1
|
Cai A, Chen Y, Wang LS, Cusick JK, Shi Y. Depicting Biomarkers for HER2-Inhibitor Resistance: Implication for Therapy in HER2-Positive Breast Cancer. Cancers (Basel) 2024; 16:2635. [PMID: 39123362 PMCID: PMC11311605 DOI: 10.3390/cancers16152635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/16/2024] [Accepted: 07/19/2024] [Indexed: 08/12/2024] Open
Abstract
HER2 (human epidermal growth factor receptor 2) is highly expressed in a variety of cancers, including breast, lung, gastric, and pancreatic cancers. Its amplification is linked to poor clinical outcomes. At the genetic level, HER2 is encoded by the ERBB2 gene (v-erb-b2 avian erythroblastic leukemia viral oncogene homolog 2), which is frequently mutated or amplified in cancers, thus spurring extensive research into HER2 modulation and inhibition as viable anti-cancer strategies. An impressive body of FDA-approved drugs, including anti-HER2 monoclonal antibodies (mAbs), antibody-drug conjugates (ADCs), and HER2-tyrosine kinase inhibitors (TKIs), have demonstrated success in enhancing overall survival (OS) and disease progression-free survival (PFS). Yet, drug resistance remains a persistent challenge and raises the risks of metastatic potential and tumor relapse. Research into alternative therapeutic options for HER2+ breast cancer therefore proves critical for adapting to this ever-evolving landscape. This review highlights current HER2-targeted therapies, discusses predictive biomarkers for drug resistance, and introduces promising emergent therapies-especially combination therapies-that are aimed at overcoming drug resistance in the context of HER2+ breast cancer.
Collapse
Affiliation(s)
- Alvan Cai
- College of Medicine, California Northstate University, Elk Grove, CA 95757, USA; (A.C.); (J.K.C.)
| | - Yuan Chen
- Section Pathology of the Institute of Forensic Medicine, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany;
| | - Lily S. Wang
- University of California, Berkeley, CA 94720, USA;
| | - John K. Cusick
- College of Medicine, California Northstate University, Elk Grove, CA 95757, USA; (A.C.); (J.K.C.)
| | - Yihui Shi
- College of Medicine, California Northstate University, Elk Grove, CA 95757, USA; (A.C.); (J.K.C.)
- California Pacific Medical Center Research Institute, Sutter Bay Hospitals, San Francisco, CA 94107, USA
| |
Collapse
|
2
|
Boscolo Bielo L, Trapani D, Nicolò E, Valenza C, Guidi L, Belli C, Kotteas E, Marra A, Prat A, Fusco N, Criscitiello C, Burstein HJ, Curigliano G. The evolving landscape of metastatic HER2-positive, hormone receptor-positive Breast Cancer. Cancer Treat Rev 2024; 128:102761. [PMID: 38772169 DOI: 10.1016/j.ctrv.2024.102761] [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: 03/02/2024] [Revised: 05/05/2024] [Accepted: 05/15/2024] [Indexed: 05/23/2024]
Abstract
Therapeutic agents targeting Human Epidermal Growth Factor Receptor 2 (HER2) demonstrated to positively impact the prognosis of HER2-positive breast cancer. HER2-positive breast cancer can present either as hormone receptor-negative or positive, defining Triple-positive breast cancer (TPBC). TPBC demonstrate unique gene expression profiles, showing reduced HER2-driven gene expression, as recapitulated by a higher proportion of Luminal-type intrinsic subtypes. The different molecular landscape of TPBC dictates distinctive clinical features, including reduced chemotherapy sensitivity, different patterns of recurrence, and better overall prognosis. Cross-talk between HER2 and hormone receptor signaling seems to be critical to determine resistance to HER2-directed agents. Accordingly, superior outcomes have been achieved with the use of endocrine therapy, representing the first subtype-specific pharmacological intervention unique to this subgroup. Additional targeted agents capable to tackle resistance mechanisms to anti-HER2, hormone agents, or both might further improve the efficacy of treatments, such as PI3K/AKT/mTOR inhibitors, particularly in a biomarker-enriched setting, and CDK4/6-inhibitors, with preliminary data suggesting a role of PAM50 subtyping to predict higher benefits in luminal tumors. Finally, the distinct biology of triple-positive tumors may yield the rationale for considering combinations within antibody-drug conjugate regimens. Accordingly, in this review, we summarized the current evidence and rationale for considering TPBC as a different entity, in which distinct therapeutical approaches leveraging on the different biological profile of TPBC may result in superior anticancer regimens and improved patient-centric outcomes.
Collapse
Affiliation(s)
- Luca Boscolo Bielo
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Dario Trapani
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Eleonora Nicolò
- Department of Medicine, Division of Hematology-Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Carmine Valenza
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Lorenzo Guidi
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Carmen Belli
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy
| | - Elias Kotteas
- Oncology Unit, Sotiria General Hospital, 3rd Dept of Internal Medicine, Athens School of Medicine, Greece
| | - Antonio Marra
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy
| | - Aleix Prat
- Department of Medical Oncology and Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Faculty of Medicine and Health Sciences, University of Barcelona, Spain
| | - Nicola Fusco
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy; Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Carmen Criscitiello
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Harold J Burstein
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy.
| |
Collapse
|
3
|
Wu JY, Shyu YK, Lee YK, Wang YC, Chiang CJ, You SL, Liao LJ, Hsu WL, Chen YC. Secular Increasing Trends in Female Thyroid Cancer Incidence in Taiwan. Life (Basel) 2024; 14:809. [PMID: 39063564 PMCID: PMC11278399 DOI: 10.3390/life14070809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 06/22/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Thyroid cancer incidence has increased globally in recent decades, especially in females, although its trends in Taiwan have not been studied extensively. This study aimed to investigate changes in female incidence and possible causes of thyroid cancer in Taiwan. METHODS Using the Taiwan Cancer Registry (TCR) Database, age-standardized incidence rates, age-specific incidence rates and birth cohorts were calculated. Correlation between female thyroid cancer incidence and cohort fertility rates were examined. RESULTS Thyroid cancer incidence increased in Taiwanese female, with age-adjusted rates per 100,000 people increasing from 7.37 during 1995-1999 to 20.53 during 2015-2019; the annual percentage change (APC) was 5.9% (95% CI, 5.3-6.5). Age-specific incidence rates increased with age, with peak rates occurring at younger ages. The APCs in the 50-54 age group were the highest (6.8%, 95% CI, 6.1-7.5). Incidence rates also increased with later birth cohorts. We observed a significant negative correlation between thyroid cancer incidence and fertility rates in the same birth cohort. CONCLUSIONS We hypothesize that overdiagnosis may be a main reason for the rapidly increasing thyroid cancer incidence in Taiwanese females. Notably, we observed a strong negative correlation between fertility and thyroid cancer incidence. However, our study is limited by the absence of individual-level cancer data in the TCR database. These associations with fertility will be an important subject for future thyroid cancer research.
Collapse
Affiliation(s)
- Jiun-Yan Wu
- School of Medicine, College of Medicine, Fu-Jen Catholic University, New Taipei City 242, Taiwan; (J.-Y.W.); (S.-L.Y.)
| | - Yuh-Kae Shyu
- Department of Nursing, College of Medicine, Fu-Jen Catholic University, New Taipei City 242, Taiwan;
| | - Yu-Kwang Lee
- Division of General Surgery, Department of Surgery, National Taiwan University Hospital, Taipei 100, Taiwan;
| | - Yu-Chiao Wang
- Master Program of Big Data in Medical Healthcare Industry, College of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan;
| | - Chun-Ju Chiang
- Graduate Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei 100, Taiwan;
| | - San-Lin You
- School of Medicine, College of Medicine, Fu-Jen Catholic University, New Taipei City 242, Taiwan; (J.-Y.W.); (S.-L.Y.)
- Data Science Center, College of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan
| | - Li-Jen Liao
- Otolaryngology Head and Neck Surgery, Far Eastern Memorial Hospital, Taipei 100, Taiwan;
- Head and Neck Cancer Surveillance and Research Group, Far Eastern Memorial Hospital, New Taipei City 242, Taiwan
- Department of Electrical Engineering, Yuan Ze University, Taoyuan 320, Taiwan
| | - Wan-Lun Hsu
- Master Program of Big Data in Medical Healthcare Industry, College of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan;
- Data Science Center, College of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan
| | - Yong-Chen Chen
- School of Medicine, College of Medicine, Fu-Jen Catholic University, New Taipei City 242, Taiwan; (J.-Y.W.); (S.-L.Y.)
- Data Science Center, College of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan
| |
Collapse
|
4
|
Simińska D, Kojder K, Jeżewski D, Tarnowski M, Tomasiak P, Piotrowska K, Kolasa A, Patrycja K, Chlubek D, Baranowska-Bosiacka I. Estrogen α and β Receptor Expression in the Various Regions of Resected Glioblastoma Multiforme Tumors and in an In Vitro Model. Int J Mol Sci 2024; 25:4130. [PMID: 38612938 PMCID: PMC11012502 DOI: 10.3390/ijms25074130] [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: 02/27/2024] [Revised: 03/29/2024] [Accepted: 04/05/2024] [Indexed: 04/14/2024] Open
Abstract
Glioblastoma multiforme (GBM) is a malignant tumor with a higher prevalence in men and a higher survival rate in transmenopausal women. It exhibits distinct areas influenced by changing environmental conditions. This study examines how these areas differ in the levels of estrogen receptors (ERs) which play an important role in the development and progression of many cancers, and whose expression levels are often correlated with patient survival. This study utilized two research models: an in vitro model employing the U87 cell line and a second model involving tumors resected from patients (including tumor core, enhancing tumor region, and peritumoral area). ER expression was assessed at both gene and protein levels, with the results validated using confocal microscopy and immunohistochemistry. Under hypoxic conditions, the U87 line displayed a decrease in ERβ mRNA expression and an increase in ERα mRNA expression. In patient samples, ERβ mRNA expression was lower in the tumor core compared to the enhancing tumor region (only in males when the study group was divided by sex). In addition, ERβ protein expression was lower in the tumor core than in the peritumoral area (only in women when the study group was divided by sex). Immunohistochemical analysis indicated the highest ERβ protein expression in the enhancing tumor area, followed by the peritumoral area, and the lowest in the tumor core. The findings suggest that ER expression may significantly influence the development of GBM, exhibiting variability under the influence of conditions present in different tumor areas.
Collapse
Affiliation(s)
- Donata Simińska
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (D.S.); (K.P.); (I.B.-B.)
| | - Klaudyna Kojder
- Department of Anaesthesiology and Intensive Care, Pomeranian Medical University in Szczecin, Unii Lubelskiej 1, 71-252 Szczecin, Poland;
| | - Dariusz Jeżewski
- Department of Neurosurgery and Pediatric Neurosurgery, Pomeranian Medical University in Szczecin, Unii Lubelskiej 1, 71-252 Szczecin, Poland;
- Department of Applied Neurocognitivistics, Pomeranian Medical University in Szczecin, Unii Lubelskiej 1, 71-252 Szczecin, Poland
| | - Maciej Tarnowski
- Department of Physiology in Health Sciences, Pomeranian Medical University in Szczecin, Żołnierska 54, 70-210 Szczecin, Poland;
| | - Patrycja Tomasiak
- Institute of Physical Culture Sciences, University of Szczecin, 70-453 Szczecin, Poland;
| | - Katarzyna Piotrowska
- Department of Physiology, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland;
| | - Agnieszka Kolasa
- Department of Histology and Embryology, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland;
| | - Kapczuk Patrycja
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (D.S.); (K.P.); (I.B.-B.)
| | - Dariusz Chlubek
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (D.S.); (K.P.); (I.B.-B.)
| | - Irena Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (D.S.); (K.P.); (I.B.-B.)
| |
Collapse
|
5
|
Li H, Seada H, Madnick S, Zhao H, Chen Z, Li F, Zhu F, Hall S, Boekelheide K. Machine learning-assisted high-content imaging analysis of 3D MCF7 microtissues for estrogenic effect prediction. Sci Rep 2024; 14:2999. [PMID: 38316851 PMCID: PMC10844358 DOI: 10.1038/s41598-024-53323-6] [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: 09/11/2023] [Accepted: 01/30/2024] [Indexed: 02/07/2024] Open
Abstract
Endocrine-disrupting chemicals (EDCs) pose a significant threat to human well-being and the ecosystem. However, in managing the many thousands of uncharacterized chemical entities, the high-throughput screening of EDCs using relevant biological endpoints remains challenging. Three-dimensional (3D) culture technology enables the development of more physiologically relevant systems in more realistic biochemical microenvironments. The high-content and quantitative imaging techniques enable quantifying endpoints associated with cell morphology, cell-cell interaction, and microtissue organization. In the present study, 3D microtissues formed by MCF-7 breast cancer cells were exposed to the model EDCs estradiol (E2) and propyl pyrazole triol (PPT). A 3D imaging and image analysis pipeline was established to extract quantitative image features from estrogen-exposed microtissues. Moreover, a machine-learning classification model was built using estrogenic-associated differential imaging features. Based on 140 common differential image features found between the E2 and PPT group, the classification model predicted E2 and PPT exposure with AUC-ROC at 0.9528 and 0.9513, respectively. Deep learning-assisted analysis software was developed to characterize microtissue gland lumen formation. The fully automated tool can accurately characterize the number of identified lumens and the total luminal volume of each microtissue. Overall, the current study established an integrated approach by combining non-supervised image feature profiling and supervised luminal volume characterization, which reflected the complexity of functional ER signaling and highlighted a promising conceptual framework for estrogenic EDC risk assessment.
Collapse
Affiliation(s)
- Hui Li
- College of Pharmaceutical Sciences, Center for Drug Safety Evaluation and Research of Zhejiang University, Zhejiang University, 866 Yuhangtang Rd, Hangzhou, 310058, China.
