1
|
Rinaldi R, Laurino S, Salvia R, Russi S, De Stefano F, Galasso R, Sgambato A, Scieuzo C, Falco G, Falabella P. Biological Activity of Peptide Fraction Derived from Hermetia illucens L. (Diptera: Stratiomyidae) Larvae Haemolymph on Gastric Cancer Cells. Int J Mol Sci 2025; 26:1885. [PMID: 40076512 PMCID: PMC11899352 DOI: 10.3390/ijms26051885] [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: 01/21/2025] [Revised: 02/15/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
Gastric cancer (GC) is one of the leading causes of cancer-related mortality worldwide, characterised by poor prognosis and limited responsiveness to chemotherapy. There is a need for new and more effective anticancer agents. Antimicrobial peptides (AMPs) represent a promising class of biomolecules for this purpose. Naturally occurring in the innate immune system, these peptides can also exert cytotoxic effects against cancer cells, earning them the designation of "anticancer peptides" (ACPs). They have the potential to be a viable support for current chemotherapy schedules due to their selectivity against cancer cells and minor propensity to induce chemoresistance in cells. Insects are an excellent source of AMPs. Among them, due to its ability to thrive in hostile and microorganism-rich environments, we isolated a peptide fraction from Hermetia illucens L. (Diptera: Stratiomyidae) haemolymph to evaluate a possible anticancer activity. We tested Peptide Fractions (PFs) against AGS and KATO III gastric cancer cell lines. Data obtained indicated that PFs, especially those resulting from Escherichia coli and Micrococcus flavus infection (to boost immune response), were able to inhibit tumour cell growth by inducing apoptosis or cell cycle arrest in a cell line-specific manner. These results support further investigation into the use of antimicrobial peptides produced from insects as possible anticancer agents.
Collapse
Affiliation(s)
- Roberta Rinaldi
- Department of Basic and Applied Sciences, University of Basilicata, Via dell’Ateneo Lucano 10, 85100 Potenza, Italy; (R.R.); (R.S.); (F.D.S.)
| | - Simona Laurino
- Centro di Riferimento Oncologico della Basilicata IRCCS (IRCCS-CROB), 85028 Rionero in Vulture, Italy; (S.L.); (S.R.); (R.G.)
| | - Rosanna Salvia
- Department of Basic and Applied Sciences, University of Basilicata, Via dell’Ateneo Lucano 10, 85100 Potenza, Italy; (R.R.); (R.S.); (F.D.S.)
- Spinoff XFlies S.R.L, University of Basilicata, Via Dell’Ateneo Lucano 10, 85100 Potenza, Italy
| | - Sabino Russi
- Centro di Riferimento Oncologico della Basilicata IRCCS (IRCCS-CROB), 85028 Rionero in Vulture, Italy; (S.L.); (S.R.); (R.G.)
| | - Federica De Stefano
- Department of Basic and Applied Sciences, University of Basilicata, Via dell’Ateneo Lucano 10, 85100 Potenza, Italy; (R.R.); (R.S.); (F.D.S.)
| | - Rocco Galasso
- Centro di Riferimento Oncologico della Basilicata IRCCS (IRCCS-CROB), 85028 Rionero in Vulture, Italy; (S.L.); (S.R.); (R.G.)
| | - Alessandro Sgambato
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
- Multiplex Spatial Profiling Facility, Fondazione Policlinico Universitario ‘Agostino Gemelli’ IRCCS, 00136 Rome, Italy
| | - Carmen Scieuzo
- Department of Basic and Applied Sciences, University of Basilicata, Via dell’Ateneo Lucano 10, 85100 Potenza, Italy; (R.R.); (R.S.); (F.D.S.)
- Spinoff XFlies S.R.L, University of Basilicata, Via Dell’Ateneo Lucano 10, 85100 Potenza, Italy
| | - Geppino Falco
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy;
| | - Patrizia Falabella
- Department of Basic and Applied Sciences, University of Basilicata, Via dell’Ateneo Lucano 10, 85100 Potenza, Italy; (R.R.); (R.S.); (F.D.S.)
- Spinoff XFlies S.R.L, University of Basilicata, Via Dell’Ateneo Lucano 10, 85100 Potenza, Italy
| |
Collapse
|
2
|
Mei W, Mei B, Chang J, Liu Y, Zhou Y, Zhu N, Hu M. Role and regulation of FOXO3a: new insights into breast cancer therapy. Front Pharmacol 2024; 15:1346745. [PMID: 38505423 PMCID: PMC10949727 DOI: 10.3389/fphar.2024.1346745] [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: 11/29/2023] [Accepted: 02/16/2024] [Indexed: 03/21/2024] Open
Abstract
Breast cancer is the most common malignancy in the world, particularly affecting female cancer patients. Enhancing the therapeutic strategies for breast cancer necessitates identifying molecular drug targets that effectively eliminate tumor cells. One of these prominent targets is the forkhead and O3a class (FOXO3a), a member of the forkhead transcription factor subfamily. FOXO3a plays a pivotal role in various cellular processes, including apoptosis, proliferation, cell cycle regulation, and drug resistance. It acts as a tumor suppressor in multiple cancer types, although its specific role in cancer remains unclear. Moreover, FOXO3a shows promise as a potential marker for tumor diagnosis and prognosis in breast cancer patients. In addition, it is actively influenced by common anti-breast cancer drugs like paclitaxel, simvastatin, and gefitinib. In breast cancer, the regulation of FOXO3a involves intricate networks, encompassing post-translational modification post-translational regulation by non-coding RNA (ncRNA) and protein-protein interaction. The specific mechanism of FOXO3a in breast cancer urgently requires further investigation. This review aims to systematically elucidate the role of FOXO3a in breast cancer. Additionally, it reviews the interaction of FOXO3a and its upstream and downstream signaling pathway-related molecules to uncover potential therapeutic drugs and related regulatory factors for breast cancer treatment by regulating FOXO3a.
