellular components and molecular networks that promote prostate cancer is crucial for disease management and therapeutic intervention. One of the potential therapeutic methods for aggressive prostate cancer is to target prostate cancer stem cells (PCSCs), which appear to be a primary focal point of cancer metastasis and recurrence and are resistant to standardized therapies. PCSCs have also been documented to play a major role in regulating tumorigenesis, sphere formation, and the metastasis ability of prostate cancer with their stemness features. Therefore, the current review highlights the origin and identification of PCSCs and their role in anti-androgen resistance, as well as stemness-related signaling pathways. In addition, the review focuses on the current advanced therapeutic strategies for targeting PCSCs that are helping to prevent prostate cancer initiation and progression, such as microRNAs (miRNAs), nanotechnology, chemotherapy, immunotherapy, the clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) gene-editing system, and photothermal ablation (PTA) therapy.
Collapse
Affiliation(s)
- Saravanan Ramesh
- Prostate Cancer Biomarker Laboratory, Faculty of Clinical Research, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| | - Preethi Selvakumar
- Prostate Cancer Biomarker Laboratory, Faculty of Clinical Research, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| | - Mohamed Yazeer Ameer
- Prostate Cancer Biomarker Laboratory, Faculty of Clinical Research, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| | - Sen Lian
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | | | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Anshuman Mishra
- Translational Research & Sustainable Healthcare Management, Institute of Advanced Materials, IAAM, Ulrika, Sweden
| | - Ashutosh Tiwari
- Translational Research & Sustainable Healthcare Management, Institute of Advanced Materials, IAAM, Ulrika, Sweden
| | - Ajeet Kaushik
- NanoBioTech Laboratory, Department of Environmental Engineering, Florida Polytechnic University, Lakeland, FL, United States
- School of Engineering, University of Petroleum and Energy Studies (UPES), Dehradun, India
| | - Young Do Jung
- Department of Biochemistry, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Salem Chouaib
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
- INSERM UMR1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy, Equipe Labellisée par la Ligue Contre le Cancer, EPHE, Faculté de Médecine, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Vinoth-Kumar Lakshmanan
- Prostate Cancer Biomarker Laboratory, Faculty of Clinical Research, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
- Translational Research & Sustainable Healthcare Management, Institute of Advanced Materials, IAAM, Ulrika, Sweden
- *Correspondence: Vinoth-Kumar Lakshmanan,
| |
Collapse
|
6
|
Costa G, Ribeiro FF, Sebastião AM, Muir EM, Vaz SH. Bridging the gap of axonal regeneration in the central nervous system: A state of the art review on central axonal regeneration. Front Neurosci 2022; 16:1003145. [PMID: 36440273 PMCID: PMC9682039 DOI: 10.3389/fnins.2022.1003145] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 10/19/2022] [Indexed: 08/26/2023] Open
Abstract
Neuronal regeneration in the central nervous system (CNS) is an important field of research with relevance to all types of neuronal injuries, including neurodegenerative diseases. The glial scar is a result of the astrocyte response to CNS injury. It is made up of many components creating a complex environment in which astrocytes play various key roles. The glial scar is heterogeneous, diverse and its composition depends upon the injury type and location. The heterogeneity of the glial scar observed in different situations of CNS damage and the consequent implications for axon regeneration have not been reviewed in depth. The gap in this knowledge will be addressed in this review which will also focus on our current understanding of central axonal regeneration and the molecular mechanisms involved. The multifactorial context of CNS regeneration is discussed, and we review newly identified roles for components previously thought to solely play an inhibitory role in central regeneration: astrocytes and p75NTR and discuss their potential and relevance for deciding therapeutic interventions. The article ends with a comprehensive review of promising new therapeutic targets identified for axonal regeneration in CNS and a discussion of novel ways of looking at therapeutic interventions for several brain diseases and injuries.
Collapse
Affiliation(s)
- Gonçalo Costa
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Faculdade de Medicina, Universidade do Porto, Porto, Portugal
| | - Filipa F. Ribeiro
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Ana M. Sebastião
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Elizabeth M. Muir
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Sandra H. Vaz
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
7
|
Tong D. Selective estrogen receptor modulators contribute to prostate cancer treatment by regulating the tumor immune microenvironment. J Immunother Cancer 2022; 10:jitc-2021-002944. [PMID: 35383112 PMCID: PMC8984050 DOI: 10.1136/jitc-2021-002944] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2022] [Indexed: 11/19/2022] Open
Abstract
Prostate cancer (PC) has previously been established as a cold tumor and develops in an inert immunosuppressive environment. Current research focuses on altering the immune microenvironment of PC from cold to hot; thus, in the present review, the diverse roles of estrogen and estrogen receptor (ER) signaling was examined in the tumor cell and tumor immune microenvironment (TIM). We hypothesized that ERα promotes PC progression and ERβ impedes epithelial-mesenchymal transition in PC cells, while in the TIM, ERβ mediates the immunosuppressive environment, and low levels of ERα is associated with disease development. Selective estrogen receptor modulators (SERMs) or selective ER degraders play diverse roles in the regulation of ER isoforms. Patients with PC may benefit from the use of SERMs, including raloxifene, in combination with anti-PD1/PD-L1 checkpoint immunotherapy, or TGF-β or Wnt antagonists. The present review demonstrated that immunotherapy-based strategies combined with SERMs may be an option for the future of PC-targeting therapy.