- Department of Pathology and Laboratory Medicine, Brown University, 70 Ship Street, Providence, RI, 02903, USA.
| | - Haitham Seada
- Department of Pathology and Laboratory Medicine, Brown University, 70 Ship Street, Providence, RI, 02903, USA
| | - Samantha Madnick
- Department of Pathology and Laboratory Medicine, Brown University, 70 Ship Street, Providence, RI, 02903, USA
| | - He Zhao
- College of Pharmaceutical Sciences, Center for Drug Safety Evaluation and Research of Zhejiang University, Zhejiang University, 866 Yuhangtang Rd, Hangzhou, 310058, China
| | - Zhaozeng Chen
- College of Pharmaceutical Sciences, Center for Drug Safety Evaluation and Research of Zhejiang University, Zhejiang University, 866 Yuhangtang Rd, Hangzhou, 310058, China
| | - Fengcheng Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Feng Zhu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Susan Hall
- Department of Pathology and Laboratory Medicine, Brown University, 70 Ship Street, Providence, RI, 02903, USA
| | - Kim Boekelheide
- Department of Pathology and Laboratory Medicine, Brown University, 70 Ship Street, Providence, RI, 02903, USA.
| |
Collapse
|
6
|
Xu Z, Wang P, Wang Z, Cui H, Gao T, Wang Z, Liu Y. ER-β accelerates the process of primary osteoporosis by promoting VEGFA-mediated apoptosis of osteoblasts. Genomics 2023; 115:110743. [PMID: 37967683 DOI: 10.1016/j.ygeno.2023.110743] [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: 07/13/2023] [Revised: 09/27/2023] [Accepted: 11/12/2023] [Indexed: 11/17/2023]
Abstract
Primary osteoporosis (POP) is a widespread and severe disorder of bone metabolism characterized by reduced bone mass and destruction of bone structure, frequently inducing fracture risk and imposing a heavy economic burden on public life. The development of POP partially revolves around the estrogen receptor β (ER-β), one of the major mediator receptors of estrogen that influences apoptosis in a range of cells. We performed KEGG and GO analysis by mining the transcriptomic dataset of POP samples showing significant enrichment of differentially expressed genes (DEGs) in multiple apoptosis-related pathways. The results of the Spearman correlation analysis and Protein-Protein Interaction (PPI) Networks screening of hub genes indicated that vascular endothelial growth factor A (VEGFA) may be a key target of ER-β in controlling osteoblast apoptosis. Further, we carried out high-throughput sequencing of ESR2-silenced MC3T3-E1 cells and noticed a substantial suppression in VEGFA expression and all apoptosis-related pathways. In addition, we determined the cell cycle and apoptosis by constructing a VEGFA-silenced cell model utilizing flow cytometry (FCM), and the results showed that ER-β could regulate the osteoblast cycle and thus promote osteoblast apoptosis by promoting VEGFA expression. And Western blot results showed that apoptosis was most likely realized through the regulation of downstream apoptosis markers c-JUN (c-Jun N-terminal kinase, JNK) and GADD45G (Growth Arrest and DNA Damage-Inducible Protein 45 gamma). The effects of ESR2 and VEGFA on the proliferation of osteoblasts were lastly assessed using the cell counting kit- 8 (CCK-8) assay. In conclusion, this study identifies that the roles of ER-β in the regulation of osteoblast apoptosis are closely related to VEGFA and provides a new target for POP treatment.
Collapse
Affiliation(s)
- Zhujie Xu
- Department of Orthopedics, The Affiliated Wuxi People's Hospital to Nanjing Medical University, Wuxi, Jiangsu 214023, PR China; Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu 214000, PR China.
| | - Peng Wang
- The First Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu 211166, PR China
| | - Zhenyu Wang
- The First Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu 211166, PR China
| | - Hao Cui
- The First Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu 211166, PR China
| | - Tianshu Gao
- The First Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu 211166, PR China
| | - Zhenting Wang
- The First Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu 211166, PR China
| | - Yi Liu
- Department of Orthopedics, The Affiliated Wuxi People's Hospital to Nanjing Medical University, Wuxi, Jiangsu 214023, PR China.
| |
Collapse
|
7
|
Saber S, Al-Qawasmeh RA, Abu-Qatouseh L, Shtaiwi A, Khanfar MA, Al-Soud YA. Novel hybrid motifs of 4-nitroimidazole-piperazinyl tagged 1,2,3-triazoles: Synthesis, crystal structure, anticancer evaluations, and molecular docking study. Heliyon 2023; 9:e19327. [PMID: 37681149 PMCID: PMC10480608 DOI: 10.1016/j.heliyon.2023.e19327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 08/08/2023] [Accepted: 08/18/2023] [Indexed: 09/09/2023] Open
Abstract
4-((4-(1-benzyl-2-methyl-4-nitro-1H-imidazole-5-yl)piperazine-1-yl)methyl)-1-substituted-1H-1,2,3-triazole motifs are designed and synthesized via click chemistry. The reaction of 1-(N1-benzyl- 2-methyl-4-nitro-1H-imidazole- 5-yl)-4-(prop-2-yn-1-yl) piperazine 5 as new scaffold with diverse primary azides to selectively produce 1,4-disubstituted-1,2,3-triazoles 9a-k, 10a-c and 11a-q. Physicochemical methods: when 1H NMR, 13C NMR, and HRMS are utilized to fully characterize all synthesized compounds. X-ray structural determination and analysis for compound 9a is also performed. The newly designed chromophores are assessed for their anti-proliferative potency against three selected human cancer cell lines (MCF-7, HepG2, and PC3), and one normal cell line (Dermal/Fibroblast). Compounds 9g and 9k have shown potent activities against the MCF-7 cell line with IC50 values of (2.00 ± 0.03 μM) and (5.00 ± 0.01 μM) respectively. ADMET studies and Molecular docking investigations are performed on the most active hybrid nitroimidazole derivatives 9g and 9k with 4-hydroxytamoxifen (4-OHT) at the human estrogen receptor alpha (hER) during binding active sites to study the ligand-protein interactions and free binding energies at atomic levels. The triazole ring in the 9g derivative forms a hydrogen bond with Asp58 with distance 3.2 Å. And it is found that polar contact with His231 amino acid residue. In silico assessment of the compounds showed very good pharmacokinetic properties based on their physicochemical values, also the ADMET criteria of the most active hybrid systems are within the acceptable range.
Collapse
Affiliation(s)
- SadeekahO.W. Saber
- Department of chemistry, School of Science, The University of Jordan, 11942, Amman, Jordan
- Faculty of Pharmacy, Jerash University, Amman-Irbid international highway, Jerash, 26150, Jordan
| | - Raed A. Al-Qawasmeh
- Department of chemistry, School of Science, The University of Jordan, 11942, Amman, Jordan
- Pure and Applied Chemistry Group, Department of Chemistry, College of Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | | | - Amneh Shtaiwi
- Faculty of Pharmacy, Middle East University, Queen Alia Airport Street, 11610, Amman, Jordan
| | - Monther A. Khanfar
- Department of chemistry, School of Science, The University of Jordan, 11942, Amman, Jordan
- Pure and Applied Chemistry Group, Department of Chemistry, College of Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Yaseen A. Al-Soud
- Chemistry Department, Faculty of Science, University of Al al-Bayt, Al-Mafraq, Jordan
| |
Collapse
|
8
|
Pegram M, Jackisch C, Johnston SRD. Estrogen/HER2 receptor crosstalk in breast cancer: combination therapies to improve outcomes for patients with hormone receptor-positive/HER2-positive breast cancer. NPJ Breast Cancer 2023; 9:45. [PMID: 37258523 DOI: 10.1038/s41523-023-00533-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/04/2023] [Indexed: 06/02/2023] Open
Abstract
The human epidermal growth factor receptor 2 (HER2) is overexpressed in 13-22% of breast cancers (BC). Approximately 60-70% of HER2+ BC co-express hormone receptors (HRs). HR/HER2 co-expression modulates response to both anti-HER2-directed and endocrine therapy due to "crosstalk" between the estrogen receptor (ER) and HER2 pathways. Combined HER2/ER blockade may be an effective treatment strategy for patients with HR+/HER2+ BC in the appropriate clinical setting(s). In this review, we provide an overview of crosstalk between the ER and HER2 pathways, summarize data from recently published and ongoing clinical trials, and discuss clinical implications for targeted treatment of HR+/HER2+ BC.
Collapse
Affiliation(s)
- Mark Pegram
- Stanford Cancer Institute, Stanford, CA, USA.
| | - Christian Jackisch
- Obstetrics and Gynaecology and Breast Cancer Center, Klinikum Offenbach GmbH, Offenbach, Germany
| | | |
Collapse
|
9
|
Ahmad W, Panicker NG, Akhlaq S, Gull B, Baby J, Khader TA, Rizvi TA, Mustafa F. Global Down-regulation of Gene Expression Induced by Mouse Mammary Tumor Virus (MMTV) in Normal Mammary Epithelial Cells. Viruses 2023; 15:v15051110. [PMID: 37243196 DOI: 10.3390/v15051110] [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: 03/28/2023] [Revised: 04/24/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
Mouse mammary tumor virus (MMTV) is a betaretrovirus that causes breast cancer in mice. The mouse mammary epithelial cells are the most permissive cells for MMTV, expressing the highest levels of virus upon infection and being the ones later transformed by the virus due to repeated rounds of infection/superinfection and integration, leading eventually to mammary tumors. The aim of this study was to identify genes and molecular pathways dysregulated by MMTV expression in mammary epithelial cells. Towards this end, mRNAseq was performed on normal mouse mammary epithelial cells stably expressing MMTV, and expression of host genes was analyzed compared with cells in its absence. The identified differentially expressed genes (DEGs) were grouped on the basis of gene ontology and relevant molecular pathways. Bioinformatics analysis identified 12 hub genes, of which 4 were up-regulated (Angp2, Ccl2, Icam, and Myc) and 8 were down-regulated (Acta2, Cd34, Col1a1, Col1a2, Cxcl12, Eln, Igf1, and Itgam) upon MMTV expression. Further screening of these DEGs showed their involvement in many diseases, especially in breast cancer progression when compared with available data. Gene Set Enrichment Analysis (GSEA) identified 31 molecular pathways dysregulated upon MMTV expression, amongst which the PI3-AKT-mTOR was observed to be the central pathway down-regulated by MMTV. Many of the DEGs and 6 of the 12 hub genes identified in this study showed expression profile similar to that observed in the PyMT mouse model of breast cancer, especially during tumor progression. Interestingly, a global down-regulation of gene expression was observed, where nearly 74% of the DEGs in HC11 cells were repressed by MMTV expression, an observation similar to what was observed in the PyMT mouse model during tumor progression, from hyperplasia to adenoma to early and late carcinomas. Comparison of our results with the Wnt1 mouse model revealed further insights into how MMTV expression could lead to activation of the Wnt1 pathway independent of insertional mutagenesis. Thus, the key pathways, DEGs, and hub genes identified in this study can provide important clues to elucidate the molecular mechanisms involved in MMTV replication, escape from cellular anti-viral response, and potential to cause cell transformation. These data also validate the use of the MMTV-infected HC11 cells as an important model to study early transcriptional changes that could lead to mammary cell transformation.
Collapse
Affiliation(s)
- Waqar Ahmad
- Department of Biochemistry & Molecular Biology, College of Medicine and Health Sciences (CMHS), United Arab Emirates (UAE) University, Al Ain 15551, United Arab Emirates
| | - Neena G Panicker
- Department of Biochemistry & Molecular Biology, College of Medicine and Health Sciences (CMHS), United Arab Emirates (UAE) University, Al Ain 15551, United Arab Emirates
| | - Shaima Akhlaq
- Department of Biochemistry & Molecular Biology, College of Medicine and Health Sciences (CMHS), United Arab Emirates (UAE) University, Al Ain 15551, United Arab Emirates
| | - Bushra Gull
- Department of Biochemistry & Molecular Biology, College of Medicine and Health Sciences (CMHS), United Arab Emirates (UAE) University, Al Ain 15551, United Arab Emirates
| | - Jasmin Baby
- Department of Biochemistry & Molecular Biology, College of Medicine and Health Sciences (CMHS), United Arab Emirates (UAE) University, Al Ain 15551, United Arab Emirates
| | - Thanumol A Khader
- Department of Biochemistry & Molecular Biology, College of Medicine and Health Sciences (CMHS), United Arab Emirates (UAE) University, Al Ain 15551, United Arab Emirates
| | - Tahir A Rizvi
- Department of Microbiology and Immunology, College of Medicine and Health Sciences (CMHS), UAE University, Al Ain 15551, United Arab Emirates
- Zayed Center for Health Sciences (ZCHS), UAE University, Al Ain 15551, United Arab Emirates
- ASPIRE Research Institute in Precision Medicine, Abu Dhabi, UAE University, Al Ain 15551, United Arab Emirates
| | - Farah Mustafa
- Department of Biochemistry & Molecular Biology, College of Medicine and Health Sciences (CMHS), United Arab Emirates (UAE) University, Al Ain 15551, United Arab Emirates
- Zayed Center for Health Sciences (ZCHS), UAE University, Al Ain 15551, United Arab Emirates
| |
Collapse
|
10
|
Anticancer or carcinogenic? The role of estrogen receptor β in breast cancer progression. Pharmacol Ther 2023; 242:108350. [PMID: 36690079 DOI: 10.1016/j.pharmthera.2023.108350] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 01/06/2023] [Accepted: 01/17/2023] [Indexed: 01/22/2023]
Abstract
Estrogen receptor β (ERβ) is closely related to breast cancer (BC) progression. Traditional concepts regard ERβ as a tumor suppressor. As studies show the carcinogenic effect of ERβ, some people have come to a new conclusion that ERβ serves as a tumor suppressor in estrogen receptor α (ERα)-positive breast cancer, while it is a carcinogen in ERα-negative breast cancer. However, we re-examine the role of ERβ and find this conclusion to be misleading based on the last decade's research. A large number of studies have shown that ERβ plays an anticancer role in both ERα-positive and ERα-negative breast cancers, and its carcinogenicity does not depend solely on the presence of ERα. Herein, we review the anticancer and oncogenic effects of ERβ on breast cancer progression in the past ten years, discuss the mechanism respectively, analyze the main reasons for the inconsistency and update ERβ selective ligand library. We believe a detailed and continuously updated review will help correct the one-sided understanding of ERβ, promoting ERβ-targeted breast cancer therapy.