Collapse
Affiliation(s)
- Wenqiu Mei
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
- Department of Neurology, Ezhou Central Hospital, Ezhou, China
| | - Bingyin Mei
- Department of Neurology, Ezhou Central Hospital, Ezhou, China
| | - Jing Chang
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Yifei Liu
- School of Biomedical Engineering, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Yanhong Zhou
- Department of Medical School of Facial Features, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Ni Zhu
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Meichun Hu
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| |
Collapse
|
3
|
Koskela SA, Figueiredo CR. From antimicrobial to anticancer: the pioneering works of Prof. Luiz Rodolpho Travassos on bioactive peptides. Braz J Microbiol 2023; 54:2561-2570. [PMID: 37725261 PMCID: PMC10689714 DOI: 10.1007/s42770-023-01118-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 08/28/2023] [Indexed: 09/21/2023] Open
Abstract
Prof. Luiz Rodolpho Travassos, a distinguished Brazilian scientist, was instrumental in fostering an interdisciplinary research approach that seamlessly combined microbiology and oncology. This work has opened new pathways into the understanding of tumorigenesis and aided in the development of innovative therapeutic tools. One significant area of his work has been the exploration of bioactive peptides, many of which were first identified for their antimicrobial properties. These peptides demonstrate promise as potential cancer therapeutics due to their selectivity, cost-effectiveness, ease of synthesis, low antigenicity, and excellent tissue penetration. Prof. Travassos' pioneering work uncovered on the potential of peptides derived from microbiological sources, such as those obtained using phage display techniques. More importantly, in international cooperation, peptides derived from complementarity-determining regions (CDRs) that showed antimicrobial activity against Candida albicans further showed to be promising tools with cytotoxic properties against cancer cells. Similarly, peptides derived from natural sources, such as the gomesin peptide, not only had shown antimicrobial properties but could treat cutaneous melanoma in experimental models. These therapeutic tools allowed Prof. Travassos and his group to navigate the intricate landscape of factors and pathways that drive cancer development, including persistent proliferative signaling, evasion of tumor suppressor genes, inhibition of programmed cell death, and cellular immortality. This review examines the mechanisms of action of these peptides, aligning them with the universally recognized hallmarks of cancer, and evaluates their potential as drug candidates. It highlights the crucial need for more selective, microbiology-inspired anti-cancer strategies that spare healthy cells, a challenge that current therapies often struggle to address. By offering a comprehensive assessment of Prof. Travassos' innovative contributions and a detailed discussion on the increasing importance of microbiology-derived peptides, this review presents an informed and robust perspective on the possible future direction of cancer therapy.
Collapse
Affiliation(s)
- Saara A Koskela
- Medical Immune Oncology Research Group (MIORG), Institute of Biomedicine, Faculty of Medicine, University of Turku, Turku, Finland
| | - Carlos R Figueiredo
- Medical Immune Oncology Research Group (MIORG), Institute of Biomedicine, Faculty of Medicine, University of Turku, Turku, Finland.
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland.
| |
Collapse
|
4
|
Specific Forms of Graphene Quantum Dots Induce Apoptosis and Cell Cycle Arrest in Breast Cancer Cells. Int J Mol Sci 2023; 24:ijms24044046. [PMID: 36835458 PMCID: PMC9968019 DOI: 10.3390/ijms24044046] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 02/10/2023] [Accepted: 02/11/2023] [Indexed: 02/19/2023] Open
Abstract
Graphene quantum dots (GQDs), nanomaterials derived from graphene and carbon dots, are highly stable, soluble, and have exceptional optical properties. Further, they have low toxicity and are excellent vehicles for carrying drugs or fluorescein dyes. Specific forms of GQDs can induce apoptosis and could be used to treat cancers. In this study, three forms of GQDs (GQD (nitrogen:carbon = 1:3), ortho-GQD, and meta-GQD) were screened and tested for their potential to inhibit breast cancer cell (MCF-7, BT-474, MDA-MB-231, and T-47D) growth. All three GQDs decreased cell viability after 72 h of treatment and specifically affected breast cancer cell proliferation. An assay for the expression of apoptotic proteins revealed that p21 and p27 were up-regulated (1.41-fold and 4.75-fold) after treatment. In particular, ortho-GQD-treated cells showed G2/M phase arrest. The GQDs specifically induced apoptosis in estrogen receptor-positive breast cancer cell lines. These results indicate that these GQDs induce apoptosis and G2/M cell cycle arrest in specific breast cancer subtypes and could potentially be used for treating breast cancers.