Collapse
Affiliation(s)
- Dali Tong
- Department of Urological Surgery, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| |
Collapse
|
8
|
Lacouture A, Lafront C, Peillex C, Pelletier M, Audet-Walsh É. Impacts of endocrine-disrupting chemicals on prostate function and cancer. ENVIRONMENTAL RESEARCH 2022; 204:112085. [PMID: 34562481 DOI: 10.1016/j.envres.2021.112085] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 09/15/2021] [Accepted: 09/16/2021] [Indexed: 06/13/2023]
Abstract
Because of their historical mode of action, endocrine-disrupting chemicals (EDCs) are associated with sex-steroid receptors, namely the two estrogen receptors (ERα and ERβ) and the androgen receptor (AR). Broadly, EDCs can modulate sex-steroid receptor functions. They can also indirectly impact the androgen and estrogen pathways by influencing steroidogenesis, expression of AR or ERs, and their respective activity as transcription factors. Additionally, many of these chemicals have multiple cellular targets other than sex-steroid receptors, which results in a myriad of potential effects in humans. The current article reviews the association between prostate cancer and the endocrine-disrupting functions of four prominent EDC families: bisphenols, phthalates, phytoestrogens, and mycoestrogens. Results from both in vitro and in vivo models are included and discussed to better assess the molecular mechanisms by which EDCs can modify prostate biology. To overcome the heterogeneity of results published, we established common guidelines to properly study EDCs in the context of endocrine diseases. Firstly, the expression of sex-steroid receptors in the models used must be determined before testing. Then, in parallel to EDCs, pharmacological compounds acting as positive (agonists) and negative controls (antagonists) have to be employed. Finally, EDCs need to be used in a precise range of concentrations to modulate sex-steroid receptors and avoid off-target effects. By adequately integrating molecular endocrinology aspects in EDC studies and identifying their underlying molecular mechanisms, we will truly understand their impact on prostate cancer and distinguish those that favor the progression of the disease from those that slow down tumor development.
Collapse
Affiliation(s)
- Aurélie Lacouture
- Department of Molecular Medicine, Faculty of Medicine, Laval University, Québec, Canada; Endocrinology - Nephrology Research Axis, CHU de Québec-Université Laval Research Center, Québec, Canada; Cancer Research Center (CRC), Laval University, Québec, Canada
| | - Camille Lafront
- Department of Molecular Medicine, Faculty of Medicine, Laval University, Québec, Canada; Endocrinology - Nephrology Research Axis, CHU de Québec-Université Laval Research Center, Québec, Canada; Cancer Research Center (CRC), Laval University, Québec, Canada
| | - Cindy Peillex
- Infectious and Immune Diseases Research Axis, CHU de Québec-Université Laval Research Center, Québec, Canada; ARThrite Research Center, Laval University, Québec, Canada; Master de Biologie, École Normale Supérieure de Lyon, Université Claude Bernard Lyon I, Université de Lyon, Lyon, France
| | - Martin Pelletier
- Infectious and Immune Diseases Research Axis, CHU de Québec-Université Laval Research Center, Québec, Canada; ARThrite Research Center, Laval University, Québec, Canada; Department of Microbiology-Infectious Diseases and Immunology, Faculty of Medicine, Laval University, Québec, Canada.
| | - Étienne Audet-Walsh
- Department of Molecular Medicine, Faculty of Medicine, Laval University, Québec, Canada; Endocrinology - Nephrology Research Axis, CHU de Québec-Université Laval Research Center, Québec, Canada; Cancer Research Center (CRC), Laval University, Québec, Canada.
| |
Collapse
|
9
|
Lafront C, Germain L, Weidmann C, Audet-Walsh É. A Systematic Study of the Impact of Estrogens and Selective Estrogen Receptor Modulators on Prostate Cancer Cell Proliferation. Sci Rep 2020; 10:4024. [PMID: 32132580 PMCID: PMC7055213 DOI: 10.1038/s41598-020-60844-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 02/17/2020] [Indexed: 01/10/2023] Open
Abstract
The estrogen signaling pathway has been reported to modulate prostate cancer (PCa) progression through the activity of estrogen receptors α and β (ERα and ERβ). Given that selective estrogen receptor modulators (SERMs) are used to treat breast cancer, ERs have been proposed as attractive therapeutic targets in PCa. However, many inconsistencies regarding the expression of ERs and the efficacy of SERMs for PCa treatment exist, notably due to the use of ERβ antibodies lacking specificity and treatments with high SERM concentrations leading to off-target effects. To end this confusion, our objective was to study the impact of estrogenic and anti-estrogenic ligands in well-studied in vitro PCa models with appropriate controls, dosages, and ER subtype-specific antibodies. When using physiologically relevant concentrations of nine estrogenic/anti-estrogenic compounds, including five SERMs, we observed no significant modulation of PCa cell proliferation. Using RNA-seq and validated antibodies, we demonstrate that these PCa models do not express ERs. In contrast, RNA-seq from PCa samples from patients have detectable expression of ERα. Overall, our study reveals that commonly used PCa models are inappropriate to study ERs and indicate that usage of alternative models is essential to properly assess the roles of the estrogen signaling pathway in PCa.