Collapse
|
11
|
Chen M, Zheng W, Fang L. Identifying liver metastasis-related hub genes in breast cancer and characterizing SPARCL1 as a potential prognostic biomarker. PeerJ 2023; 11:e15311. [PMID: 37180578 PMCID: PMC10174054 DOI: 10.7717/peerj.15311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 04/06/2023] [Indexed: 05/16/2023] Open
Abstract
Background The liver is the third most common metastatic site for advanced breast cancer (BC), and liver metastases predict poor prognoses. However, the characteristic biomarkers of BC liver metastases and the biological role of secreted protein acidic and rich in cysteine-like 1 (SPARCL1) in BC remain unclear. The present study aimed to identify potential biomarkers for liver metastasis of BC and to investigate the effect of SPARCL1 on BC. Methods The publicly available GSE124648 dataset was used to identify differentially expressed genes (DEGs) between BC and liver metastases. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were conducted to annotate these DEGs and understand the biological functions in which they are involved. A protein-protein interaction (PPI) network was constructed to identify metastasis-related hub genes and further validated in a second independent dataset (GSE58708). Clinicopathological correlation of hub gene expression in patients with BC was determined. Gene set enrichment analysis (GSEA) was performed to explore DEG-related signaling pathways. SPARCL1 expression in BC tissues and cell lines was verified by RT-qPCR. Further in vitro experiments were performed to investigate the biological functions of SPARCL1 in BC cells. Results We identified 332 liver metastasis-related DEGs from GSE124648 and 30 hub genes, including SPARCL1, from the PPI network. GO and KEGG enrichment analyses of liver-metastasis-related DEGs revealed several enriched terms associated with the extracellular matrix and pathways in cancer. Clinicopathological correlation analysis of SPARCL1 revealed that its expression in BC was associated with age, TNM stage, estrogen receptor status, progesterone receptor status, histological type, molecular type, and living status of patients. GSEA results suggested that low SPARCL1 expression in BC was related to the cell cycle, DNA replication, oxidative phosphorylation, and homologous recombination. Lower expression levels of SPARCL1 were detected in BC tissues compared to adjacent tissues. The in vitro experiments showed that SPARCL1 knockdown significantly increased the proliferation and migration of BC cells, whereas the proliferation and migration were suppressed after elevating the expression of SPARCL1. Conclusion We identified SPARCL1 as a tumor suppressor in BC, which shows potential as a target for BC and liver metastasis therapy and diagnosis.
Collapse
Affiliation(s)
- Mingkuan Chen
- Tongji University School of Medicine, Department of Thyroid and Breast Division of General Surgery Shanghai Tenth People’s Hospital, Shanghai, Jing’an District, China
| | - Wenfang Zheng
- Tongji University School of Medicine, Department of Thyroid and Breast Division of General Surgery Shanghai Tenth People’s Hospital, Shanghai, Jing’an District, China
| | - Lin Fang
- Tongji University School of Medicine, Department of Thyroid and Breast Division of General Surgery Shanghai Tenth People’s Hospital, Shanghai, Jing’an District, China
| |
Collapse
|
12
|
Hu Q, Luo K, Liu P, Mei Y. To discuss the mechanism of colchicine in the treatment of acute cerebral infarction based on network pharmacology. Medicine (Baltimore) 2022; 101:e30720. [PMID: 36197265 PMCID: PMC9509177 DOI: 10.1097/md.0000000000030720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
To explore the mechanism of action of colchicine in the treatment of acute cerebral infarction (ACI) based on network pharmacology. The Swiss Target Prediction Database and CTD database were used to predict the target information of colchicine. ACI-related targets were retrieved using the GeneCards database, and the target protein interaction network (PPI) and active ingredient-target network were obtained by combining Cytoscape 3.7.1 software and R language. Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis and gene function analysis (GO) enrichment analysis were performed using R language to preliminarily explore the multiple pharmacological mechanisms of action of colchicine. There were 200 targets identified by network parameter analysis; 958 ACI targets were identified. Overlapping comparisons allowed the extraction of 143 overlapping targets, and the top 30 targets were screened according to the topological isomerization parameters. Component-target networks were constructed. A PPI of overlapping targets was established to identify key targets. In addition, Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis and GO functional enrichment analysis were performed to explore the multiple mechanisms of action of colchicine in the treatment of ACI. Colchicine treatment of ACI is characterized by multi-component, multi-target and multi-pathway, and can exert complex network regulation through the interaction between different targets, providing a new idea and new basis for further exploration of the mechanism of action of colchicine in the treatment of ACI.
Collapse
Affiliation(s)
- Qiaoxia Hu
- Department of Geriatrics, the Affiliated Hospital of Medical School of Ningbo University, Institute of Geriatrics of Ningbo University, Ningbo, China
- *Correspondence: Qiaoxia Hu, Department of Geriatrics, the Affiliated Hospital of Medical School of Ningbo University, Institute of Geriatrics of Ningbo University, 247 Renmin Road, Ningbo 315020, China (e-mail: )
| | - Kena Luo
- Department of Geriatrics, the Affiliated Hospital of Medical School of Ningbo University, Institute of Geriatrics of Ningbo University, Ningbo, China
| | - Puheng Liu
- Department of Geriatrics, the Affiliated Hospital of Medical School of Ningbo University, Institute of Geriatrics of Ningbo University, Ningbo, China
| | - Yifei Mei
- Department of Geriatrics, the Affiliated Hospital of Medical School of Ningbo University, Institute of Geriatrics of Ningbo University, Ningbo, China
| |
Collapse
|
13
|
Zhu X, Xue C, Kang X, Jia X, Wang L, Younis MH, Liu D, Huo N, Han Y, Chen Z, Fu J, Zhou C, Yao X, Du Y, Cai W, Kang L, Lyu Z. DNMT3B-mediated FAM111B methylation promotes papillary thyroid tumor glycolysis, growth and metastasis. Int J Biol Sci 2022; 18:4372-4387. [PMID: 35864964 PMCID: PMC9295055 DOI: 10.7150/ijbs.72397] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 05/13/2022] [Indexed: 11/15/2022] Open
Abstract
Over the past decades, the incidence of thyroid cancer (TC) rapidly increased all over the world, with the papillary thyroid cancer (PTC) accounting for the vast majority of TC cases. It is crucial to investigate novel diagnostic and therapeutic targets for PTC and explore more detailed molecular mechanisms in the carcinogenesis and progression of PTC. Based on the TCGA and GEO databases, FAM111B is downregulated in PTC tissues and predicts better prognosis in PTC patients. FAM111B suppresses the growth, migration, invasion and glycolysis of PTC both in vitro and in vivo. Furthermore, estrogen inhibits FAM111B expression by DNMT3B methylation via enhancing the recruitment of DNMT3B to FAM111B promoter. DNMT3B-mediated FAM111B methylation accelerates the growth, migration, invasion and glycolysis of PTC cells. In clinical TC patient specimens, the expression of FAM111B is inversely correlated with the expressions of DNMT3B and the glycolytic gene PGK1. Besides, the expression of FAM111B is inversely correlated while DNMT3B is positively correlated with glucose uptake in PTC patients. Our work established E2/DNMT3B/FAM111B as a crucial axis in regulating the growth and progression of PTC. Suppression of DNMT3B or promotion of FAM111B will be potential promising strategies in the estrogen induced PTC.
Collapse
Affiliation(s)
- Xiang Zhu
- Department of Endocrinology, the First Medical Center of PLA General Hospital, Beijing, China.,Department of Genetic Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Chunyuan Xue
- Department of Genetic Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Xiaofeng Kang
- Department of Genetic Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Xiaomeng Jia
- Department of Endocrinology, the First Medical Center of PLA General Hospital, Beijing, China
| | - Lin Wang
- Department of Endocrinology, the First Medical Center of PLA General Hospital, Beijing, China
| | - Muhsin H Younis
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI, USA
| | - Donghui Liu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Nan Huo
- Department of Genetic Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Yuchen Han
- Department of Genetic Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Zhao Chen
- Department of Nuclear Medicine, Peking University First Hospital, Beijing, China
| | - Jing Fu
- Department of Pathology, Beijing Haidian Hospital, Beijing, China
| | - Chunyu Zhou
- Department of Pathology, Beijing Haidian Hospital, Beijing, China
| | - Xiaoxiang Yao
- Department of Pathology, Beijing Haidian Hospital, Beijing, China
| | - Yimeng Du
- Department of Genetic Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI, USA
| | - Lei Kang
- Department of Nuclear Medicine, Peking University First Hospital, Beijing, China
| | - Zhaohui Lyu
- Department of Endocrinology, the First Medical Center of PLA General Hospital, Beijing, China
| |
Collapse
|
14
|
Cai Q, Yang HS, Li YC, Zhu J. Dissecting the Roles of PDCD4 in Breast Cancer. Front Oncol 2022; 12:855807. [PMID: 35795053 PMCID: PMC9251513 DOI: 10.3389/fonc.2022.855807] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 05/12/2022] [Indexed: 11/29/2022] Open
Abstract
The human programmed cell death 4 (PDCD4) gene was mapped at chromosome 10q24 and encodes the PDCD4 protein comprised of 469 amino acids. PDCD4 inhibits protein translation PDCD4 inhibits protein translation to suppress tumor progression, and its expression is frequently decreased in breast cancer. PDCD4 blocks translation initiation complex by binding eIF4A via MA-3 domains or by directly binding 5’ mRNA internal ribosome entry sites with an RNA binding domain to suppress breast cancer progression and proliferation. Numerous regulators and biological processes including non-coding RNAs, proteasomes, estrogen, natural compounds and inflammation control PDCD4 expression in breast cancer. Loss of PDCD4 expression is also responsible for drug resistance in breast cancer. HER2 activation downregulates PDCD4 expression by activating MAPK, AKT, and miR-21 in aromatase inhibitor-resistant breast cancer cells. Moreover, modulating the microRNA/PDCD4 axis maybe an effective strategy for overcoming chemoresistance in breast cancer. Down-regulation of PDCD4 is significantly associated with short overall survival of patients, which suggests that PDCD4 may be an independent prognostic marker for breast cancer.
Collapse
Affiliation(s)
- Qian Cai
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovasular Proteomics of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
| | - Hsin-Sheng Yang
- Department of Toxicology and Cancer Biology, Collage of Medicine, University of Kentucky, Lexington, KY, United States
| | - Yi-Chen Li
- Department of Breast Surgery, General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Jiang Zhu
- Department of Breast Surgery, General Surgery, Qilu Hospital of Shandong University, Jinan, China
- *Correspondence: Jiang Zhu,
| |
Collapse
|
15
|
Ge Y, Li F, He Y, Cao Y, Guo W, Hu G, Liu J, Fu S. L-arginine stimulates the proliferation of mouse mammary epithelial cells and the development of mammary gland in pubertal mice by activating the GPRC6A/PI3K/AKT/mTOR signalling pathway. J Anim Physiol Anim Nutr (Berl) 2022; 106:1383-1395. [PMID: 35616019 DOI: 10.1111/jpn.13730] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/25/2022] [Accepted: 04/20/2022] [Indexed: 11/30/2022]
Abstract
Amino acids have been shown to affect the development of mammary gland (MG). However, it is unclear whether L-arginine promotes the development of pubertal MG. Therefore, our study aims to explore the effect of L-arginine on the development of MG in pubertal mice. To investigate its internal mechanism of action, we will use mouse mammary epithelial cells (mMECs) line. Whole-mount staining showed that L-arginine can promote the extension of MG duct. In vitro, 0.4 mM L-arginine could activate the G protein-coupled receptor family C, group 6, subtype A (GPRC6A)/phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) signalling pathway and increase the phosphorylation of eukaryotic initiation factor 4E binding protein 1 (4EBP1) to promote the synthesis of cell cycle regulatory protein D1 (Cyclin D1), leading to the dissociation of the retinoblastoma tumour suppressor protein (Rb)-E2F1 transcription factor (E2F1) complex in mMECs and releasing E2F1 to promote cell proliferation. Furthermore, GPRC6A was knocked down or inhibition of the PI3K/AKT/mTOR signalling pathway with corresponding inhibitors completely abolished the arginine-induced promotion of mMECs proliferation. In vivo, it was further confirmed that 0.1% L-arginine can activate the PI3K/AKT/mTOR signalling pathway in the MG of pubertal mice. These results were able to indicate that L-arginine stimulates the development of MG in pubertal mice through the GPRC6A/PI3K/AKT/mTOR signalling pathway.
Collapse
Affiliation(s)
- Yusong Ge
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Feng Li
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Yuan He
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Yu Cao
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Wenjin Guo
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Guiqiu Hu
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Juxiong Liu
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Shoupeng Fu
- Department of Theoretic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| |
Collapse
|
16
|
Raloxifene Suppresses Tumor Growth and Metastasis in an Orthotopic Model of Castration-Resistant Prostate Cancer. Biomedicines 2022; 10:biomedicines10040853. [PMID: 35453603 PMCID: PMC9033055 DOI: 10.3390/biomedicines10040853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/01/2022] [Accepted: 04/01/2022] [Indexed: 02/01/2023] Open
Abstract
Androgen receptor (AR)-castrate-resistant prostate cancer (CRPC) is an aggressive form of prostate cancer that does not have clinically approved targeted treatment options. To this end, the cytotoxic potential of raloxifene and the synthetic curcumin derivative 2,6-bis (pyridin-4-ylmethylene)-cyclohexanone (RL91) was examined in AR-(PC3 and DU145) cells and AR+ (LnCaP) CRPC cells. The results showed that both raloxifene and RL91 elicited significant cytotoxicity across three cell lines with the lowest EC50 values in PC3 cells. Additionally, the two drugs were synergistically cytotoxic toward the PC3, DU-145 and LNCaP cell lines. To determine the effect of the drug combination in vivo, an orthotopic model of CRPC was used. Male mice were injected with PC3 prostate cancer cells and then treated with vehicle (5 mL/kg), raloxifene (8.5 mg/kg, po), RL91 (8.5 mg/kg, po) or a combination of raloxifene and RL91 for six weeks. Sham animals were subjected to the surgical procedure but were not implanted with PC3 cells. The results showed that raloxifene decreased tumor size and weight as well as metastasis to renal lymph nodes. However, combination treatment reversed the efficacy of raloxifene as tumor volume and metastasis returned to control levels. The results suggest that raloxifene has tumor suppressive and anti-metastatic effects and has potential for further clinical use in AR-CRPC.