Collapse
|
5
|
Jin ML, Yang L, Jeong KW. SETD1A-SOX2 axis is involved in tamoxifen resistance in estrogen receptor α-positive breast cancer cells. Theranostics 2022; 12:5761-5775. [PMID: 35966598 PMCID: PMC9373809 DOI: 10.7150/thno.72599] [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] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 07/02/2022] [Indexed: 12/03/2022] Open
Abstract
Rationale: Approximately 30-40% of estrogen receptor (ER)-positive breast cancer (BC) cases recur after tamoxifen therapy. Thus, additional studies on the mechanisms underlying tamoxifen resistance and more specific prognostic biomarkers are required. In this study, we investigated the role of the SET domain containing 1A (SETD1A), a histone H3-lysine 4 (H3K4) methyltransferase, in the development of tamoxifen resistance in BC. Methods: The relationship between tamoxifen resistance and SETD1A protein level was investigated using resistant cell lines derived from the parent BC cells. Biochemical and molecular assays, such as RNA-sequencing, reverse transcription-quantitative polymerase chain reaction, chromatin-immunoprecipitation, and protein-binding assays, were used to identify the SETD1A target gene in tamoxifen-resistant BC cells. Additionally, the role of SETD1A in cancer stem cells (CSCs) was investigated using CSCs isolated from tamoxifen-resistant BC cells. Comprehensive transcriptome analysis and immunofluorescence staining using clinical datasets and tissue microarray were performed to determine the correlation between the expression of the SETD1A-SRY-box transcription factor 2 (SOX2) pair and recurrence in tamoxifen-treated patients with BC. Results: SETD1A was expressed at higher levels in tamoxifen-resistant BC cells than in primary BC cells. Notably, SETD1A-depleted tamoxifen-resistant MCF-7 cells showed restored sensitivity to tamoxifen, whereas SETD1A overexpression in MCF-7 cells resulted in decreased sensitivity. SETD1A is recruited to the SOX2 gene via its interaction with SOX2, thereby enhancing the expression of SOX2 genes in tamoxifen-resistant BC cells. The growth of tamoxifen-resistant cells and CSCs was effectively suppressed by SETD1A knockdown. In addition, high levels of SETD1A and SOX2 were significantly correlated with a low survival rate in patients with ER-positive tamoxifen-resistant BC. Conclusion: Our findings provide the first evidence of the critical role of the SETD1A-SOX2 axis in tamoxifen-resistant BC cells, implying that SETD1A may serve as a molecular target and prognostic indicator of a therapeutic response in patients with tamoxifen-resistant BC.
Collapse
Affiliation(s)
- Ming Li Jin
- Gachon Research Institute of Pharmaceutical Sciences, College of Pharmacy, Gachon University, 191 Hambakmoero, Yeonsu-gu, Incheon, 21936, Republic of Korea
| | - Liu Yang
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Kwang Won Jeong
- Gachon Research Institute of Pharmaceutical Sciences, College of Pharmacy, Gachon University, 191 Hambakmoero, Yeonsu-gu, Incheon, 21936, Republic of Korea
| |
Collapse
|
6
|
Sarwar S, Alamro A, Huq F, Alghamdi A. Insights Into the Role of Epigenetic Factors Determining the Estrogen Response in Estrogen-Positive Ovarian Cancer and Prospects of Combining Epi-Drugs With Endocrine Therapy. Front Genet 2022; 13:812077. [PMID: 35873467 PMCID: PMC9306913 DOI: 10.3389/fgene.2022.812077] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 04/19/2022] [Indexed: 11/25/2022] Open
Abstract
Ovarian cancer is one of the most lethal malignancies. The population at the risk is continually on the rise due to the acquired drug resistance, high relapse rate, incomplete knowledge of the etiology, cross-talk with other gynecological malignancies, and diagnosis at an advanced stage. Most ovarian tumors are thought to arise in surface epithelium somehow in response to changes in the hormonal environment. Prolonged treatment with hormone replacement therapy (HRT) is also considered a contributing factor. Estrogens influence the etiology and progression of the endocrine/hormone-responsive cancers in a patient-specific manner. The concept of hormonal manipulations got attention during the last half of the 20th century when tamoxifen was approved by the FDA as the first selective estrogen receptor modulator (SERM). Endocrine therapy that has been found to be effective against breast cancer can be an option for ovarian cancer. It is now established that global changes in the epigenetic landscape are not only the hallmark of tumor development but also contribute to the development of resistance to hormone therapy. A set of functionally related genes involved in epigenetic reprogramming are controlled by specific transcription factors (TFs). Thus, the activities of TFs mediate important mechanisms through which epigenetic enzymes and co-factors modify chromatin for the worst outcome in a site-specific manner. Furthermore, the role of epigenetic aberrations involving histone modifications is established in ovarian cancer pathogenesis. This review aims to provide insights on the role of key epigenetic determinants of response as well as resistance to the hormone therapy, the current status of research along with its limitations, and future prospects of epigenetic agents as biomarkers in early diagnosis, prognosis, and personalized treatment strategies. Finally, the possibility of small phytoestrogenic molecules in combination with immunotherapy and epi-drugs targeting ovarian cancer has been discussed.
Collapse
Affiliation(s)
- Sadia Sarwar
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
- *Correspondence: Sadia Sarwar,
| | - Abir Alamro
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Fazlul Huq
- Eman Research Journal, Eman Research, Sydney, NSW, Australia
| | - Amani Alghamdi
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
7
|
S L, A S, Dv S, Bs R, R S, Sharaf S, Sa A, G R. Comparative differential cytotoxicity of clinically used SERMs in human cancer lines of different origin and its predictive molecular docking studies of key target genes involved in cancer progression and treatment responses. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2021; 3:100080. [PMID: 35059624 PMCID: PMC8760488 DOI: 10.1016/j.crphar.2021.100080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 12/18/2021] [Accepted: 12/21/2021] [Indexed: 12/24/2022] Open
Abstract
SERMS like Tamoxifene, 5-hydroxy tamoxifene, raloxifene and endoxifene has been used for the treatment of hormonal imbalances and dependent cancers owing to their action via Estrogen receptors as in the treatment of estrogen sensitive breast cancers. Due to the adverse side effects, modifications and development of the existing or newer SERMS has always been of immense interest. Ormeloxifene, a SERM molecule manufactured by HLL Lifecare Ltd, India as birth control under the trade names Saheli, Novex, and Novex-DS which is also investigated against mastalgia, fibro-adenoma and abnormal uterine bleeding. Anti-cancer effects have been reported in estrogen dependent and independent cancers which shows its wide scope to be implemented in cancer therapy. Current investigation is a comprehensive effort to find the cytotoxic potential of Ormeloxifene in comparison with clinically used four SERMS in twenty six cancer cell lines of different origin using Adriamycin as positive control. Also the computational studies pertaining to selected target/ligand with respect to tumor progression, development, treatment responses and apoptosis. The studies proved effective cytotoxicity of Ormeloxifene on cancer cell lines with lower TGI, GI50 and LC50 values which are significantly comparable. Also the in silico studies proved that the docking score of the compound suggests the interaction of the compound which could tightly regulate key target genes controlling cancer like ER, EGFR kinase, EGFR-cSRC, HDAC-2, PARP-1 and BRAF. This study brings out the superior efficacy of Ormeloxifene compared to other SERMS with proven safety profile to be repositioned as an anti-cancer drug to treat diverse cancer types.