Collapse
Affiliation(s)
- Camille Lafront
- Department of molecular medicine, Faculty of Medicine, Université Laval, Québec City, G1V 0A6, Canada
- Endocrinology - Nephrology Research Axis, Centre de recherche du CHU de Québec - Université Laval, Québec City, Canada
- Centre de recherche sur le cancer (CRC) of Université Laval, Québec City, Canada
| | - Lucas Germain
- Endocrinology - Nephrology Research Axis, Centre de recherche du CHU de Québec - Université Laval, Québec City, Canada
- Centre de recherche sur le cancer (CRC) of Université Laval, Québec City, Canada
- Department of biochemistry, microbiology and bioinformatics, Faculty of Sciences and Engineering, Université Laval, Québec City, G1V 0A6, Canada
| | - Cindy Weidmann
- Endocrinology - Nephrology Research Axis, Centre de recherche du CHU de Québec - Université Laval, Québec City, Canada
- Centre de recherche sur le cancer (CRC) of Université Laval, Québec City, Canada
| | - Étienne Audet-Walsh
- Department of molecular medicine, Faculty of Medicine, Université Laval, Québec City, G1V 0A6, Canada.
- Endocrinology - Nephrology Research Axis, Centre de recherche du CHU de Québec - Université Laval, Québec City, Canada.
- Centre de recherche sur le cancer (CRC) of Université Laval, Québec City, Canada.
| |
Collapse
|
10
|
Ventral prostate and mammary gland phenotype in mice with complete deletion of the ERβ gene. Proc Natl Acad Sci U S A 2020; 117:4902-4909. [PMID: 32075916 PMCID: PMC7060692 DOI: 10.1073/pnas.1920478117] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Disagreements about the phenotype of estrogen receptor β (ERβ) knockout mouse, created by removing the DNA-binding domain of the ERβ gene or interruption of the gene with a neocassette (Oliver Smithies ERβ knockout mice [ERβOS-/-]), prompted us to create an ERβ knockout mouse by deleting the ERβ gene with the use of CRISPR/Cas9 technology. We confirmed that the ERβ gene was eliminated from the mouse genome and that no ERβ mRNA or protein was detectable in tissues of this mouse. Overall the phenotype of the ventral prostate (VP) and mammary gland (MG) in ERβcrispr-/- mice was similar to, but more severe than, that in the ERβOS-/-mice. In the VP of 6-mo-old ERβcrispr-/- mice there was epithelial hyperplasia, fibroplasia, inflammation, stromal overgrowth, and intraductal cancer-like lesions. This was accompanied by an increase in Ki67 and P63 and loss in DACH1 and PURα, two androgen receptor (AR) repressors. In the MG there was overexpression of estrogen receptor α and progesterone receptor, loss of collagen, increase in proliferation and expression of metalloproteases, and invasive epithelium. Surprisingly, by 18 mo of age, the number of hyperplastic foci was reduced, the ducts of the VP and MG became atrophic, and, in the VP, there was massive immune infiltration and massive desquamation of the luminal epithelial cells. These changes were coincident with reduced levels of androgens in males and estrogens in females. We conclude that ERβ is a tumor suppressor gene in the VP and MG where its loss increases the activity AR and ERα, respectively.
Collapse
|
11
|
ZFHX3 is indispensable for ERβ to inhibit cell proliferation via MYC downregulation in prostate cancer cells. Oncogenesis 2019; 8:28. [PMID: 30979864 PMCID: PMC6461672 DOI: 10.1038/s41389-019-0138-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 03/20/2019] [Indexed: 12/22/2022] Open
Abstract
Both estrogen receptor 2 (ESR2, also known as estrogen receptor beta (ERβ)) and the zinc-finger homeobox 3 (ZFHX3, also known as ATBF1 for AT motif-binding factor 1) modulate prostate development and suppress prostatic tumorigenesis in mice. ZFHX3 is integral to proper functions of ESR1 (i.e., estrogen receptor alpha (ERα)), which belongs to the same family of proteins as ESR2, but is hardly expressed in prostate epithelial cells. It is not clear how ZFHX3 suppresses prostatic tumorigenesis. In this study, we investigated whether ZFHX3 and ERβ functionally interact with each other in the suppression of prostatic tumorigenesis. In two androgen receptor (AR)-positive prostate cancer cell lines, C4-2B and LNCaP, we first validated ERβ’s tumor suppressor activity indicated by the inhibition of cell proliferation and repression of MYC expression. We found that loss of ZFHX3 increased cell proliferation and MYC expression, and downregulation of MYC was necessary for ZFHX3 to inhibit cell proliferation in the same cell lines. Importantly, loss of ZFHX3 prevented ERβ from suppressing cell proliferation and repressing MYC transcription. Biochemically, ERβ and ZFHX3 physically interacted with each other and they both occupied the same region of the common MYC promoter, even though ZFHX3 also bound to another region of the MYC promoter. Higher levels of ZFHX3 and ERβ in human prostate cancer tissue samples correlated with better patient survival. These findings establish MYC repression as a mechanism for ZFHX3’s tumor suppressor activity and ZFHX3 as an indispensable factor for ERβ’s tumor suppressor activity in prostate cancer cells. Our data also suggest that intact ZFHX3 function is required for using ERβ-selective agonists to effectively treat prostate cancer.