Collapse
|
17
|
Li Y, Zheng N, Li Y, Li P, Sun S, Wang S, Song X. Exposure of childbearing-aged female to phthalates through the use of personal care products in China: An assessment of absorption via dermal and its risk characterization. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 807:150980. [PMID: 34662603 DOI: 10.1016/j.scitotenv.2021.150980] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 10/08/2021] [Accepted: 10/10/2021] [Indexed: 06/13/2023]
Abstract
Phthalates (PAEs) are widely used in personal care products (PCPs) and skin care packaging materials. Through national representative sampling, 328 childbearing-aged females in China were investigated by questionnaire, whose contact factors for 30 cosmetic products were collected. According to the daily exposure method and adverse cumulative effects of PAE exposure on female reproduction, we derived the ERα, ERβ binding, and AR anti-androgenic effects. The utilization rates of acne cleanser, acne cream, cleanser (non-acne), and cream (non-acne) in volunteers were 21.90%, 22.22%, 51.63%, and 51.96%, respectively. Examining the data for PAEs in PCPs, the content of DBP (dibutyl phthalate) in them was significantly higher for tubes (0.26 ± 0.05 μg/g) and other packaging (pump type and metal tube) (0.25 ± 0.03 μg/g) than bowl (0.17 ± 0.04 μg/g). The DBP content of acne cream (0.27 ± 0.03 μg/g) was significantly higher than that of non-acne cream (0.17 ± 0.03 μg/g); likewise, there was significantly more DEHP (di (2-ethylhexyl) phthalate) in acne cleanser (0.87 ± 0.15 μg/g) than non-acne cleanser (0.64 ± 0.36 μg/g). Students and office worker were the main consumers of PCPs; however, among all occupation groups, the daily exposure dose of PCPs for workers was highest (mean = 0.0004, 0.0002, 0.0009 μg/kg bw/day for DEP (diethyl phthalate), DBP, and DEHP, respectively). The cumulative indices of PAEs' exposure revealed that the level of ERα and ERβ binding and AR anti-androgenic effects in workers was respectively 0.4935, 0.0186, and 0.2411 μg/kg bw/day. The risk index (HITDI and HIRfDs) of DEP, DBP, and DEHP was lower than their corresponding reference value (hazard index <1), but using PCPs may cause potential health risks. Therefore, we should pay attention to the adverse effects of PAEs on female reproductive functioning, especially the cumulative exposure of females of childbearing age.
Collapse
Affiliation(s)
- Yunyang Li
- Northeast Institute of Geography and Agricultural Ecology, Chinese Academy of Sciences, Changchun, Jilin, China; Key Laboratory of Groundwater Resources and Environment of the Ministry of Education, College of Environment and Resources, Jilin University, China; Graduate University of Chinese Academy of Sciences, Beijing, China
| | - Na Zheng
- Northeast Institute of Geography and Agricultural Ecology, Chinese Academy of Sciences, Changchun, Jilin, China; Key Laboratory of Groundwater Resources and Environment of the Ministry of Education, College of Environment and Resources, Jilin University, China.
| | - Yang Li
- Key Laboratory of Groundwater Resources and Environment of the Ministry of Education, College of Environment and Resources, Jilin University, China
| | - Pengyang Li
- Key Laboratory of Groundwater Resources and Environment of the Ministry of Education, College of Environment and Resources, Jilin University, China
| | - Siyu Sun
- Key Laboratory of Groundwater Resources and Environment of the Ministry of Education, College of Environment and Resources, Jilin University, China
| | - Sujing Wang
- Key Laboratory of Groundwater Resources and Environment of the Ministry of Education, College of Environment and Resources, Jilin University, China
| | - Xue Song
- Key Laboratory of Groundwater Resources and Environment of the Ministry of Education, College of Environment and Resources, Jilin University, China
| |
Collapse
|
18
|
Chen P, Li B, Ou-Yang L. Role of estrogen receptors in health and disease. Front Endocrinol (Lausanne) 2022; 13:839005. [PMID: 36060947 PMCID: PMC9433670 DOI: 10.3389/fendo.2022.839005] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 07/26/2022] [Indexed: 12/14/2022] Open
Abstract
Estrogen receptors (ERs) regulate multiple complex physiological processes in humans. Abnormal ER signaling may result in various disorders, including reproductive system-related disorders (endometriosis, and breast, ovarian, and prostate cancer), bone-related abnormalities, lung cancer, cardiovascular disease, gastrointestinal disease, urogenital tract disease, neurodegenerative disorders, and cutaneous melanoma. ER alpha (ERα), ER beta (ERβ), and novel G-protein-coupled estrogen receptor 1 (GPER1) have been identified as the most prominent ERs. This review provides an overview of ERα, ERβ, and GPER1, as well as their functions in health and disease. Furthermore, the potential clinical applications and challenges are discussed.
Collapse
Affiliation(s)
| | - Bo Li
- *Correspondence: Bo Li, libo‐‐
| | | |
Collapse
|
19
|
Karim K, Giribabu N, Salleh N. Marantodes pumilum (blume) Kuntze (Kacip Fatimah) leaves aqueous extract prevents downregulation of Wnt/β-catenin pathway and upregulation of apoptosis in osteoblasts of estrogen-deficient, diabetes-induced rats. JOURNAL OF ETHNOPHARMACOLOGY 2021; 280:114236. [PMID: 34044074 DOI: 10.1016/j.jep.2021.114236] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 05/02/2021] [Accepted: 05/20/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Marantodes pumilum (Blume) Kuntze has been claimed to be beneficial in protecting the bone against loss in post-menopausal women. In view of increased incidence of diabetes mellitus (DM) in post-menopausal period, M. pumilum ability to overcome the detrimental effect of estrogen-deficiency and DM on the bones were identified. AIM OF THE STUDY To identify the mechanisms underlying protective effect of MPLA on the bone in estrogen-deficient, diabetic condition. METHODS Adult female, estrogen-deficient, diabetic rats (225 ± 10 g) were divided into untreated group and treated with M. pumilum leaf aqueous extract (MPLA) (50 mg/kg/day and 100 mg/kg/day) and estrogen for 28 days (n = 6 per group). Fasting blood glucose (FBG) levels were weekly monitored and at the end of treatment, rats were sacrificed and femur bones were harvested. Bone collagen distribution was observed by Masson's trichome staining. Levels of bone osteoblastogenesis, apoptosis and proliferative markers were evaluated by Realtime PCR, Western blotting, immunofluorescence and immunohistochemistry. RESULTS MPLA treatment was able to ameliorate the increased in FBG levels in estrogen deficient, diabetic rats. In these rats, decreased bone collagen content, expression level of osteoblastogenesis markers (Wnt3a, β-catenin, Frizzled, Dvl and LRP-5) and proliferative markers (PCNA and c-Myc) and increased expression of anti-osteoblastogenesis marker (Gsk-3β) and apoptosis markers (Caspase-3, Caspase-9 and Bax) but not Bcl-2 were ameliorated. Effects of 100 mg/kg/day MPLA were greater than estrogen. CONCLUSION MPLA was able to protect against bone loss, thus making it a promising agent for the treatment of osteoporosis in women with estrogen deficient, diabetic condition.
Collapse
Affiliation(s)
- Kamarulzaman Karim
- Department of Physiology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Nelli Giribabu
- Department of Physiology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia.
| | - Naguib Salleh
- Department of Physiology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia.
| |
Collapse
|
20
|
CTI-2 Inhibits Metastasis and Epithelial-Mesenchymal Transition of Breast Cancer Cells by Modulating MAPK Signaling Pathway. Int J Mol Sci 2021; 22:ijms222212229. [PMID: 34830111 PMCID: PMC8622910 DOI: 10.3390/ijms222212229] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/26/2021] [Accepted: 10/29/2021] [Indexed: 02/07/2023] Open
Abstract
Although some breast cancer patients die due to tumor metastasis rather than from the primary tumor, the molecular mechanism of metastasis remains unclear. Therefore, it is necessary to inhibit breast cancer metastasis during cancer treatment. In this case, after designing and synthesizing CTI-2, we found that CTI-2 treatment significantly reduced breast cancer cell metastasis in vivo and in vitro. Notably, with the treatment of CTI-2 in breast cancer cells, the expression level of E-cadherin increased, while the expression level of N-cadherin and vimentin decreased. In addition, after CTI-2 treatment, those outflow levels for p-ERK, p-p38, and p-JNK diminished, while no significant changes in the expression levels of ERK, JNK, or p38 were observed. Our conclusion suggested that CTI-2 inhibits the epithelial-mesenchymal transition (EMT) of breast carcinoma cells by inhibiting the activation of the mitogen-activated protein kinase (MAPK) signaling pathway, thereby inhibiting the metastasis of breast tumor cells. Therefore, we believe that CTI-2 is another candidate for breast tumor medication.
Collapse
|
21
|
Mendonca P, Alghamdi S, Messeha S, Soliman KFA. Pentagalloyl glucose inhibits TNF-α-activated CXCL1/GRO-α expression and induces apoptosis-related genes in triple-negative breast cancer cells. Sci Rep 2021; 11:5649. [PMID: 33707603 PMCID: PMC7952910 DOI: 10.1038/s41598-021-85090-z] [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: 09/22/2020] [Accepted: 02/24/2021] [Indexed: 12/13/2022] Open
Abstract
In triple-negative breast cancer (TNBC), the tumor microenvironment is associated with increased proliferation, suppressing apoptotic mechanisms, an altered immune response, and drug resistance. The current investigation was designed to examine the natural compound pentagalloyl glucose (PGG) effects on TNF-α activated TNBC cell lines, MDA-MB-231 and MDA-MB-468. The results obtained showed that PGG reduced the expression of the cytokine GRO-α/CXCL1. PGG also inhibited IƙBKE and MAPK1 genes and the protein expression of IƙBKE and MAPK, indicating that GRO-α downregulation is possibly through NFƙB and MAPK signaling pathway. PGG also inhibited cell proliferation in both cell lines. Moreover, PGG induced apoptosis, modulating caspases, and TNF superfamily receptor genes. It also augmented mRNA of receptors DR4 and DR5 expression, which binds to TNF-related apoptosis-induced ligand, a potent and specific stimulator of apoptosis in tumors. Remarkably, PGG induced a 154-fold increase in TNF expression in MDA-MB-468 compared to a 14.6-fold increase in MDA-MB-231 cells. These findings indicate PGG anti-cancer ability in inhibiting tumor cell proliferation and GRO-α release and inducing apoptosis by increasing TNF and TNF family receptors' expression. Thus, PGG use may be recommended as an adjunct therapy for TNBC to increase chemotherapy effectiveness and prevent cancer progression.
Collapse
Affiliation(s)
- Patricia Mendonca
- Division of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Room G134 H Pharmacy Building, 1415 ML King Blvd, Tallahassee, FL, 32307, USA
| | - Sumaih Alghamdi
- Division of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Room G134 H Pharmacy Building, 1415 ML King Blvd, Tallahassee, FL, 32307, USA
| | - Samia Messeha
- Division of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Room G134 H Pharmacy Building, 1415 ML King Blvd, Tallahassee, FL, 32307, USA
| | - Karam F A Soliman
- Division of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Room G134 H Pharmacy Building, 1415 ML King Blvd, Tallahassee, FL, 32307, USA.
| |
Collapse
|
22
|
Ramírez-de-Arellano A, Pereira-Suárez AL, Rico-Fuentes C, López-Pulido EI, Villegas-Pineda JC, Sierra-Diaz E. Distribution and Effects of Estrogen Receptors in Prostate Cancer: Associated Molecular Mechanisms. Front Endocrinol (Lausanne) 2021; 12:811578. [PMID: 35087479 PMCID: PMC8786725 DOI: 10.3389/fendo.2021.811578] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 12/15/2021] [Indexed: 12/19/2022] Open
Abstract
Estrogens are hormones that have been extensively presented in many types of cancer such as breast, uterus, colorectal, prostate, and others, due to dynamically integrated signaling cascades that coordinate cellular growth, differentiation, and death which can be potentially new therapeutic targets. Despite the historical use of estrogens in the pathogenesis of prostate cancer (PCa), their biological effect is not well known, nor their role in carcinogenesis or the mechanisms used to carry their therapeutic effects of neoplastic in prostate transformation. The expression and regulation of the estrogen receptors (ERs) ERα, ERβ, and GPER stimulated by agonists and antagonists, and related to prostate cancer cells are herein reviewed. Subsequently, the structures of the ERs and their splice variants, the binding of ligands to ERs, and the effect on PCa are provided. Finally, we also assessed the contribution of molecular simulation which can help us to search and predict potential estrogenic activities.