Collapse
Affiliation(s)
- Lakshmi S
- Corporate R&D Centre, HLL Lifecare Limited, Thiruvananthapuram, Pincode- 695 017, India
| | - Shanitha A
- Dept. of Computational Biology & Bioinformatics, University of Kerala, Thiruvananthapuram, Pincode-695581, India
| | - Shiny Dv
- Corporate R&D Centre, HLL Lifecare Limited, Thiruvananthapuram, Pincode- 695 017, India
| | - Rahul Bs
- Corporate R&D Centre, HLL Lifecare Limited, Thiruvananthapuram, Pincode- 695 017, India
| | - Saikant R
- Corporate R&D Centre, HLL Lifecare Limited, Thiruvananthapuram, Pincode- 695 017, India
| | - Shehna Sharaf
- Corporate R&D Centre, HLL Lifecare Limited, Thiruvananthapuram, Pincode- 695 017, India
| | - Abi Sa
- Corporate R&D Centre, HLL Lifecare Limited, Thiruvananthapuram, Pincode- 695 017, India
| | - Rajmohan G
- Corporate R&D Centre, HLL Lifecare Limited, Thiruvananthapuram, Pincode- 695 017, India
| |
Collapse
|
8
|
Synergistic Cytotoxicity between Elephantopus scaber and Tamoxifen on MCF-7-Derived Multicellular Tumor Spheroid. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:6355236. [PMID: 34712346 PMCID: PMC8548115 DOI: 10.1155/2021/6355236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 08/25/2021] [Accepted: 10/04/2021] [Indexed: 11/18/2022]
Abstract
Elephantopus scaber Linn, a traditional herb, exhibited anticancer properties, and it was cytotoxic against the monolayer estrogen receptor-positive breast cancer cell line, MCF-7, in the previous study. In order to determine the potential of E. scaber as a complementary medicine for breast cancer, this study aimed to evaluate the synergism between E. scaber and tamoxifen in cytotoxicity using MCF-7 in the form of 3-dimensional multicellular tumor spheroid (MCTS) cultures. MCTS represents a more reliable model for studying drug penetration as compared to monolayer cells due to its greater resemblance to solid tumor. Combination of E. scaber ethanol extract and tamoxifen, which were used in concentrations lower than their respective IC50 values, had successfully induced apoptosis on MCTS in this study. The combinatorial treatment showed >58% increase of lactate dehydrogenase release in cell media, cell cycle arrest at the S phase, and 1.3 fold increase in depolarization of mitochondrial membrane potential. The treated MCTS also experienced DNA fragmentation; this had been quantified by TUNEL-positive assay, which showed >64% increase in DNA damaged cells. Higher externalization of phospatidylserine and distorted and disintegrated spheroids stained by acridine orange/propidium iodide showed that the cell death was mainly due to apoptosis. Further exploration showed that the combinatorial treatment elevated caspases-8 and 9 activities involving both extrinsic and intrinsic pathways of apoptosis. The treatment also upregulated the expression of proapoptotic gene HSP 105 and downregulated the expression of prosurvival genes such as c-Jun, ICAM1, and VEGF. In conclusion, these results suggested that the coupling of E. scaber to low concentration of tamoxifen showed synergism in cytotoxicity and reducing drug resistance in estrogen receptor-positive breast cancer.
Collapse
|
9
|
Tang J, Luo Y, Long G, Zhou L. MINDY1 promotes breast cancer cell proliferation by stabilizing estrogen receptor α. Cell Death Dis 2021; 12:937. [PMID: 34645792 PMCID: PMC8514509 DOI: 10.1038/s41419-021-04244-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 07/16/2021] [Accepted: 07/20/2021] [Indexed: 12/13/2022]
Abstract
Breast cancer is the most commonly diagnosed malignant tumor among females. Estrogen receptor α (ERα) is initially expressed in 70% of breast cancers and is a well-known target of endocrine therapy for ERα-positive breast cancer. In the present study, we identified MINDY1, a member belongs to the motif interacting with Ubcontaining novel DUB family (MINDY), as a potential deubiquitylase of ERα in breast cancer. There was a positive correlation between ERα and MINDY1 protein levels in human breast cancer tissues. We found that high expression of MINDY1 was associated with poor prognosis. MINDY1 interacted with ERα, thereby mediating the deubiquitination of ERα and increased its stability in a deubiquitylation activity-dependent manner. MINDY1 depletion significantly decreased the ERα protein level and ERα signaling activity in breast cancer cells. Specifically, MINDY1 associated with the N-terminal of ERα via its catalytic domain, thus inhibiting K48-specific poly-ubiquitination process on ERα protein. In addition, MINDY1 depletion led to growth inhibition and cell cycle arrest of ERα-positive breast cancer cells. Finally, overexpression of ERα could rescue the MINDY1 depletion-induced growth inhibition both in vitro and in vivo, suggesting that MINDY1 promotes breast carcinogenesis through increasing ERα stability. Overall, our study proposed a novel post-translational mechanism of ERα in supporting breast cancer progression. Targeting the MINDY1 may prove to be a promising strategy for patients with ERα-positive breast cancer.