Collapse
|
12
|
In Vivo Expression of miR-32 Induces Proliferation in Prostate Epithelium. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:2546-2557. [DOI: 10.1016/j.ajpath.2017.07.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 06/25/2017] [Accepted: 07/13/2017] [Indexed: 12/19/2022]
|
13
|
Geybels MS, Fang M, Wright JL, Qu X, Bibikova M, Klotzle B, Fan JB, Feng Z, Ostrander EA, Nelson PS, Stanford JL. PTEN loss is associated with prostate cancer recurrence and alterations in tumor DNA methylation profiles. Oncotarget 2017; 8:84338-84348. [PMID: 29137428 PMCID: PMC5663600 DOI: 10.18632/oncotarget.20940] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Accepted: 07/08/2017] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Prostate cancer (PCa) with loss of the tumor suppressor gene PTEN has an unfavorable prognosis. DNA methylation profiles associated with PTEN loss may provide further insights into the mechanisms underlying these more aggressive, clinically relevant tumors. METHODS The cohort included patients with clinically localized PCa. Samples taken from the primary tumor were used to determine PTEN genomic deletions using FISH, and to analyze epigenome-wide DNA methylation profiles. Patients were followed for PCa recurrence on average for 8 years after diagnosis. RESULTS The study included 471 patients with data on PTEN loss, and the frequency of hemi- and homozygous PTEN loss was 10.0% and 4.5%, respectively. Loss of PTEN was associated with a significantly higher risk of recurrence (any vs. no PTEN loss; HR = 1.74; 95% CI: 1.03-2.93). Hazard ratios for hemi- and homozygous loss were 1.39 (95% CI: 0.73-2.64) and 2.84 (95% CI: 1.30-6.19), respectively. Epigenome-wide methylation profiling identified 4,208 differentially methylated CpGs (FDR Q-value < 0.01) in tumors with any versus no PTEN loss. There were no genome-wide significant differentially methylated CpGs in homo- versus hemizygous deleted tumors. Tumor methylation data were used to build a methylation signature of PTEN loss in our cohort, which was confirmed in TCGA, and included CpGs in ATP11A, GDNF, JAK1, JAM3, and VAPA. CONCLUSION Loss of PTEN was positively associated with PCa recurrence. Prostate tumors with PTEN loss harbor a distinct methylation signature, and these aberrantly methylated CpG sites may mediate tumor progression when PTEN is deleted.
Collapse
Affiliation(s)
- Milan S. Geybels
- Department of Epidemiology, GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
- Division of Public Health Sciences, Fred Hutchison Cancer Research Center, Seattle, Washington, USA
| | - Min Fang
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Jonathan L. Wright
- Division of Public Health Sciences, Fred Hutchison Cancer Research Center, Seattle, Washington, USA
- Department of Urology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Xiaoyu Qu
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Cytogenetics, Seattle Cancer Care Alliance, Seattle, Washington, USA
| | - Marina Bibikova
- Department of Oncology, Illumina, Inc., San Diego, California, USA
| | - Brandy Klotzle
- Department of Oncology, Illumina, Inc., San Diego, California, USA
| | - Jian-Bing Fan
- Department of Oncology, Illumina, Inc., San Diego, California, USA
- Current address: AnchorDx Corp., Guangzhou 510300, People's Republic of China
| | - Ziding Feng
- Department of Biostatistics, MD Anderson Cancer Center, Houston, Texas, USA
| | - Elaine A. Ostrander
- Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland, USA
| | - Peter S. Nelson
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| | - Janet L. Stanford
- Division of Public Health Sciences, Fred Hutchison Cancer Research Center, Seattle, Washington, USA
- Department of Epidemiology, University of Washington School of Public Health, Seattle, Washington, USA
| |
Collapse
|
14
|
Tao X, Xu L, Yin L, Han X, Qi Y, Xu Y, Song S, Zhao Y, Peng J. Dioscin induces prostate cancer cell apoptosis through activation of estrogen receptor-β. Cell Death Dis 2017; 8:e2989. [PMID: 28796245 PMCID: PMC5596577 DOI: 10.1038/cddis.2017.391] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 06/22/2017] [Accepted: 07/10/2017] [Indexed: 12/31/2022]
Abstract
Recent researches have shown that estrogen receptor-β (ERβ) activator may be a potent anticancer agent for prostate cancer (PCa), and our previous study also indicated that dioscin can upregulate the expression of ERβ in MC3T3-E1 cell. In the present work, the activity and mechanism of dioscin, a natural product, against PCa were investigated. The results showed that dioscin markedly inhibited cell viability, colony formation, motility and induced apoptosis in PC3 cells. Moreover, dioscin disrupted the formation of PC3 cell-derived mammospheres and reduced aldehyde dehydrogenase (ALDH) level and the CD133+/CD44+ cells, indicating that dioscin had a potent inhibitory activity on prostate cancer stem cells (PCSCs). In vivo results also showed that dioscin significantly suppressed the tumor growth of PC3 cell xenografts in nude mice. Furthermore, mechanism investigation showed that dioscin markedly upregulated ERβ expression level, subsequently increased prolyl hydroxylase 2 level, decreased the levels of hypoxia-inducible factor-1α, vascular endothelial growth factor A and BMI-1, and thus induced cell apoptosis by regulating the expression levels of caspase-3 and Bcl-2 family proteins. In addition, transfection experiment of ERβ-siRNA further indicated that diosicn showed excellent activity against PCa in vitro and in vivo by increasing ERβ expression level. The co-immunoprecipitation (Co-IP) results further suggested that dioscin promoted the interaction of c-ABL and ERβ, but did not change c-ABL expression. Moreover, the molecular docking assay showed that dioscin processed powerful affinity toward to ERβ mainly through the strong hydrogen bonding and hydrophobic effects, and the actions of dioscin on ERβ activation and tumor cells inhibition were significantly weakened in the mutational (Phe-336, Phe-468) PC3 cells. Collectively, these findings proved that dioscin exerted efficient anti-PCa activity via activation of ERβ, which should be developed as an efficient candidate in clinical for treating this cancer in the future.