Collapse
Affiliation(s)
- Adrián Ramírez-de-Arellano
- Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
| | - Ana Laura Pereira-Suárez
- Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
- Departamento de Microbiología y Patología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
| | - Cecilia Rico-Fuentes
- Centro Universitario de los Altos, Universidad de Guadalajara, Tepatitlán de Morelos, Mexico
| | - Edgar Iván López-Pulido
- Centro Universitario de los Altos, Universidad de Guadalajara, Tepatitlán de Morelos, Mexico
| | - Julio César Villegas-Pineda
- Departamento de Microbiología y Patología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
| | - Erick Sierra-Diaz
- Departamentos de Clínicas Quirúrgicas y Salud Pública, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
- Departamento de Urología, Hospital de Especialidades Centro Médico Nacional de Occidente, Guadalajara, Mexico
- *Correspondence: Erick Sierra-Diaz,
| |
Collapse
|
23
|
Selective activation of the estrogen receptor-β by the polysaccharide from Cynanchum wilfordii alleviates menopausal syndrome in ovariectomized mice. Int J Biol Macromol 2020; 165:1029-1037. [PMID: 32991896 DOI: 10.1016/j.ijbiomac.2020.09.165] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 09/11/2020] [Accepted: 09/20/2020] [Indexed: 12/14/2022]
Abstract
The menopausal syndrome caused by rapid changes in hormone levels greatly influences the quality of life of women. Though hormone replacement therapy (HRT) is widely used to treat the menopausal syndrome, it exhibits many side effects, including the risk of thrombosis, cardiovascular diseases, and increased incidence of breast cancer; thus, diversifying the interest for phytotherapy-based materials as alternatives to HRT. Here, we isolated a crude polysaccharide fraction (CWPF) from Cynanchum wilfordii root that alleviated the ovariectomy-induced uterine atrophy and bone loss without changes in plasma estradiol concentration in mice. Increased plasma levels of follicle-stimulating hormone (FSH), alkaline phosphatase (ALP), osteocalcin (OC) in ovariectomized mice were also reduced to normal levels by CWPF administration. We found that the inhibitory effects of CWPF on menopausal symptoms were mediated by the estrogen receptor β (ER-β) specific activation, not ER-α. Moreover, CWPF treatment suppressed the phosphorylation of Akt, suggesting that CWPF alleviates post-menopausal symptoms by regulating ER-β related Akt signaling pathway. These results demonstrate that the polysaccharides corresponding to CWPF among the water-soluble extracts of CW could be used as a beneficial herbal alternative for the development of therapeutic agents to prevent menopausal syndrome in women.
Collapse
|
24
|
Zhu J, Lv Y, Hao J, Shi T, Wang S, Wang K, Fan X, Guo Y, Zhang J, Li J. N-myc downstream-regulated gene 2 promotes the protein stability of estrogen receptor beta via inhibition of ubiquitin-protein ligase E3A to suppress colorectal cancer. J Gastrointest Oncol 2020; 11:1200-1213. [PMID: 33456993 DOI: 10.21037/jgo-20-557] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background N-myc downstream-regulated gene 2 (NDRG2) and estrogen receptor beta (ERβ) both play key roles in cellular differentiation in colorectal cancer (CRC). Previous studies have demonstrated that ERβ co-locates with and directly transactivates NDRG2. However, the effect of NDRG2 on ERβ and its underlying mechanism remain largely unknown. Our aim of the study is to explore the effect of NDRG2 on ERβ and their contributions to progression of CRC. Methods The Cancer Genome Atlas (TCGA) database was first utilized to validate the clinical significance of ERβ and NDRG2 in CRC. MTT and scratch migration assays were carried out to verify the role of ERβ and NDRG2 in CRC cells. Western blotting and polymerase chain reaction were performed to analyze the effect of NDRG2 on ERβ, and an immunoprecipitation assay was conducted to explore the protein-protein interaction. Results ERβ and NDRG2 were both found to be significantly down-regulated in tumor tissues from the TCGA-CRC database. NDRG2 was also observed to enhance the protein stability of ERβ while could not change messenger RNA (mRNA) level of ESR2 (encoding ERβ). A positive relationship was found to exist between the two proteins in CRC cells, with NDRG2 prolonging the half-life of ERβ and improving its nuclear translocation. Through detecting expression of ERβ downstream genes (such as TP53 and JNK) and performing related function experiment, we demonstrated that NDRG2 could promote transcriptional activation of ERβ target genes and enhance the function of tumor suppressors when the ERβ agonist diarylpropionitrile (DPN). The immunoprecipitation assay showed that NDRG2 could affect the complex components of ubiquitin-protein ligase E3A (UBE3A, known as E6AP) and ERβ, reducing the ubiquitin-mediated proteasome degradation of ERβ. Conclusions In the current study, we found that NDRG2 could bind with UBE3A to hinder the binding of UBE3A with ERβ. Moreover, a positive feedback loop was discovered between NDRG2 and ERβ, which provides a novel insight and therapeutic target for CRC.
Collapse
Affiliation(s)
- Jun Zhu
- State Key Laboratory of Cancer Biology, Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yongzhi Lv
- The First Affiliated Hospital of Xi'an Medical University, Xi'an, China
| | - Jun Hao
- Department of Experiment Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Tingyu Shi
- Department of Basic Medicine, The Fourth Military Medical University, Xi'an, China
| | - Shuai Wang
- State Key Laboratory of Cancer Biology, Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Ke Wang
- State Key Laboratory of Cancer Biology, Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Xiaoyan Fan
- Department of Experiment Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yuan Guo
- School of Clinical Medicine, Xi'an Medical University, Xi'an, China
| | - Jian Zhang
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, The Fourth Military Medical University, Xi'an, China
| | - Jipeng Li
- State Key Laboratory of Cancer Biology, Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
25
|
Mal R, Magner A, David J, Datta J, Vallabhaneni M, Kassem M, Manouchehri J, Willingham N, Stover D, Vandeusen J, Sardesai S, Williams N, Wesolowski R, Lustberg M, Ganju RK, Ramaswamy B, Cherian MA. Estrogen Receptor Beta (ERβ): A Ligand Activated Tumor Suppressor. Front Oncol 2020; 10:587386. [PMID: 33194742 PMCID: PMC7645238 DOI: 10.3389/fonc.2020.587386] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 09/15/2020] [Indexed: 12/12/2022] Open
Abstract
Estrogen receptor alpha (ERα) and estrogen receptor beta (ERβ) belong to a superfamily of nuclear receptors called steroid hormone receptors, which, upon binding ligand, dimerize and translocate to the nucleus where they activate or repress the transcription of a large number of genes, thus modulating critical physiologic processes. ERβ has multiple isoforms that show differing association with prognosis. Expression levels of the full length ERβ1 isoform are often lower in aggressive cancers as compared to normal tissue. High ERβ1 expression is associated with improved overall survival in women with breast cancer. The promise of ERβ activation, as a potential targeted therapy, is based on concurrent activation of multiple tumor suppressor pathways with few side effects compared to chemotherapy. Thus, ERβ is a nuclear receptor with broad-spectrum tumor suppressor activity, which could serve as a potential treatment target in a variety of human cancers including breast cancer. Further development of highly selective agonists that lack ERα agonist activity, will be necessary to fully harness the potential of ERβ.
Collapse
Affiliation(s)
- Rahul Mal
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Alexa Magner
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Joel David
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Jharna Datta
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Meghna Vallabhaneni
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Mahmoud Kassem
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States.,Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH, United States
| | - Jasmine Manouchehri
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Natalie Willingham
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Daniel Stover
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States.,Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH, United States
| | - Jeffery Vandeusen
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States.,Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH, United States
| | - Sagar Sardesai
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States.,Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH, United States
| | - Nicole Williams
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States.,Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH, United States
| | - Robert Wesolowski
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States.,Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH, United States
| | - Maryam Lustberg
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States.,Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH, United States
| | - Ramesh K Ganju
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Bhuvaneswari Ramaswamy
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States.,Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH, United States
| | - Mathew A Cherian
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States.,Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
26
|
Abstract
Breast cancer, a malignant tumor originating from mammary epithelial tissue, is the most common cancer among women worldwide. Challenges facing the diagnosis and treatment of breast cancer necessitate the search for new mechanisms and drugs to improve outcomes. Estrogen receptor (ER) is considered to be important for determining the diagnosis and treatment strategy. The discovery of the second estrogen receptor, ERβ, provides an opportunity to understand estrogen action. The emergence of ERβ can be traced back to 1996. Over the past 20 years, an increasing body of evidence has implicated the vital effect of ERβ in breast cancer. Although there is controversy among scholars, ERβ is generally thought to have antiproliferative effects in disease progression. This review summarizes available evidence regarding the involvement of ERβ in the clinical treatment and prognosis of breast cancer and describes signaling pathways associated with ERβ. We hope to highlight the potential of ERβ as a therapeutic target.
Collapse
|
27
|
Important roles of estrogen receptor alpha in tumor progression and anti-estrogen therapy of pancreatic ductal adenocarcinoma. Life Sci 2020; 260:118302. [PMID: 32827543 DOI: 10.1016/j.lfs.2020.118302] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 08/06/2020] [Accepted: 08/17/2020] [Indexed: 02/07/2023]
Abstract
AIMS The roles of estrogen receptors (ERs) and the efficacy of anti-estrogen (E2) therapies in pancreatic cancer stay controversial. The main objectives of this study were to investigate the potential roles of ERs in tumor progression and endocrine therapies. MAIN METHODS The ER expression status in PANC-1 and SW1990 pancreatic cancer cell lines was determined. SRB assay, colony formation assay and proliferation assay were used to investigate the responses of these cells to E2. ERα-selective agonist propylpyrazoletriol (PPT), ERβ-selective agonist diarylpropionitrile (DPN), ERα over-expressed SW1990 cells, ERα knock-out PANC-1 cells and patient-derived xenografts (PDX) were applied to investigate the potential roles of ERα in pancreatic cancer. The phosphorylation of ERα-related signaling molecules extracellular regulated protein kinases (ERK1/2) and protein kinase B (AKT) were investigated. The in vivo anti-tumor efficacy and safety of letrozole (LTZ) combined with leuprorelin acetate (LA) and gemcitabine (GEM) were also preliminarily studied. KEY FINDINGS PANC-1 cells expressed much more ERα than SW1990 cells, and ERβ level was with less diversity. Accordingly, the proliferation of PANC-1 rather than SW1990 cells could be stimulated by E2, and only PANC-1 could respond to LTZ endocrine therapy in female but not male mice. The phosphorylation of ERK1/2 but not AKT was altered by over-expressed or knocking out of ERα with or without the addition of E2 and LTZ. The combination therapy of LTZ and GEM showed acceptable efficacy and safety. SIGNIFICANCE This study showed the important roles of ERα in tumor progression and endocrine therapies of pancreatic cancer in women.
Collapse
|
28
|
Klinge CM. Estrogenic control of mitochondrial function. Redox Biol 2020; 31:101435. [PMID: 32001259 PMCID: PMC7212490 DOI: 10.1016/j.redox.2020.101435] [Citation(s) in RCA: 131] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 01/14/2020] [Accepted: 01/16/2020] [Indexed: 12/15/2022] Open
Abstract
Sex-based differences in human disease are caused in part by the levels of endogenous sex steroid hormones which regulate mitochondrial metabolism. This review updates a previous review on how estrogens regulate metabolism and mitochondrial function that was published in 2017. Estrogens are produced by ovaries and adrenals, and in lesser amounts by adipose, breast stromal, and brain tissues. At the cellular level, the mechanisms by which estrogens regulate diverse cellular functions including reproduction and behavior is by binding to estrogen receptors α, β (ERα and ERβ) and G-protein coupled ER (GPER1). ERα and ERβ are transcription factors that bind genomic and mitochondrial DNA to regulate gene transcription. A small proportion of ERα and ERβ interact with plasma membrane-associated signaling proteins to activate intracellular signaling cascades that ultimately alter transcriptional responses, including mitochondrial morphology and function. Although the mechanisms and targets by which estrogens act directly and indirectly to regulate mitochondrial function are not fully elucidated, it is clear that estradiol regulates mitochondrial metabolism and morphology via nuclear and mitochondrial-mediated events, including stimulation of nuclear respiratory factor-1 (NRF-1) transcription that will be reviewed here. NRF-1 is a transcription factor that interacts with coactivators including peroxisome proliferator-activated receptor gamma, coactivator 1 alpha (PGC-1α) to regulate nuclear-encoded mitochondrial genes. One NRF-1 target is TFAM that binds mtDNA to regulate its transcription. Nuclear-encoded miRNA and lncRNA regulate mtDNA-encoded and nuclear-encoded transcripts that regulate mitochondrial function, thus acting as anterograde signals. Other estrogen-regulated mitochondrial activities including bioenergetics, oxygen consumption rate (OCR), and extracellular acidification (ECAR), are reviewed.
Collapse
Affiliation(s)
- Carolyn M Klinge
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, 40292, KY, USA.
| |
Collapse
|
29
|
Jiang W, Wang X, Zhang C, Xue L, Yang L. Expression and clinical significance of MAPK and EGFR in triple-negative breast cancer. Oncol Lett 2020; 19:1842-1848. [PMID: 32194678 PMCID: PMC7038935 DOI: 10.3892/ol.2020.11274] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 09/27/2019] [Indexed: 12/26/2022] Open
Abstract
To investigate the expression and clinical significance of mitogen-activated protein kinase (MAPK) and epidermal growth factor receptor (EGFR) in triple-negative breast cancer (TNBC), a total of 300 TNBC and 120 paired paracancerous tissues were examined. Immunohistochemistry was conducted to determine the expression levels of MAPK and EGFR, and the correlation between MAPK and EGFR expression was evaluated using Cramer's V test. The association between MAPK and EGFR expression, and various clinicopathological variables (such as lymph node metastasis, clinical stage, recurrence and metastasis) was also evaluated, using the χ2 test. MAPK and EGFR expression levels in TNBC tissues were significantly higher than in the paired paracancerous tissues. Moreover, MAPK expression was associated with that of EGFR in TNBC tissues. The positive expression rates of MAPK and EGFR in patients with lymph node metastasis, advanced clinical stage, tumor recurrence and metastasis were higher than those without. Patients with positive expression of MAPK and EGFR in TNBC tissues had poorer prognoses and lower overall survival times than those without expression. In summary, the expression of MAPK and EGFR is closely associated with tumor invasion and the metastasis of TNBC, and may therefore be used as an indicator of poor prognosis in patients with TNBC.