Collapse
Affiliation(s)
- Jianing Tang
- Department of Liver Surgery, Xiangya Hospital, Central South University, Changsha, China.
| | - Yongwen Luo
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Guo Long
- Department of Liver Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Ledu Zhou
- Department of Liver Surgery, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
10
|
El-Kaream SAA, Ebied SAEM, Sadek NA, Saad DM, Nadwan EA. Serum estrogen and its soluble receptor levels in Egyptian patients with acute leukemia: case-control study. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2021. [DOI: 10.1186/s43042-021-00186-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Abstract
Background
Acute leukemias are malignant neoplastic diseases that arise from either lymphoid [ALL] or myeloid [AML] cell lines that are distinguished by the proliferation of BM non-functional immature cells and subsequently released into the bloodstream. ALL is prevalent malignancy in young, while AML in older. Diagnosis is usually routinely performed through peripheral blood count and smear then confirmed by BM aspirate. It is remarkable to notice that leukemia can be manifested at high, low, and even at normal leucocyte count. While treatment results have improved steadily over the last decades in younger and adults, limited changes have been in survival among subjects of age > 60 years. Aim of the work is to measure the serum estrogen [E2] and its soluble receptor [ER] levels in acute leukemia patients and extrapolate its possible clinical significance. This study included 40 [20 females and 20 males] healthy volunteers clinically free from any disease, 40 [20 females and 20 males] AML patients, and 40 [20 females and 20 males] ALL. To all subjects, serum E2 and its soluble ER level were investigated by ELISA.
Results
Serum E2 [pg/ml] level was lower in AML and ALL female and male patients groups than control group. Serum ER [ng/ml] level was lower in AML and ALL female and male patients groups than control group.
Conclusion
Estimation of serum E2 and its soluble ER level is of edifying diagnostic value. Determination of serum E2 and its soluble ER level in AML and ALL patients is of value in deciding treatment therapeutic target protocol.
Collapse
|
11
|
Gruslova A, McClellan B, Balinda HU, Viswanadhapalli S, Alers V, Sareddy GR, Huang T, Garcia M, deGraffenried L, Vadlamudi RK, Brenner AJ. FASN inhibition as a potential treatment for endocrine-resistant breast cancer. Breast Cancer Res Treat 2021; 187:375-386. [PMID: 33893909 DOI: 10.1007/s10549-021-06231-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 04/14/2021] [Indexed: 01/18/2023]
Abstract
PURPOSE The majority of breast cancers are estrogen receptor (ERα) positive making endocrine therapy a mainstay for these patients. Unfortunately, resistance to endocrine therapy is a common occurrence. Fatty acid synthase (FASN) is a key enzyme in lipid biosynthesis and its expression is commensurate with tumor grade and resistance to numerous therapies. METHODS The effect of the FASN inhibitor TVB-3166 on ERα expression and cell growth was characterized in tamoxifen-resistant cell lines, xenografts, and patient explants. Subcellular localization of ERα was assessed using subcellular fractionations. Palmitoylation and ubiquitination of ERα were assessed by immunoprecipitation. ERα and p-eIF2α protein levels were analyzed by Western blotting after treatment with TVB-3166 with or without the addition of palmitate or BAPTA. RESULTS TVB-3166 treatment leads to a marked inhibition of proliferation in tamoxifen-resistant cells compared to the parental cells. Additionally, TVB-3166 significantly inhibited tamoxifen-resistant breast tumor growth in mice and decreased proliferation of primary tumor explants compared to untreated controls. FASN inhibition significantly reduced ERα levels most prominently in endocrine-resistant cells and altered its subcellular localization. Furthermore, we showed that the reduction of ERα expression upon TVB-3166 treatment is mediated through the induction of endoplasmic reticulum stress. CONCLUSION Our preclinical data provide evidence that FASN inhibition by TVB-3166 presents a promising therapeutic strategy for the treatment of endocrine-resistant breast cancer. Further clinical development of FASN inhibitors for endocrine-resistant breast cancer should be considered.
Collapse
Affiliation(s)
| | | | | | | | - Victoria Alers
- UT Health San Antonio MD Anderson Cancer Center, San Antonio, TX, USA
| | - Gangadhara R Sareddy
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Tim Huang
- UT Health San Antonio MD Anderson Cancer Center, San Antonio, TX, USA
| | - Michael Garcia
- UT Health San Antonio MD Anderson Cancer Center, San Antonio, TX, USA
| | | | - Ratna K Vadlamudi
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Andrew J Brenner
- UT Health San Antonio MD Anderson Cancer Center, San Antonio, TX, USA.
- South Texas Research Facility, University of Texas Health San Antonio, STRF 2.208.58403 Floyd Curl Dr, San Antonio, TX, 78229, USA.