Collapse
Affiliation(s)
- Xufeng Tao
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Lina Xu
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Lianhong Yin
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Xu Han
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Yan Qi
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Youwei Xu
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Shasha Song
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Yanyan Zhao
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Jinyong Peng
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| |
Collapse
|
15
|
Perez PA, Petiti JP, Picech F, Guido CB, dV Sosa L, Grondona E, Mukdsi JH, De Paul AL, Torres AI, Gutierrez S. Estrogen receptor β regulates the tumoral suppressor PTEN to modulate pituitary cell growth. J Cell Physiol 2017; 233:1402-1413. [PMID: 28542730 DOI: 10.1002/jcp.26025] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 05/19/2017] [Indexed: 01/16/2023]
Abstract
In this study, we focused on ERβ regulation in the adenohypophysis under different estrogenic milieu, by analyzing whether ER modulates the phosphatase and tensin homolog deleted on chromosome 10 (PTEN) expression and its subcellular localization on anterior pituitary glands from Wistar rats and GH3 lactosomatotroph cells that over-expressed ERβ. ERβ was regulated in a cyclic manner, and underwent dynamic changes throughout the estrous cycle, with decreased ERβ+ cells in estrus and under E2 treatment, but increased in ovariectomized rats. In addition, the ERα/β ratio increased in estrus and under E2 stimulation, but decreased in ovariectomized rats. Double immunofluorescence revealed that lactotroph and somatotroph ERβ+ were significantly decreased in estrus. Also, variations in the PTEN expression was observed, which was diminished with high E2 conditions but augmented with low E2 milieu. The subcellular localization of this phosphatase was cell cycle-dependent, with remarkable changes in the immunostaining pattern: nuclear in arrested pituitary cells but cytoplasmic in stimulated cells, and responding differently to ER agonists, with only DPN being able to increase PTEN expression and retaining it in the nucleus. Finally, ERβ over-expression increased PTEN with a noticeable subcellular redistribution, and with a significant nuclear signal increase in correlation with an increase of cells in G0/G1 phase. These results showed that E2 is able to inhibit ERβ expression and suggests that the tumoral suppressor PTEN might be one of the signaling proteins by which E2, through ERβ, acts to modulate pituitary cell proliferation, thereby adapting endocrine populations in relation with hormonal necessities.
Collapse
Affiliation(s)
- Pablo A Perez
- Centro de Microscopia Electrónica, Instituto de Investigaciones en Ciencias de la Salud (INICSA-CONICET), Facultad de Ciencias Medicas, Universidad Nacional de Cordoba, Cordoba, Argentina
| | | | - Florencia Picech
- Centro de Microscopia Electrónica, Instituto de Investigaciones en Ciencias de la Salud (INICSA-CONICET), Facultad de Ciencias Medicas, Universidad Nacional de Cordoba, Cordoba, Argentina
| | - Carolina B Guido
- Centro de Microscopia Electrónica, Instituto de Investigaciones en Ciencias de la Salud (INICSA-CONICET), Facultad de Ciencias Medicas, Universidad Nacional de Cordoba, Cordoba, Argentina
| | - Liliana dV Sosa
- Centro de Microscopia Electrónica, Instituto de Investigaciones en Ciencias de la Salud (INICSA-CONICET), Facultad de Ciencias Medicas, Universidad Nacional de Cordoba, Cordoba, Argentina
| | - Ezequiel Grondona
- Centro de Microscopia Electrónica, Instituto de Investigaciones en Ciencias de la Salud (INICSA-CONICET), Facultad de Ciencias Medicas, Universidad Nacional de Cordoba, Cordoba, Argentina
| | - Jorge H Mukdsi
- Centro de Microscopia Electrónica, Instituto de Investigaciones en Ciencias de la Salud (INICSA-CONICET), Facultad de Ciencias Medicas, Universidad Nacional de Cordoba, Cordoba, Argentina
| | - Ana L De Paul
- Centro de Microscopia Electrónica, Instituto de Investigaciones en Ciencias de la Salud (INICSA-CONICET), Facultad de Ciencias Medicas, Universidad Nacional de Cordoba, Cordoba, Argentina
| | - Alicia I Torres
- Centro de Microscopia Electrónica, Instituto de Investigaciones en Ciencias de la Salud (INICSA-CONICET), Facultad de Ciencias Medicas, Universidad Nacional de Cordoba, Cordoba, Argentina
| | - Silvina Gutierrez
- Centro de Microscopia Electrónica, Instituto de Investigaciones en Ciencias de la Salud (INICSA-CONICET), Facultad de Ciencias Medicas, Universidad Nacional de Cordoba, Cordoba, Argentina
| |
Collapse
|
16
|