Collapse
Affiliation(s)
- Weihua Jiang
- Department of Breast Surgery, The 3rd Affiliated Teaching Hospital of Xinjiang Medical University (Affiliated Cancer Hospital), Urumqi, Xinjiang 830011, P.R. China
| | - Xiaowen Wang
- Department of Breast Surgery, The 3rd Affiliated Teaching Hospital of Xinjiang Medical University (Affiliated Cancer Hospital), Urumqi, Xinjiang 830011, P.R. China
| | - Chenguang Zhang
- Department of Breast Surgery, The 3rd Affiliated Teaching Hospital of Xinjiang Medical University (Affiliated Cancer Hospital), Urumqi, Xinjiang 830011, P.R. China
| | - Laiti Xue
- Department of Breast Surgery, The 3rd Affiliated Teaching Hospital of Xinjiang Medical University (Affiliated Cancer Hospital), Urumqi, Xinjiang 830011, P.R. China
| | - Liang Yang
- Department of Breast Surgery, The 3rd Affiliated Teaching Hospital of Xinjiang Medical University (Affiliated Cancer Hospital), Urumqi, Xinjiang 830011, P.R. China
| |
Collapse
|
30
|
Pattarawat P, Wallace S, Pfisterer B, Odoi A, Wang HCR. Formulation of a triple combination gemcitabine plus romidepsin + cisplatin regimen to efficaciously and safely control triple-negative breast cancer tumor development. Cancer Chemother Pharmacol 2019; 85:141-152. [PMID: 31865420 DOI: 10.1007/s00280-019-04013-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Accepted: 12/13/2019] [Indexed: 12/22/2022]
Abstract
PURPOSE Triple-negative breast cancer (TNBC) is an aggressive, lethal, and heterogeneous subtype of breast cancers, tending to have lower 5-year survival rates than other BC subtypes in response to conventional chemotherapies. This study's aim was to identify advanced regimens to effectively control TNBC tumor development. METHODS We investigated the combination of the DNA synthesis inhibitor gemcitabine, the DNA-damaging agent cisplatin, and the histone deacetylase inhibitor romidepsin to control a variety of breast cells in vitro. We studied the toxicity of drug doses and administration schedules to determine tolerable combination regimens in immune-deficient nude and -competent BALB/c mice. We then studied the efficacy of tolerable regimens in controlling TNBC cell-derived xenograft development in nude mice. By reducing clinically equivalent doses of each agent in combination, we formulated tolerable regimens in animals. We verified that the tolerable triple combination gemcitabine plus romidepsin + cisplatin regimen more efficacious than double combination regimens in controlling xenograft tumor development in nude mice. RESULTS A triple combination of gemcitabine + romidepsin + cisplatin synergistically induced death of the TNBC M.D. Anderson-Metastatic Breast cancer (MDA-MB) 231 and MDA-MB468, as well as Michigan Cancer Foundation (MCF) 7, MCF10A, and MCF10A-Ras cells. Cell death induced by gemcitabine + romidepsin + cisplatin was in a reactive oxygen species-dependent manner. CONCLUSION Considering the high costs for developing a new anticancer agent, we used the FDA-approved drugs gemcitabine, romidepsin (is approved for T-cell lymphoma and is under clinical trial for TNBC), and cisplatin to economically formulate an efficacious and safe combination regimen. The highly efficacious gemcitabine plus romidepsin + cisplatin regimen should be poised for efficient translation into clinical trials, ultimately contributing to reduced mortality and improved quality of life for TNBC patients.
Collapse
Affiliation(s)
- Pawat Pattarawat
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, 2407 River Drive, Knoxville, TN, 37996, USA.,UT-ORNL Graduate School of Genome Science and Technology, University of Tennessee, Knoxville, TN, 37996, USA
| | - Shelby Wallace
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, 2407 River Drive, Knoxville, TN, 37996, USA
| | - Bianca Pfisterer
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, 2407 River Drive, Knoxville, TN, 37996, USA
| | - Agricola Odoi
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, 2407 River Drive, Knoxville, TN, 37996, USA
| | - Hwa-Chain Robert Wang
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, 2407 River Drive, Knoxville, TN, 37996, USA. .,UT-ORNL Graduate School of Genome Science and Technology, University of Tennessee, Knoxville, TN, 37996, USA.
| |
Collapse
|
31
|
Direito I, Fardilha M, Helguero LA. Contribution of the unfolded protein response to breast and prostate tissue homeostasis and its significance to cancer endocrine response. Carcinogenesis 2019; 40:203-215. [PMID: 30596981 DOI: 10.1093/carcin/bgy182] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Revised: 12/05/2018] [Accepted: 12/14/2018] [Indexed: 12/25/2022] Open
Abstract
Resistant breast and prostate cancers remain a major clinical problem, new therapeutic approaches and better predictors of therapeutic response are clearly needed. Because of the involvement of the unfolded protein response (UPR) in cell proliferation and apoptosis evasion, an increasing number of publications support the hypothesis that impairments in this network trigger and/or exacerbate cancer. Moreover, UPR activation could contribute to the development of drug resistance phenotypes in both breast and prostate cancers. Therefore, targeting this pathway has recently emerged as a promising strategy in anticancer therapy. This review addresses the contribution of UPR to breast and prostate tissues homeostasis and its significance to cancer endocrine response with focus on the current progress on UPR research related to cancer biology, detection, prognosis and treatment.
Collapse
Affiliation(s)
| | - Margarida Fardilha
- Signal Transduction Laboratory, Department of Medical Sciences, Institute for Biomedicine (iBiMED), Universidade de Aveiro, Aveiro, Portugal
| | | |
Collapse
|
32
|
Kim JN, Kim BJ. Depolarization of pacemaker potentials by caffeic acid phenethyl ester in interstitial cells of Cajal from the murine small intestine. Can J Physiol Pharmacol 2019; 98:201-210. [PMID: 31689119 DOI: 10.1139/cjpp-2019-0452] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Interstitial cells of Cajal (ICCs) are pacemaker cells in the gastrointestinal (GI) tract and generate pacemaker potentials. In this study, we investigated the effects of caffeic acid phenethyl ester (CAPE) on the pacemaker potentials of ICCs from the mouse small or large intestine. Using the whole-cell patch-clamp configuration, we found that CAPE depolarized the pacemaker potentials of cultured ICCs from the murine small intestine in a dose-dependent manner. The estrogen receptor (ER) β antagonist PHTPP completely inhibited CAPE-induced depolarization, but the ERα antagonist BHPI did not. Intracellular GDP-β-S and pretreatment with Ca2+-free solution or thapsigargin also blocked CAPE-induced depolarization. To investigate the mechanisms of CAPE-mediated depolarization of ICCs, we used the nonselective cation channel (NSCC) inhibitor flufenamic acid, the Cl- channel blocker, mitogen-activated protein kinase (MAPK) inhibitors PD98059, SB203580, or SP600125, and PI3 kinase inhibitor LY294002. All inhibitors blocked the CAPE-induced pacemaker potential depolarization of ICCs. These results suggest that CAPE induces pacemaker potential depolarization through ERβ in a G protein, NSCC, Cl- channel, MAPK- and PI3 kinase dependent manner via intracellular and extracellular Ca2+ regulation in the murine small intestine. CAPE may therefore modulate GI motility by acting on ICCs in the murine small intestine.
Collapse
Affiliation(s)
- Jeong Nam Kim
- Division of Longevity and Biofunctional Medicine, Pusan National University School of Korean Medicine, Yangsan 50612, Republic of Korea.,Healthy Aging Korean Medical Research Center, Pusan National University School of Korean Medicine, Yangsan 50612, Republic of Korea
| | - Byung Joo Kim
- Division of Longevity and Biofunctional Medicine, Pusan National University School of Korean Medicine, Yangsan 50612, Republic of Korea.,Healthy Aging Korean Medical Research Center, Pusan National University School of Korean Medicine, Yangsan 50612, Republic of Korea
| |
Collapse
|
33
|
Makar S, Saha T, Swetha R, Gutti G, Kumar A, Singh SK. Rational approaches of drug design for the development of selective estrogen receptor modulators (SERMs), implicated in breast cancer. Bioorg Chem 2019; 94:103380. [PMID: 31757413 DOI: 10.1016/j.bioorg.2019.103380] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 10/18/2019] [Accepted: 10/21/2019] [Indexed: 12/20/2022]
Abstract
Drug discovery and development have gained momentum due to the rational drug design by engaging computational tools and bioinformatics methodologies. Bioisosteric replacements and hybrid molecular approaches are the other inventive processes, used by medicinal chemists for the desired modifications of leads for clinical drug candidates. SERMs, ought to produce inhibitory activity in breast, uterus and agonist activity in other tissues, are beneficial for estrogen-like actions. ER subtypes α and β are hormone dependent modulators of intracellular signaling and gene expression, and development of ER selective ligands could be an effective approach for treatment of breast cancer. This report has critically investigated the possible designing considerations of SERMs, their in silico interactions, and potent pharmacophore generation approaches viz. indole, restricted benzothiophene [3, 2-b] indole, carborane, xanthendione, combretastatin A-4, organometallic heterocycles, OBHS-SAHA hybrids, benzopyranones, tetrahydroisoquinolines, Dig G derivatives and their specifications in drug design and development, to rationally improve the understanding in drug discovery. This also includes various strategies for the development of dual inhibitors for the management of antiestrogenic resistance.
Collapse
Affiliation(s)
- Subhajit Makar
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi 221005, U.P, India
| | - Tanmay Saha
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi 221005, U.P, India
| | - Rayala Swetha
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi 221005, U.P, India
| | - Gopichand Gutti
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi 221005, U.P, India
| | - Ashok Kumar
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi 221005, U.P, India
| | - Sushil K Singh
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi 221005, U.P, India.
| |
Collapse
|
34
|
Saha T, Makar S, Swetha R, Gutti G, Singh SK. Estrogen signaling: An emanating therapeutic target for breast cancer treatment. Eur J Med Chem 2019; 177:116-143. [PMID: 31129450 DOI: 10.1016/j.ejmech.2019.05.023] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 04/29/2019] [Accepted: 05/06/2019] [Indexed: 12/15/2022]
Abstract
Breast cancer, a most common malignancy in women, was known to be associated with steroid hormone estrogen. The discovery of estrogen receptor (ER) gave us not only a powerful predictive and prognostic marker, but also an efficient target for the treatment of hormone-dependent breast cancer with various estrogen ligands. ER consists of two subtypes i.e. ERα and ERβ, that are mostly G-protein-coupled receptors and activated by estrogen, specially 17β-estradiol. The activation is followed by translocation into the nucleus and binding with DNA to modulate activities of different genes. ERs can manage synthesis of RNA through genomic actions without directly binding to DNA. Receptors are tethered by protein-protein interactions to a transcription factor complex to communicate with DNA. Estrogens also exhibit nongenomic actions, a characteristic feature of steroid hormones, which are so rapid to be considered by the activation of RNA and translation. These are habitually related to stimulation of different protein kinase cascades. Majority of post-menopausal breast cancer is estrogen dependent, mostly potent biological estrogen (E2) for continuous growth and proliferation. Estrogen helps in regulating the differentiation and proliferation of normal breast epithelial cells. In this review we have investigated the important role of ER in development and progression of breast cancer, which is complicated by receptor's interaction with co-regulatory proteins, cross-talk with other signal transduction pathways and development of treatment strategies viz. selective estrogen receptor modulators (SERMs), selective estrogen receptor down regulators (SERDs), aromatase and sulphatase inhibitors.
Collapse
Affiliation(s)
- Tanmay Saha
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, 221005, U.P, India
| | - Subhajit Makar
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, 221005, U.P, India
| | - Rayala Swetha
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, 221005, U.P, India
| | - Gopichand Gutti
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, 221005, U.P, India
| | - Sushil K Singh
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, 221005, U.P, India.
| |
Collapse
|
35
|
Wei S, Zhou X, Niu M, Zhang H, Liu X, Wang R, Li P, Li H, Cai H, Zhao Y. Network pharmacology exploration reveals the bioactive compounds and molecular mechanisms of Li-Ru-Kang against hyperplasia of mammary gland. Mol Genet Genomics 2019; 294:1159-1171. [PMID: 31053932 DOI: 10.1007/s00438-019-01569-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Accepted: 04/24/2019] [Indexed: 12/13/2022]
Abstract
Li-Ru-Kang (LRK) has been commonly used in the treatment of hyperplasia of mammary gland (HMG) as a cipher prescription and achieved obvious therapeutic effects. However, the bioactive compounds and underlying pharmacological mechanisms remain unclear. This study aims to decipher the bioactive compounds and potential action mechanisms of LRK in the treatment of HMG using an integrated pharmacology approach. The ingredients of LRK and the corresponding drug targets were retrieved through drug target databases and were used to construct the "compound-target-disease" network and function-pathway network. Ultimately, 89 compounds and 2150 drug targets were collected. Gene ontology enrichment analysis revealed that mammary gland alveolus development and mammary gland lobule development were the key biological processes and were regulated simultaneously by three direct targets, including androgen receptor (AR), estrogen receptor (ER) and cyclin-D1. Moreover, 14 compounds of LRK were directly involved in the regulation of the three aforementioned targets. KEGG pathway enrichment analysis found that five signaling pathways and seven direct targets were closely related with HMG treatment by LRK. The results of animal experiments showed that LRK significantly improved the histopathological status of HMG in rats. Additionally, LRK markedly regulated the protein expressions of AR, cyclin-D1, MMP2, MMP3 and MMP9. But interestingly, the effect of LRK on ER was not obvious. This study demonstrated that LRK exerted its therapeutic efficacy based on multi-components, multi-targets and multi-pathways. This research confirms the advantages of network pharmacology analyses and the necessity for experimental verification.