| |
Collapse
|
12
|
Kashyap D, Garg VK, Sandberg EN, Goel N, Bishayee A. Oncogenic and Tumor Suppressive Components of the Cell Cycle in Breast Cancer Progression and Prognosis. Pharmaceutics 2021; 13:pharmaceutics13040569. [PMID: 33920506 PMCID: PMC8072616 DOI: 10.3390/pharmaceutics13040569] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/03/2021] [Accepted: 04/13/2021] [Indexed: 02/08/2023] Open
Abstract
Cancer, a disease of inappropriate cell proliferation, is strongly interconnected with the cell cycle. All cancers consist of an abnormal accumulation of neoplastic cells, which are propagated toward uncontrolled cell division and proliferation in response to mitogenic signals. Mitogenic stimuli include genetic and epigenetic changes in cell cycle regulatory genes and other genes which regulate the cell cycle. This suggests that multiple, distinct pathways of genetic alterations lead to cancer development. Products of both oncogenes (including cyclin-dependent kinase (CDKs) and cyclins) and tumor suppressor genes (including cyclin-dependent kinase inhibitors) regulate cell cycle machinery and promote or suppress cell cycle progression, respectively. The identification of cyclins and CDKs help to explain and understand the molecular mechanisms of cell cycle machinery. During breast cancer tumorigenesis, cyclins A, B, C, D1, and E; cyclin-dependent kinase (CDKs); and CDK-inhibitor proteins p16, p21, p27, and p53 are known to play significant roles in cell cycle control and are tightly regulated in normal breast epithelial cells. Following mitogenic stimuli, these components are deregulated, which promotes neoplastic transformation of breast epithelial cells. Multiple studies implicate the roles of both types of components-oncogenic CDKs and cyclins, along with tumor-suppressing cyclin-dependent inhibitors-in breast cancer initiation and progression. Numerous clinical studies have confirmed that there is a prognostic significance for screening for these described components, regarding patient outcomes and their responses to therapy. The aim of this review article is to summarize the roles of oncogenic and tumor-suppressive components of the cell cycle in breast cancer progression and prognosis.
Collapse
Affiliation(s)
- Dharambir Kashyap
- Department of Histopathology, Postgraduate Institute of Medical Education and Research, Chandigarh 160 012, Punjab, India;
| | | | - Elise N. Sandberg
- Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA;
| | - Neelam Goel
- University Institute of Engineering and Technology, Panjab University, Chandigarh 160 014, Punjab, India
- Correspondence: (N.G.); or (A.B.)
| | - Anupam Bishayee
- Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA;
- Correspondence: (N.G.); or (A.B.)
| |
Collapse
|
13
|
Huang C, Xia X, He J, Liu Y, Shao Z, Hu T, Yu C, Liu X, Xu Q, Liu B, Liu N, Liao Y, Huang H. ERα is a target for butein-induced growth suppression in breast cancer. Am J Cancer Res 2020; 10:3721-3736. [PMID: 33294263 PMCID: PMC7716169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/23/2020] [Indexed: 06/12/2023] Open
Abstract
Breast cancer (BCa) has the highest incidence and mortality among malignant diseases in female worldwide. BCa is frequently caused by estrogen receptor α (ERα), a ligand-dependent receptor that highly expressed in about 70% of breast tumors. Therefore, ERα has become a well-characterized and the most effective target for treating ERα-expressing BCa (ERα+ BCa). However, the acquire resistance was somehow developed in patients who received current ERα signaling-targeted endocrine therapies. Hence, discovery of novel anti-estrogen/ERα strategies is urgent. In the present study, we identified butein as a potential agent for breast cancer treatment by the use of a natural product library. We showed that butein inhibits the growth of ERα+ BCa both in vitro and in vivo which is associated with cell cycle arrest that partially triggered by butein-induced ERα downregulation. Mechanically, butein binds to a specific pocket of ERα and promotes proteasome-mediated degradation of the receptor. Collectively, this work reveals that butein is a candidate to diminish ERα signaling which represents a potentially novel strategy for treating BCa.
Collapse
Affiliation(s)
- Chuyi Huang
- Affiliated Cancer Hospital & Institute of Guangzhou Medical UniversityGuangzhou 510095, Guangdong, China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical UniversityGuangzhou 511436, Guangdong, China
| | - Xiaohong Xia
- Affiliated Cancer Hospital & Institute of Guangzhou Medical UniversityGuangzhou 510095, Guangdong, China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical UniversityGuangzhou 511436, Guangdong, China
| | - Jinchan He
- Affiliated Cancer Hospital & Institute of Guangzhou Medical UniversityGuangzhou 510095, Guangdong, China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical UniversityGuangzhou 511436, Guangdong, China
| | - Yuan Liu
- Affiliated Cancer Hospital & Institute of Guangzhou Medical UniversityGuangzhou 510095, Guangdong, China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical UniversityGuangzhou 511436, Guangdong, China
| | - Zhenlong Shao
- Affiliated Cancer Hospital & Institute of Guangzhou Medical UniversityGuangzhou 510095, Guangdong, China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical UniversityGuangzhou 511436, Guangdong, China
| | - Tumei Hu
- Affiliated Cancer Hospital & Institute of Guangzhou Medical UniversityGuangzhou 510095, Guangdong, China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical UniversityGuangzhou 511436, Guangdong, China
| | - Cuifu Yu
- Affiliated Cancer Hospital & Institute of Guangzhou Medical UniversityGuangzhou 510095, Guangdong, China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical UniversityGuangzhou 511436, Guangdong, China
| | - Xiaolin Liu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical UniversityGuangzhou 510260, Guangdong, China
| | - Qiong Xu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical UniversityGuangzhou 510260, Guangdong, China
| | - Bin Liu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical UniversityGuangzhou 510260, Guangdong, China
| | - Ningning Liu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical UniversityGuangzhou 510260, Guangdong, China
| | - Yuning Liao
- Affiliated Cancer Hospital & Institute of Guangzhou Medical UniversityGuangzhou 510095, Guangdong, China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical UniversityGuangzhou 511436, Guangdong, China
| | - Hongbiao Huang
- Affiliated Cancer Hospital & Institute of Guangzhou Medical UniversityGuangzhou 510095, Guangdong, China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical UniversityGuangzhou 511436, Guangdong, China
| |
Collapse
|
14
|