Roubaud G, Liaw BC, Oh WK, Mulholland DJ. Strategies to avoid treatment-induced lineage crisis in advanced prostate cancer. Nat Rev Clin Oncol 2017; 14:269-283. [PMID: 27874061 PMCID: PMC5567685 DOI: 10.1038/nrclinonc.2016.181] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The increasing potency of therapies that target the androgen receptor (AR) signalling axis has correlated with a rise in the proportion of patients with prostate cancer harbouring an adaptive phenotype, termed treatment-induced lineage crisis. This phenotype is characterized by features that include soft-tissue metastasis and/or resistance to standard anticancer therapies. Potent anticancer treatments might force cancer cells to evolve and develop alternative cell lineages that are resistant to primary therapies, a mechanism similar to the generation of multidrug- resistant microorganisms after continued antibiotic use. Herein, we assess the hypothesis that treatment-adapted phenotypes harbour reduced AR expression and/or activity, and acquire compensatory strategies for cell survival. We highlight the striking similarities between castration-resistant prostate cancer and triple-negative breast cancer, another poorly differentiated endocrine malignancy. Alternative treatment paradigms are needed to avoid therapy-induced resistance. Herein, we present a new clinical trial strategy designed to evaluate the potential of rapid drug cycling as an approach to delay the onset of resistance and treatment-induced lineage crisis in patients with metastatic castration-resistant prostate cancer.
Collapse
Affiliation(s)
- Guilhem Roubaud
- Department of Medical Oncology, Institut Bergonié, 229 Cours de l'Argonne, Bordeaux 33076, France
| | - Bobby C Liaw
- Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, 1470 Madison Avenue, New York, New York 10029, USA
| | - William K Oh
- Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, 1470 Madison Avenue, New York, New York 10029, USA
| | - David J Mulholland
- Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, 1470 Madison Avenue, New York, New York 10029, USA
| |
Collapse
|
17
|
Nelson AW, Groen AJ, Miller JL, Warren AY, Holmes KA, Tarulli GA, Tilley WD, Katzenellenbogen BS, Hawse JR, Gnanapragasam VJ, Carroll JS. Comprehensive assessment of estrogen receptor beta antibodies in cancer cell line models and tissue reveals critical limitations in reagent specificity. Mol Cell Endocrinol 2017; 440:138-150. [PMID: 27889472 PMCID: PMC5228587 DOI: 10.1016/j.mce.2016.11.016] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 11/01/2016] [Accepted: 11/20/2016] [Indexed: 11/20/2022]
Abstract
Estrogen Receptor-β (ERβ) has been implicated in many cancers. In prostate and breast cancer its function is controversial, but genetic studies implicate a role in cancer progression. Much of the confusion around ERβ stems from antibodies that are inadequately validated, yet have become standard tools for deciphering its role. Using an ERβ-inducible cell system we assessed commonly utilized ERβ antibodies and show that one of the most commonly used antibodies, NCL-ER-BETA, is non-specific for ERβ. Other antibodies have limited ERβ specificity or are only specific in one experimental modality. ERβ is commonly studied in MCF-7 (breast) and LNCaP (prostate) cancer cell lines, but we found no ERβ expression in either, using validated antibodies and independent mass spectrometry-based approaches. Our findings question conclusions made about ERβ using the NCL-ER-BETA antibody, or LNCaP and MCF-7 cell lines. We describe robust reagents, which detect ERβ across multiple experimental approaches and in clinical samples.
Collapse
Affiliation(s)
- Adam W Nelson
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, CB2 ORE, UK; Academic Urology Group, Department of Surgery, University of Cambridge, Cambridge, CB2 0QQ, UK; Department of Urology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge, CB2 0QQ, UK
| | - Arnoud J Groen
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, CB2 ORE, UK
| | - Jodi L Miller
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, CB2 ORE, UK
| | - Anne Y Warren
- Department of Histopathology, Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge, CB2 0QQ, UK
| | - Kelly A Holmes
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, CB2 ORE, UK
| | - Gerard A Tarulli
- Dame Roma Mitchell Cancer Research Laboratories, Hanson Institute Building, School of Medicine, Faculty of Health Sciences, The University of Adelaide, SA 5005, Australia
| | - Wayne D Tilley
- Dame Roma Mitchell Cancer Research Laboratories, Hanson Institute Building, School of Medicine, Faculty of Health Sciences, The University of Adelaide, SA 5005, Australia
| | - Benita S Katzenellenbogen
- Departments of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - John R Hawse
- Department of Biochemistry and Molecular Biology, Mayo Clinic, 200 1st Street SW, Rochester, MN 55905 USA
| | - Vincent J Gnanapragasam
- Academic Urology Group, Department of Surgery, University of Cambridge, Cambridge, CB2 0QQ, UK; Department of Urology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge, CB2 0QQ, UK
| | - Jason S Carroll
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, CB2 ORE, UK.