Collapse
Affiliation(s)
- Shizhang Wei
- Department of Pharmacy, the Fifth Medical Center of PLA General Hospital, 100 Western 4th Ring Road, Beijing, 100039, China
| | - Xuelin Zhou
- Department of Pharmacy, the Fifth Medical Center of PLA General Hospital, 100 Western 4th Ring Road, Beijing, 100039, China
| | - Ming Niu
- China Military Institute of Chinese Medicine, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
| | - Haizhu Zhang
- College of Pharmacy and Chemistry, Dali University, Dali, 671000, China
| | - Xiaoyi Liu
- China Military Institute of Chinese Medicine, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
| | - Ruilin Wang
- Department of Integrative Medical Center, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
| | - Pengyan Li
- China Military Institute of Chinese Medicine, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
| | - Haotian Li
- Department of Pharmacy, the Fifth Medical Center of PLA General Hospital, 100 Western 4th Ring Road, Beijing, 100039, China
| | - Huadan Cai
- Department of Pharmacy, the Fifth Medical Center of PLA General Hospital, 100 Western 4th Ring Road, Beijing, 100039, China
| | - Yanling Zhao
- Department of Pharmacy, the Fifth Medical Center of PLA General Hospital, 100 Western 4th Ring Road, Beijing, 100039, China.
| |
Collapse
|
36
|
ERβ modulates genistein’s cisplatin-enhancing activities in breast cancer MDA-MB-231 cells via P53-independent pathway. Mol Cell Biochem 2019; 456:205-216. [DOI: 10.1007/s11010-019-03505-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 02/01/2019] [Indexed: 11/25/2022]
|
37
|
Wang Y, Wei S, Gao T, Yang Y, Lu X, Zhou X, Li H, Wang T, Qian L, Zhao Y, Zou W. Anti-Inflammatory Effect of a TCM Formula Li-Ru-Kang in Rats With Hyperplasia of Mammary Gland and the Underlying Biological Mechanisms. Front Pharmacol 2018; 9:1318. [PMID: 30524280 PMCID: PMC6262177 DOI: 10.3389/fphar.2018.01318] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 10/29/2018] [Indexed: 12/22/2022] Open
Abstract
Li-Ru-Kang (LRK), a formula of eight traditional Chinese medicines (TCM), has been used to treat hyperplasia of mammary gland (HMG) in TCM clinics. However, how LRK works in HMG patients is unclear. To explore the possible mechanisms of LRK against HMG, the network pharmacology was used to screen the potential targets and possible pathways that involved in LRK treated HMG. Rat HMG model induced by estrogen and progesterone was used to further verify the effects of the key molecules of LRK selected from the enriched pathways on HMG. Nipple heights and diameters were measured and uterus index was calculated. The histopathological changes of mammary gland tissue were detected by hematoxylin-eosin (H&E) staining. Western blot was used to detect the phosphorylation of ERK, JNK, and P38. And immunohistochemistry staining was performed to evaluate the levels of estrogen receptor α (ERα), progesterone receptor (PR), nuclear factor-(NF-)κB (p65), interleukin-1β (IL-1β), tumor necrosis factor α (TNF-α), cyclooxygenases 2 (COX-2), inducible nitric oxide synthase (iNOS), 8-hydroxy-2′deoxyguanosine (8-OHdG), and nitrotyrosine (NT). Our results indicate that LRK treatment rescues significantly nipples height and diameter, decreases uterus index and ameliorates HMG. LRK treatment also markedly attenuates the over-expression of IL-1β, TNF-α, COX-2, and iNOS, and suppressed the formation of 8-OHdG and NT. Furthermore, LRK treatment significantly inhibits the phosphorylation of JNK, ERK, and p38 and expression of NF-κB (p65), interestingly, LRK treatment has no effect on the expression of ERα and PR. Our data suggest that the LRK treatment protects the mammary glands from the damage of oxidative stress and inflammation induced by estrogen and progesterone, via suppresses of MAPK/NF-κB signaling pathways without affecting on the expression of ERα and PR.
Collapse
Affiliation(s)
- Yingying Wang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,Department of Pharmacy, 302 Military Hospital of China, Beijing, China
| | - Shizhang Wei
- Department of Pharmacy, 302 Military Hospital of China, Beijing, China
| | - Tian Gao
- Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuxue Yang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,Department of Pharmacy, 302 Military Hospital of China, Beijing, China
| | - Xiaohua Lu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,Department of Pharmacy, 302 Military Hospital of China, Beijing, China
| | - Xuelin Zhou
- Department of Pharmacy, 302 Military Hospital of China, Beijing, China
| | - Haotian Li
- Department of Pharmacy, 302 Military Hospital of China, Beijing, China
| | - Tao Wang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,Department of Pharmacy, 302 Military Hospital of China, Beijing, China
| | - Liqi Qian
- Department of Traditional Chinese Medicine, First Affiliated Hospital of Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yanling Zhao
- Department of Pharmacy, 302 Military Hospital of China, Beijing, China
| | - Wenjun Zou
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
38
|
Ambhore NS, Katragadda R, Raju Kalidhindi RS, Thompson MA, Pabelick CM, Prakash YS, Sathish V. Estrogen receptor beta signaling inhibits PDGF induced human airway smooth muscle proliferation. Mol Cell Endocrinol 2018; 476:37-47. [PMID: 29680290 PMCID: PMC6120801 DOI: 10.1016/j.mce.2018.04.007] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 04/09/2018] [Accepted: 04/18/2018] [Indexed: 02/07/2023]
Abstract
Airway smooth muscle (ASM) cell hyperplasia driven by persistent inflammation is a hallmark feature of remodeling in asthma. Sex steroid signaling in the lungs is of considerable interest, given epidemiological data showing more asthma in pre-menopausal women and aging men. Our previous studies demonstrated that estrogen receptor (ER) expression increases in asthmatic human ASM; however, very limited data are available regarding differential roles of ERα vs. ERβ isoforms in human ASM cell proliferation. In this study, we evaluated the effect of selective ERα and ERβ modulators on platelet-derived growth factor (PDGF)-stimulated ASM proliferation and the mechanisms involved. Asthmatic and non-asthmatic primary human ASM cells were treated with PDGF, 17β-estradiol, ERα-agonist and/or ERβ-agonist and/or G-protein-coupled estrogen receptor 30 (GPR30/GPER) agonist and proliferation was measured using MTT and CyQuant assays followed by cell cycle analysis. Transfection of small interfering RNA (siRNA) ERα and ERβ significantly altered the human ASM proliferation. The specificity of siRNA transfection was confirmed by Western blot analysis. Gene and protein expression of cell cycle-related antigens (PCNA and Ki67) and C/EBP were measured by RT-PCR and Western analysis, along with cell signaling proteins. PDGF significantly increased ASM proliferation in non-asthmatic and asthmatic cells. Treatment with PPT showed no significant effect on PDGF-induced proliferation, whereas WAY interestingly suppressed proliferation via inhibition of ERK1/2, Akt, and p38 signaling. PDGF-induced gene expression of PCNA, Ki67 and C/EBP in human ASM was significantly lower in cells pre-treated with WAY. Furthermore, WAY also inhibited PDGF-activated PCNA, C/EBP, cyclin-D1, and cyclin-E. Overall, we demonstrate ER isoform-specific signaling in the context of ASM proliferation. Activation of ERβ can diminish remodeling in human ASM by inhibiting pro-proliferative signaling pathways, and may point to a novel perception for blunting airway remodeling.
Collapse
Affiliation(s)
| | - Rathnavali Katragadda
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND, USA
| | | | - Michael A Thompson
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Christina M Pabelick
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Y S Prakash
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Venkatachalem Sathish
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND, USA; Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
39
|
Pharmacological activation of estrogen receptor beta augments innate immunity to suppress cancer metastasis. Proc Natl Acad Sci U S A 2018; 115:E3673-E3681. [PMID: 29592953 DOI: 10.1073/pnas.1803291115] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Metastases constitute the greatest causes of deaths from cancer. However, no effective therapeutic options currently exist for cancer patients with metastasis. Estrogen receptor β (ERβ), as a member of the nuclear receptor superfamily, shows potent tumor-suppressive activities in many cancers. To investigate whether modulation of ERβ could serve as a therapeutic strategy for cancer metastasis, we examined whether the selective ERβ agonist LY500307 could suppress lung metastasis of triple-negative breast cancer (TNBC) and melanoma. Mechanistically, while we observed that LY500307 potently induced cell death of cancer cells metastasized to lung in vivo, it does not mediate apoptosis of cancer cells in vitro, indicating that the cell death-inducing effects of LY500307 might be mediated by the tumor microenvironment. Pathological examination combined with flow cytometry assays indicated that LY500307 treatment induced significant infiltration of neutrophils in the metastatic niche. Functional experiments demonstrated that LY500307-treated cancer cells show chemotactic effects for neutrophils and that in vivo neutrophil depletion by Ly6G antibody administration could reverse the effects of LY500307-mediated metastasis suppression. RNA sequencing analysis showed that LY500307 could induce up-regulation of IL-1β in TNBC and melanoma cells, which further triggered antitumor neutrophil chemotaxis. However, the therapeutic effects of LY500307 treatment for suppression of lung metastasis was attenuated in IL1B-/- murine models, due to failure to induce antitumor neutrophil infiltration in the metastatic niche. Collectively, our study demonstrated that pharmacological activation of ERβ could augment innate immunity to suppress cancer metastatic colonization to lung, thus providing alternative therapeutic options for cancer patients with metastasis.
Collapse
|
40
|
Ge H, Yan Y, Tian F, Wu D, Huang Y. Prognostic value of estrogen receptor α and estrogen receptor β in gastric cancer based on a meta-analysis and The Cancer Genome Atlas (TCGA) datasets. Int J Surg 2018; 53:24-31. [PMID: 29555527 DOI: 10.1016/j.ijsu.2018.03.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Revised: 02/11/2018] [Accepted: 03/09/2018] [Indexed: 12/11/2022]
Abstract
PURPOSE Numbers of studies have demonstrated that estrogen receptor α and estrogen receptor β have been involved in development of gastric cancer. Hence, we analyzed studies and The Cancer Genome Atlas (TCGA) data of ERs expression and perform this meta-analysis to access the association between ERα or ER β and the clinicopathological characteristics, overall survival time, in GC. METHOD A literature search was performed in PubMed, Cochrane Library, Web of Science, EMBASE database, and Chinese CNKI. Data on the relationship between ERα or ERβ expression and clinicopathological features were extracted. A TCGA dataset including information of the ERs expression and clinical data of GC patients was analyzed. Pooled odds ratios (ORs) and hazard ratios (HRs) with 95% confidence intervals (CIs) were calculated. RESULTS ERα was not associated with cancer risk, lymph node metastasis, infiltration degree, gender, and TNM stage. However, ERβ was negatively associated with lymph node metastasis. ERα expression may be associated with poor prognosis in GC patients. CONCLUSION Estrogen receptors may be related to the progression and deterioration of gastric cancer. However, further high-quality studies are needed to provide more reliable evidence.
Collapse
Affiliation(s)
- Hua Ge
- Department of Gastrointestinal Surgery, The First People's Hospital of Zunyi, Zunyi Medical University, Zunyi, Guizhou, People's Republic of China.
| | - Yan Yan
- Quality Control Department, The First People's Hospital of Zunyi, Zunyi Medical University, Zunyi, Guizhou, People's Republic of China
| | - Fei Tian
- Department of Gastrointestinal Surgery, The First People's Hospital of Zunyi, Zunyi Medical University, Zunyi, Guizhou, People's Republic of China
| | - Di Wu
- Department of Gastrointestinal Surgery, The First People's Hospital of Zunyi, Zunyi Medical University, Zunyi, Guizhou, People's Republic of China
| | - Yongsheng Huang
- Department of Gastrointestinal Surgery, The First People's Hospital of Zunyi, Zunyi Medical University, Zunyi, Guizhou, People's Republic of China
| |
Collapse
|
41
|
Zhang B, Zhang CG, Ji LH, Zhao G, Wu ZY. Estrogen receptor β selective agonist ameliorates liver cirrhosis in rats by inhibiting the activation and proliferation of hepatic stellate cells. J Gastroenterol Hepatol 2018; 33:747-755. [PMID: 28884481 DOI: 10.1111/jgh.13976] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 08/27/2017] [Accepted: 08/28/2017] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIM The aim of this study is to explore the roles of estrogen receptor (ER) subtypes and corresponding agonists/antagonists on the development of cirrhosis and activation and proliferation of hepatic stellate cells (HSCs). METHODS Carbon tetrachloride (CCl4 )-induced cirrhotic ovariectomized rats were administered non-selective ER agonist (β-estradiol, E2), ER selective agonists (ERα agonist, propylpyrazoletriol; ERβ agonist, diarylpropionitrile [DPN]; and G-protein-coupled ER [GPER] agonist, G1), or E2 + ER selective antagonists (ERα antagonist, MPP; ERβ antagonist, PHTPP; and GPER antagonist, G15) for 12 weeks. The expression of the three ER subtypes in livers and HSCs and the effects of the drugs on hepatic fibrosis, isolated HSCs, and uteri were evaluated. RESULTS Selective ER agonists/antagonists had various effects on CCl4 -induced cirrhosis. The cirrhotic rats in the CCl4 + E2, CCl4 + DPN, CCl4 + E2 + MPP, and CCl4 + E2 + G15 groups presented reduced fibrosis scores, compared with those in the CCl4 group. The cirrhotic rats in the E2 + PHTPP group presented increased fibrosis scores that similar to those in the CCl4 group. The ovariectomized rats had enlarged uteri with increased uterus indexes after E2 administration; however, the proliferative effects of E2 were partially blocked by MPP or G15, but not PHTPP. In the in vitro study, DPN attenuated the transformation of quiescent HSCs to activated phenotype, suppressed collagen I, and α-smooth muscle actin expression. DPN also suppressed platelet-derived growth factor-induced proliferation in cultured HSCs, which was reversed by PHTPP. CONCLUSIONS The antifibrogenic effects of estrogen were mediated by ERβ but not ERα or GPER. The ERβ selective agonist exerted a fibrosuppressive effect by inhibiting the activation and proliferation of HSCs, but did not induce uterine hyperplasia.