Alarcón R, Rivera OE, Ingaramo PI, Tschopp MV, Dioguardi GH, Milesi MM, Muñoz-de-Toro M, Luque EH. Neonatal exposure to a glyphosate-based herbicide alters the uterine differentiation of prepubertal ewe lambs. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 265:114874. [PMID: 32599332 DOI: 10.1016/j.envpol.2020.114874] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/30/2020] [Accepted: 05/23/2020] [Indexed: 06/11/2023]
Abstract
The exposure to endocrine-disrupting compounds (EDCs), such as glyphosate-based herbicides (GBHs), during early life might alter female fertility. The aim of the present study was to evaluate the effects of neonatal exposure to a GBH on sheep uterine development. To achieve this, Friesian ewe lambs were exposed to GBH (2 mg/kg of body weight/day; n = 12) or vehicle (controls; n = 10) through s.c. injections, from postnatal day (PND) 1 to PND14; on PND45, the uteri were obtained to evaluate histomorphological and molecular parameters. Morphological parameters were determined by picrosirius-hematoxylin staining. Protein expression of Ki67 (as a cell proliferation marker), p27, and molecules involved in uterine organogenetic differentiation was measured by immunohistochemistry. We also determined the mRNA expression of the IGF molecular pathway by RT-PCR. Although histomorphology was not modified, the uteri of GBH-exposed ewe lambs showed lower cell proliferation, together with higher p27 protein expression. In addition, the uteri of GBH-exposed ewe lambs showed increased gene expression of insulin-like growth factor binding protein 3 (IGFBP-3), decreased expression of ERα in the luminal (LE) and glandular (GE) epithelia and in the subepithelial stroma (SS), and lower PR expression in the LE but higher in the GE and SS. In addition, GBH treatment decreased the uterine expression of Wnt5a in the GE, of Wnt7a in the SS, of β-catenin in the LE and GE, of Hoxa10 in the SS, and of Foxa2 in the GE as compared with controls. In conclusion, neonatal exposure to GBH decreased cell proliferation and altered the expression of molecules that control proliferation and development in the uterus. All these changes might have adverse consequences on uterine differentiation and functionality, affecting the female reproductive health of sheep. GBH may be responsible for uterine subfertility, acting as an EDC.
Collapse
Affiliation(s)
- Ramiro Alarcón
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Oscar E Rivera
- Instituto de Investigación sobre Producción Agropecuaria, Ambiente y Salud (IIPAAS), Facultad de Ciencias Agrarias, Universidad Nacional de Lomas de Zamora, Buenos Aires, Argentina
| | - Paola I Ingaramo
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional del Litoral, Santa Fe, Argentina
| | - María V Tschopp
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Gisela H Dioguardi
- Instituto de Investigación sobre Producción Agropecuaria, Ambiente y Salud (IIPAAS), Facultad de Ciencias Agrarias, Universidad Nacional de Lomas de Zamora, Buenos Aires, Argentina
| | - Mercedes M Milesi
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Mónica Muñoz-de-Toro
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Enrique H Luque
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional del Litoral, Santa Fe, Argentina.
| |
Collapse
|
15
|
Xia X, He J, Liu B, Shao Z, Xu Q, Hu T, Yu C, Liu X, Liao Y, Liu N, Huang H. Targeting ERα degradation by L-Tetrahydropalmatine provides a novel strategy for breast cancer treatment. Int J Biol Sci 2020; 16:2192-2204. [PMID: 32549765 PMCID: PMC7294940 DOI: 10.7150/ijbs.44005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 05/02/2020] [Indexed: 02/07/2023] Open
Abstract
The incidence and mortality of breast cancer (BCa) are the highest among female cancers. There are approximate 70% BCa that are classified as estrogen receptor alpha (ERα) positive. Therefore, targeting ERα is the most significantly therapeutic schedule. However, patients with breast cancer develop resistance to ERα or estrogen (E2) antagonists such as fulvestrant and tamoxifen. In the present study, we found that L-Tetrahydropalmatine (L-THP) significantly suppressed cell proliferation in ERα+ BCa cells via inducing cell cycle arrest rather than apoptosis. Additionally, L-THP enhanced the sensitivity of ERα+ BCa cells to tamoxifen and fulvestrant. Mechanically, the application of L-THP promotes ERα degradation through accumulating ubiquitin chains on ERα. Overexpressing ERα abrogates L-THP induced-antiproliferation in ERα+ BCa cells. Collectively, our work indicates that L-THP may represent a potentially novel therapeutic medicine for ERα+ breast cancer patient.
Collapse
Affiliation(s)
- Xiaohong Xia
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510260, China.,Affiliated Cancer Hospital & institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou, Guangdong 511436, China
| | - Jinchan He
- Affiliated Cancer Hospital & institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou, Guangdong 511436, China
| | - Bin Liu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510260, China
| | - Zhenlong Shao
- Affiliated Cancer Hospital & institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou, Guangdong 511436, China
| | - Qiong Xu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510260, China
| | - Tumei Hu
- Affiliated Cancer Hospital & institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou, Guangdong 511436, China
| | - Cuifu Yu
- Affiliated Cancer Hospital & institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou, Guangdong 511436, China
| | - Xiaolin Liu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510260, China
| | - Yuning Liao
- Affiliated Cancer Hospital & institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou, Guangdong 511436, China
| | - Ningning Liu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510260, China
| | - Hongbiao Huang
- Affiliated Cancer Hospital & institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou, Guangdong 511436, China
| |
Collapse
|
16
|
Ferraiuolo RM, Wagner KU. Regulation and New Treatment Strategies in Breast Cancer. JOURNAL OF LIFE SCIENCES (WESTLAKE VILLAGE, CALIF.) 2019; 1:23-38. [PMID: 32095785 PMCID: PMC7039658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Breast cancer classifications are based on the presence or absence of estrogen receptor and progesterone receptor along with the overexpression or amplification of the Her2 receptor. Although the overall 5-year survival rate of breast cancer patients has increased due to the use of targeted therapies, a subset of patients can acquire resistance over time or are unresponsive when presented in the clinic. Novel therapies focusing on molecular pathways and cell cycle regulation currently being used in the clinic may lead to increased response in this subset of patients.