| |
Collapse
|
18
|
Kozłowska E, Puszynski K. Application of bifurcation theory and siRNA-based control signal to restore the proper response of cancer cells to DNA damage. J Theor Biol 2016; 408:213-221. [DOI: 10.1016/j.jtbi.2016.08.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 07/17/2016] [Accepted: 08/10/2016] [Indexed: 10/21/2022]
|
19
|
Mak P, Li J, Samanta S, Mercurio AM. ERβ regulation of NF-kB activation in prostate cancer is mediated by HIF-1. Oncotarget 2016; 6:40247-54. [PMID: 26450901 PMCID: PMC4741892 DOI: 10.18632/oncotarget.5377] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 09/21/2015] [Indexed: 12/26/2022] Open
Abstract
We examined the regulation of NF-κB in prostate cancer by estrogen receptor β (ERβ) based on the inverse correlation between p65 and ERβ expression that exists in prostate carcinomas and reports that ERβ can inhibit NF-κB activation, although the mechanism is not known. We demonstrate that ERβ functions as a gate-keeper for NF-κB p65 signaling by repressing its expression and nuclear translocation. ERβ regulation of NF-κB signaling is mediated by HIF-1. Loss of ERβ or hypoxia stabilizes HIF-1α, which we found to be a direct driver of IKKβ transcription through a hypoxia response element present in the promoter of the IKKβ gene. The increase of IKKβ expression in ERβ-ablated cells correlates with an increase in phospho-IκBα and concomitant p65 nuclear translocation. An inverse correlation between the expression of ERβ and IKKβ/p65 was also observed in the prostates of ERβ knockout (BERKO) mice, Gleason grade 5 prostate tumors and analysis of prostate cancer databases. These findings provide a novel mechanism for how ERβ prevents NF-κB activation and raise the exciting possibility that loss of ERβ expression is linked to chronic inflammation in the prostate, which contributes to the development of high-grade prostate cancer.
Collapse
Affiliation(s)
- Paul Mak
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Jiarong Li
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Sanjoy Samanta
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Arthur M Mercurio
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| |
Collapse
|
20
|
Kowalska K, Piastowska-Ciesielska AW. Oestrogens and oestrogen receptors in prostate cancer. SPRINGERPLUS 2016; 5:522. [PMID: 27186486 PMCID: PMC4844569 DOI: 10.1186/s40064-016-2185-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 04/19/2016] [Indexed: 03/25/2023]
Abstract
The role of androgens in prostate cancer is obvious due to the fact that androgen signalling is the main regulator of prostate growth and function. Androgen deprivation therapy is a mainstay treatment for advanced prostate cancer. However, prostate cancer often becomes androgen-independent, which in consequence leads to lethal and incurable disease. In addition, oestrogens play a crucial role in prostate cancer, especially in elder men in whom the overall ratio of oestrogens to androgens is increasing. This review summarizes the current knowledge on molecular mechanisms through which oestrogens are involved in prostate cancer development. We focused on commonly alternated molecular signalling pathways contributing to tumourgenesis in prostate cancer.
Collapse
Affiliation(s)
- Karolina Kowalska
- Department of Comparative Endocrinology, Faculty of Biomedical Sciences and Postgraduate Training, Medical University of Lodz, Zeligowskiego 7/9, 90-752 Lodz, Poland
| | - Agnieszka Wanda Piastowska-Ciesielska
- Department of Comparative Endocrinology, Faculty of Biomedical Sciences and Postgraduate Training, Medical University of Lodz, Zeligowskiego 7/9, 90-752 Lodz, Poland
| |
Collapse
|
21
|
Karamouzis MV, Papavassiliou KA, Adamopoulos C, Papavassiliou AG. Targeting Androgen/Estrogen Receptors Crosstalk in Cancer. Trends Cancer 2016; 2:35-48. [PMID: 28741499 DOI: 10.1016/j.trecan.2015.12.001] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 12/01/2015] [Accepted: 12/02/2015] [Indexed: 01/04/2023]
Abstract
The actions of estrogens are mediated by estrogen receptors, ERα and ERβ. Recent genomic landscaping of ERα- and ERβ-binding sites has revealed important distinctions regarding their transcriptional activity. ERβ and its isoforms have been correlated with endocrine treatment responsiveness in breast tumors, while post-translational modifications, receptor dimerization patterns, and subcellular localization are increasingly recognized as crucial modulators in prostate carcinogenesis. Androgen receptor (AR) is essential for the development and progression of prostate cancer as well as of certain breast cancer types. The balance between the activity of these two hormone receptors and their molecular interactions in different clinical settings is influenced by several coregulators. This comprises a dynamic regulatory network enhancing or limiting the activity of AR-directed treatments in breast and prostate tumorigenesis. In this review, we discuss the molecular background regarding the therapeutic targeting of androgen/estrogen receptor crosstalk in breast and prostate cancer.