Collapse
Affiliation(s)
- Bin Zhang
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Cheng-Gang Zhang
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lin-Hua Ji
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Gang Zhao
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhi-Yong Wu
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
42
|
Cui ML, Yang HY, He GQ. Apoptosis induction of colorectal cancer cells HTL-9 in vitro by the transformed products of soybean isoflavones by Ganoderma lucidum. J Zhejiang Univ Sci B 2017; 18:1101-1112. [PMID: 29204990 PMCID: PMC5742293 DOI: 10.1631/jzus.b1700189] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 06/08/2017] [Indexed: 12/15/2022]
Abstract
Soybean isoflavones have been one of the potential preventive candidates for antitumor research in recent years. In this paper, we first studied the transformation of soybean isoflavones with the homogenized slurry of Ganoderma lucidum. The resultant transformed products (TSI) contained (703.21±4.35) mg/g of genistein, with transformed rates of 96.63% and 87.82% of daidzein and genistein, respectively, and TSI also could enrich the bioactive metabolites of G. lucidum. The antitumor effects of TSI on human colorectal cancer cell line HTL-9, human breast cancer cell line MCF-7, and human immortalized gastric epithelial cell line GES-1 were also studied. The 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT) assay showed that TSI could dramatically reduce the viability rates of HTL-9 cells and MCF-7 cells without detectable cytotoxicity on GES-1 normal cells when the TSI concentration was lower than 100 μg/ml. With 100 μg/ml of TSI, HTL-9 cells were arrested in the G1 phase, and late-apoptosis was primarily induced, accompanied with partial early-apoptosis. TSI could induce primarily early-apoptosis by arresting cells in the G1 phase of MCF-7 cells. For HTL-9 cells, Western-blot and reverse-transcriptase polymerase chain reaction (RT-PCR) analysis showed that TSI (100 μg/ml) can up-regulate the expression of Bax, Caspase-3, Caspase-8, and cytochrome c (Cyto-c), indicating that TSI could induce cell apoptosis mainly through the mitochondrial pathway. In addition, the expression of p53 was up-regulated, while the expression of Survivin and nuclear factor κB (NF-κB) was down-regulated. All these results showed that TSI could induce apoptosis of HTL-9 cells by the regulation of multiple apoptosis-related genes.
Collapse
Affiliation(s)
- Mei-lin Cui
- College of Food Science, Shanxi Normal University, Linfen 041004, China
| | - Huan-yi Yang
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China
- Zhejiang Provincial Key Laboratory of Food Microbiology, Zhejiang University, Hangzhou 310058, China
| | - Guo-qing He
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China
- Zhejiang Provincial Key Laboratory of Food Microbiology, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
43
|
Bialesova L, Xu L, Gustafsson JÅ, Haldosen LA, Zhao C, Dahlman-Wright K. Estrogen receptor β2 induces proliferation and invasiveness of triple negative breast cancer cells: association with regulation of PHD3 and HIF-1α. Oncotarget 2017; 8:76622-76633. [PMID: 29100336 PMCID: PMC5652730 DOI: 10.18632/oncotarget.20635] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 08/17/2017] [Indexed: 12/25/2022] Open
Abstract
The two estrogen receptor (ER) subtypes, ERα and ERβ, belong to the nuclear receptor superfamily. The human ERβ variant ERβ2 is proposed to be expressed at higher levels than ERβ1 in many breast tumors and it has been suggested that ERβ2, in contrast to ERβ1, is associated with aggressive phenotypes of various cancers. However, the role of endogenous ERβ2 in breast cancer cells remains elusive. In this study, we identified that triple negative breast cancer (TNBC) cell lines express endogenous ERβ2, but not ERα or ERβ1. This allows novel studies of endogenous ERβ2 functions independent of ERα and ERβ1. We show that overexpression of ERβ2 in TNBC cells increased whereas knockdown of endogenous ERβ2 decreased cell proliferation and cell invasion. To elucidate the molecular mechanism responsible for these cellular phenotypes, we assayed ERβ2 dependent global gene expression profiles. We show that ERβ2 decreases prolyl hydroxylase 3 (PHD3) gene expression and further show that this is associated with increased hypoxia inducible factor 1α (HIF-1α) protein levels, thus providing a possible mechanism for the invasive phenotype. These results are further supported by analysing the expression of ERβ2 and PHD3 in breast tumor samples where a negative correlation between ERβ2 and PHD3 expression was observed. Together, we demonstrate that ERβ2 has an important role in enhancing cell proliferation and invasion, beyond modulation of ERβ and ERβ1 signalling which might contribute to the invasive characteristics of TNBC. The invasive phenotype could potentially be mediated through transcriptional repression of PHD3 and increased HIF-1α protein levels.
Collapse
Affiliation(s)
- Lucia Bialesova
- Department of Biosciences and Nutrition, Novum, Karolinska Institutet, Huddinge S-141 83, Sweden
| | - Li Xu
- Department of Biosciences and Nutrition, Novum, Karolinska Institutet, Huddinge S-141 83, Sweden
| | - Jan-Åke Gustafsson
- Department of Biosciences and Nutrition, Novum, Karolinska Institutet, Huddinge S-141 83, Sweden.,Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX 77204-5056, USA
| | - Lars-Arne Haldosen
- Department of Biosciences and Nutrition, Novum, Karolinska Institutet, Huddinge S-141 83, Sweden
| | - Chunyan Zhao
- Department of Biosciences and Nutrition, Novum, Karolinska Institutet, Huddinge S-141 83, Sweden
| | - Karin Dahlman-Wright
- Department of Biosciences and Nutrition, Novum, Karolinska Institutet, Huddinge S-141 83, Sweden
| |
Collapse
|
44
|
Mansouri S, Farahmand L, Hosseinzade A, Eslami-S Z, Majidzadeh-A K. Estrogen can restore Tamoxifen sensitivity in breast cancer cells amidst the complex network of resistance. Biomed Pharmacother 2017; 93:1320-1325. [PMID: 28747013 DOI: 10.1016/j.biopha.2017.07.057] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 06/24/2017] [Accepted: 07/11/2017] [Indexed: 01/01/2023] Open
Abstract
Breast cancer-related deaths have been on the decline ever since the application of systemic therapies. Chiefly, endocrine therapy, such as Tamoxifen, enhances the survival of estrogen receptor (ER)-positive patients. More than a decade has passed since the introduction of Tamoxifen, however, drug resistance, particularly to Tamoxifen, still remains a major challenge. It has been shown that not only does chronic Tamoxifen exposures induce resistance, but estrogen deprivation can as well. There are two Tamoxifen resistant cell lines, long term estrogen deprived (LTED) cells and cells that have acquired resistance due to long-term exposure to Tamoxifen (Tam-R). Despite having similar cytosolic pathways over-activated in Tam-R and LTED-R cells during the development of resistance, the administration of receptor tyrosine kinases (RTKs) inhibitors fail to restore Tamoxifen sensitivity in LTED-Rs. This alludes to existing differences in the underlying molecular mechanisms of resistance. Surprisingly, despite estrogen being recognized as a breast cancer stimulator; it has recently been introduced as an apoptotic inducer in unresponsive cells. Furthermore, the addition of estrogen to the media of LTED and Tam-R cells triggers cell death, perhaps is functioning as an anti-proliferative agent. In this review, we outline the molecular pathways potentially facilitating estrogen-induced apoptosis in resistant cells.
Collapse
Affiliation(s)
- Sepideh Mansouri
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Leila Farahmand
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Aysooda Hosseinzade
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Zahra Eslami-S
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Keivan Majidzadeh-A
- Genetics Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran.
| |
Collapse
|
45
|
The histone H2A isoform Hist2h2ac is a novel regulator of proliferation and epithelial–mesenchymal transition in mammary epithelial and in breast cancer cells. Cancer Lett 2017; 396:42-52. [DOI: 10.1016/j.canlet.2017.03.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 03/01/2017] [Accepted: 03/03/2017] [Indexed: 01/11/2023]
|
46
|
Oza A, Ma CX. New Insights in Estrogen Receptor (ER) Biology and Implications for Treatment. CURRENT BREAST CANCER REPORTS 2017. [DOI: 10.1007/s12609-017-0231-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
47
|
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: 10.6] [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
|
48
|
Geng J, Li H, Huang C, Chai J, Zheng R, Li F, Jiang S. Functional analysis of HSPA1A and HSPA8 in parturition. Biochem Biophys Res Commun 2017; 483:371-379. [DOI: 10.1016/j.bbrc.2016.12.136] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Accepted: 12/20/2016] [Indexed: 10/20/2022]
|
49
|
Li X, Zhou Y, Liu Y, Zhang X, Chen T, Chen K, Ba Q, Li J, Liu H, Wang H. Preclinical Efficacy and Safety Assessment of Artemisinin-Chemotherapeutic Agent Conjugates for Ovarian Cancer. EBioMedicine 2016; 14:44-54. [PMID: 27939426 PMCID: PMC5161434 DOI: 10.1016/j.ebiom.2016.11.026] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 11/21/2016] [Accepted: 11/21/2016] [Indexed: 01/11/2023] Open
Abstract
Artemisinin (ARS) and its derivatives, which are clinically used antimalarial agents, have shown antitumor activities. Their therapeutic potencies, however, are limited by their low solubility and poor bioavailability. Here, through a pharmacophore hybridization strategy, we synthesized ARS-drug conjugates, in which the marketed chemotherapeutic agents chlorambucil, melphalan, flutamide, aminoglutethimide, and doxifluridine, were separately bonded to Dihydroartemisinin (DHA) through various linkages. Of these, the artemisinin-melphalan conjugate, ARS4, exhibited most toxicity to human ovarian cancer cells but had low cytotoxicity to normal cells. ARS4 inhibited the growth and proliferation of ovarian cancer cells and resulted in S-phase arrest, apoptosis, and inhibition of migration; these effects were stronger than those of its parent drugs, DHA and melphalan. Furthermore, ARS4 modulated the expression of proteins involved in cell cycle progression, apoptosis, and the epithelial–mesenchymal transition (EMT). Moreover, in mice, ARS4 inhibited growth and intraperitoneal dissemination and metastasis of ovarian cancer cells without observable toxic effects. Our results provide a basis for development of the compound as a chemotherapeutic agent. Research in context Artemisinin compounds have recently received attention as anticancer agents because of their clinical safety profiles and broad efficacy. However, their therapeutic potencies are limited by low solubility and poor bioavailability. Here, we report that ARS4, an artemisinin-melphalan conjugate, possesses marked in-vitro and in-vivo antitumor activity against ovarian cancer, the effects of which are stronger than those for its parent drugs, Dihydroartemisinin and melphalan. In mice, ARS4 inhibits localized growth of ovarian cancer cells and intraperitoneal dissemination and metastasis without appreciable host toxicity. Thus, for patients with ovarian cancer, ARS4 is a promising chemotherapeutic agent. Artemisinin-drug conjugates were designed via pharmacophore hybridization strategy ARS4 induced apoptosis of ovarian cancer cells and cell cycle arrest and reversed the EMT polarity In mice, ARS4 inhibited growth and intraperitoneal dissemination of ovarian cancer cells with no appreciable host toxicity
Collapse
Affiliation(s)
- Xiaoguang Li
- School of Public health, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yu Zhou
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yanling Liu
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xu Zhang
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Tao Chen
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Kerong Chen
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Qian Ba
- School of Public health, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jingquan Li
- School of Public health, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Hong Liu
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
| | - Hui Wang
- School of Public health, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Key Laboratory of Food Safety Risk Assessment, Ministry of Health, Beijing, China.
| |
Collapse
|
50
|
Lau KM, To KF. Importance of Estrogenic Signaling and Its Mediated Receptors in Prostate Cancer. Int J Mol Sci 2016; 17:E1434. [PMID: 27589731 PMCID: PMC5037713 DOI: 10.3390/ijms17091434] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 08/16/2016] [Accepted: 08/23/2016] [Indexed: 12/12/2022] Open
Abstract
Prostate cancer (PCa) treatment was first established by Huggins and Hodges in 1941, primarily described as androgen deprivation via interference of testicular androgen production. The disease remains incurable with relapse of hormone-refractory cancer after treatments. Epidemiological and clinical studies disclosed the importance of estrogens in PCa. Discovery of estrogen receptor ERβ prompted direct estrogenic actions, in conjunction with ERα, on PCa cells. Mechanistically, ERs upon ligand binding transactivate target genes at consensus genomic sites via interactions with various transcriptional co-regulators to mold estrogenic signaling. With animal models, Noble revealed estrogen dependencies of PCa, providing insight into potential uses of antiestrogens in the treatment. Subsequently, various clinical trials were conducted and molecular and functional consequences of antiestrogen treatment in PCa were delineated. Besides, estrogens can also trigger rapid non-genomic signaling responses initiated at the plasma membrane, at least partially via an orphan G-protein-coupled receptor GPR30. Activation of GPR30 significantly inhibited in vitro and in vivo PCa cell growth and the underlying mechanism was elucidated. Currently, molecular networks of estrogenic and antiestrogenic signaling via ERα, ERβ and GPR30 in PCa have not been fully deciphered. This crucial information could be beneficial to further developments of effective estrogen- and antiestrogen-based therapy for PCa patients.
Collapse
Affiliation(s)
- Kin-Mang Lau
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Oncology in Southern China, and Sir YK Pao Centre for Cancer, The Chinese University of Hong Kong, Hong Kong, China.
| | - Ka-Fai To
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Oncology in Southern China, and Sir YK Pao Centre for Cancer, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|