Collapse
Affiliation(s)
- Michalis V Karamouzis
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece.
| | - Kostas A Papavassiliou
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Christos Adamopoulos
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Athanasios G Papavassiliou
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece.
| |
Collapse
|
22
|
Latonen L, Kujala P, Visakorpi T. Incidence of Mucinous Metaplasia in the Prostate of FVB/N Mice (Mus musculus). Comp Med 2016; 66:286-289. [PMID: 27538859 PMCID: PMC4983170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 12/17/2015] [Accepted: 01/21/2016] [Indexed: 06/06/2023]
Abstract
Prostate epithelium in mice is considered to be relatively resistant to aged-related changes, as compared with human prostate epithelium, which is prone to spontaneous hyperplasia and cancer, for example. In addition, the incidence of metaplasia in mouse prostate typically is considered to be low. Here we report the incidence of mucinous metaplasia in the prostates of wild-type FVB/N mice. Our histologic study shows that mucinous metaplasia involving goblet cells occurs much more frequently (incidence as high as 50%) in the prostates of aged mice (17-24 mo) than has been reported previously. Mucinous metaplasia in the prostates of laboratory mice may be considerably more frequent than previously appreciated.
Collapse
Affiliation(s)
- Leena Latonen
- Institute of Biosciences and Medical Technology (BioMediTech), University of Tampere, Fimlab Laboratories, Tampere University Hospital, Tampere, Finland.
| | - Paula Kujala
- Fimlab Laboratories, Tampere University Hospital, Tampere, Finland
| | - Tapio Visakorpi
- Institute of Biosciences and Medical Technology (BioMediTech), University of Tampere, Fimlab Laboratories, Tampere University Hospital, Tampere, Finland
| |
Collapse
|
23
|
Gravina GL, Mancini A, Scarsella L, Colapietro A, Jitariuc A, Vitale F, Marampon F, Ricevuto E, Festuccia C. Dual PI3K/mTOR inhibitor, XL765 (SAR245409), shows superior effects to sole PI3K [XL147 (SAR245408)] or mTOR [rapamycin] inhibition in prostate cancer cell models. Tumour Biol 2015. [PMID: 26219891 DOI: 10.1007/s13277-015-3725-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Deregulation of phosphatidylinositol-3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) signaling pathway contributes to prostate cancer development and progression. Here, we compared the in vitro effects of the dual PI3K/mTOR inhibitor (XL765) with those observed with the sole PI3K (XL147) or mTOR (rapamycin) inhibition in 2 non-tumor prostate epithelial cell lines, 8 prostate cancer cell lines, and 11 prostate cancer cell derivatives. We demonstrated that the XL765 treatment showed superior and proliferative effects of XL147 or rapamycin. The XL765 effects were associated to increasing the chromosome region maintenance 1 (CRM1)-mediated nuclear localization of glycogen synthase kinase 3 beta (GSK3β) and Foxo-1a with higher induction of apoptosis when compared to those observed in XL147 and rapamycin treatments. IC50 values were calculated in phosphatase and tensin homologue deleted on chromosome 10 (PTEN)-positive and PTEN-negative cell lines as well as after PTEN transfection or PTEN downmodulation by siRNA strategy revealing that the presence of this protein was associated with reduced sensitivity to PI3K and mTOR inhibitors. The comparison of IC50 values was also calculated for androgen-dependent and -independent cell lines as well as after androgen receptor (AR) transfection or the AR downmodulation by siRNA strategy revealing that androgen independence was associated with enhanced responsiveness. Our results provide a rationale to use the dual PI3K/Akt/mTOR inhibitors in hormone-insensitive prostate cancer models due to the overactivity of PI3K/Akt/mTOR in this disease condition.
Collapse
Affiliation(s)
- Giovanni Luca Gravina
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Radiobiology, University of L'Aquila, L'Aquila, Italy
- Department of Biotechnological and Applied Clinical Sciences, Division of Radiation Oncology, University of L'Aquila, L'Aquila, Italy
| | - Andrea Mancini
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Radiobiology, University of L'Aquila, L'Aquila, Italy
| | - Luca Scarsella
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Radiobiology, University of L'Aquila, L'Aquila, Italy
| | - Alessandro Colapietro
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Radiobiology, University of L'Aquila, L'Aquila, Italy
| | - Ana Jitariuc
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Radiobiology, University of L'Aquila, L'Aquila, Italy
| | - Flora Vitale
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Radiobiology, University of L'Aquila, L'Aquila, Italy
| | - Francesco Marampon
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Radiobiology, University of L'Aquila, L'Aquila, Italy
| | - Enrico Ricevuto
- Department of Biotechnological and Applied Clinical Sciences, Division of Medical Oncology, University of L'Aquila, L'Aquila, Italy
| | - Claudio Festuccia
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Radiobiology, University of L'Aquila, L'Aquila, Italy.
| |
Collapse
|
24
|
Liu FT, Ou-Yang X, Zhang GP, Luo HL. Progress in research of colorectal intraepithelial neoplasia and adenoma. Shijie Huaren Xiaohua Zazhi 2015; 23:3413-3420. [DOI: 10.11569/wcjd.v23.i21.3413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer is a common malignant tumor in the digestive system, and the early diagnosis of colorectal cancer has been the focus of its prevention and control. Colorectal intraepithelial neoplasia and adenoma are considered to be the most important precancerous lesions of colorectal cancer. In recent years, with the development of biological medicine, genetics,
and other disciplines, many studies have explored the relationship between intraepithelial neoplasia and adenoma and colorectal cancer, and some new research progress has been achieved to provide some guidance for the future clinical screening, regular follow-up and chemical prevention. However, it remains to be studied how colorectal intraepithelial neoplasia and adenoma form and evolve to colorectal cancer.
Collapse
|