Collapse
Affiliation(s)
- Rosa-Maria Ferraiuolo
- Karmanos Cancer Institute at Wayne State University
School of Medicine, Detroit, MI 48202
| | - Kay-Uwe Wagner
- Karmanos Cancer Institute at Wayne State University
School of Medicine, Detroit, MI 48202
| |
Collapse
|
17
|
Deubiquitination and stabilization of estrogen receptor α by ubiquitin-specific protease 7 promotes breast tumorigenesis. Cancer Lett 2019; 465:118-128. [DOI: 10.1016/j.canlet.2019.09.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 09/03/2019] [Accepted: 09/06/2019] [Indexed: 01/19/2023]
|
18
|
Farghadani R, Seifaddinipour M, Rajarajeswaran J, Abdulla MA, Mohd Hashim NB, Khaing SL, Salehen NB. In vivo acute toxicity evaluation and in vitro molecular mechanism study of antiproliferative activity of a novel indole Schiff base β-diiminato manganese III complex in hormone-dependent and triple negative breast cancer cells. PeerJ 2019; 7:e7686. [PMID: 31608167 PMCID: PMC6786247 DOI: 10.7717/peerj.7686] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 08/19/2019] [Indexed: 12/28/2022] Open
Abstract
Breast cancer is the most frequently diagnosed cancer among women worldwide. Recently, increasing attention has been paid to the anticancer effects of transition metal complexes of indole Schiff bases. β-diiminato ManganeseIII complex has shown promising cell cycle arrest and apoptosis induction against MCF-7 and MDA-MB-231 breast cancer cells. In this study, time- and dose- dependent inhibitory activity were evaluated using MTT assay after 48 h and 72 h exposure time. In addition, median effect analysis was conducted according to Chou-Talalay method to investigate whether MnIII complex has synergistic effect in combination with chemotherapeutic drugs on inhibiting breast cancer cell growth. The molecular mechanisms underlying its potent antiproliferative effect was determined through bioluminescent caspase-3/7, -8 and -9 activity assays and quantitative expression analysis of cell cycle- and apoptosis-related genes. Furthermore, safety evaluation of MnIII complex was assessed through the acute oral toxicity test in in vivo model. The MTT assay results revealed that it potently reduced the viability of MCF-7 (IC50 of 0.63 ± 0.07 µg/mL for 48 h and 0.39 ± 0.08 µg/mL for 72 h) and MDA-MB-231 (1.17 ± 0.06 µg/mL for 48 h, 1.03 ± 0.15 µg/mL for 72 h) cells in dose- and time-dependent manner. Combination treatment also enhanced the cytotoxic effects of doxorubicin but not tamoxifen on inhibiting breast cancer cell growth. The involvement of intrinsic and extrinsic pathway in apoptosis induction was exhibited through the increased activity of caspase-9 and caspase-8, respectively, leading to enhanced downstream executioner caspase-3/7 activity in treated MCF-7 and MDA-MB-231 cells. In addition, gene expression analysis revealed that MnIII complex exerts its antiproliferative effect via up-and down-regulation of p21 and cyclin D1, respectively, along with increased expression of Bax/Bcl-2 ratio, TNF-α, initiator caspase-8 and -10 and effector caspase-3 in MCF-7 and MDA-MB-231 cells. However, the results did not show increased caspase-8 activity in treated MCF-7 cells. Furthermore, in vivo acute oral toxicity test revealed no signs of toxicity and mortality in treated animal models compared to the control group. Collectively, the promising inhibitory effect and molecular and mechanistic evidence of antiproliferative activity of MnIII complex and its safety characterization have demonstrated that it may have therapeutic value in breast cancer treatment worthy of further investigation and development.
Collapse
Affiliation(s)
- Reyhaneh Farghadani
- Department of Molecular Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Maryam Seifaddinipour
- Institute of Biological Sciences, Faculty of Science, University of Malaya, Kuala Lumpur, Malaysia
| | - Jayakumar Rajarajeswaran
- Department of Molecular Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Mahmood Ameen Abdulla
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | | | - Si Lay Khaing
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Nur'ain Binti Salehen
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
19
|
Xue M, Zhang K, Mu K, Xu J, Yang H, Liu Y, Wang B, Wang Z, Li Z, Kong Q, Li X, Wang H, Zhu J, Zhuang T. Regulation of estrogen signaling and breast cancer proliferation by an ubiquitin ligase TRIM56. Oncogenesis 2019; 8:30. [PMID: 31000690 PMCID: PMC6473003 DOI: 10.1038/s41389-019-0139-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 03/27/2019] [Accepted: 03/29/2019] [Indexed: 11/13/2022] Open
Abstract
Breast cancer ranks no. 1 in women cancer worldwide, while 60–70% are estrogen receptor alpha positive. The estrogen selective modulators, such as tamoxifen, become the effective drugs for controlling ER alpha breast cancer progression. However, tamoxifen resistance will develop during long-time treatment and cancer progression. Thus, further understanding of ER alpha signaling becomes necessary for the improvement of breast cancer therapy. Here, we identify TRIM56 as a novel regulatory factor in ER alpha signaling. TRIM56 expression is positively correlated with ER alpha and PR in breast cancer samples and is related to poor prognosis in endocrine therapy patients. TRIM56 depletion significantly decreases ER alpha signaling activity and ER-alpha-positive breast cancer proliferation in vitro and in vivo. TRIM56 associates with AF1 domain of ER alpha via its WD40 domain in the cytoplasm. TRIM56 prolongs ER alpha protein stability, possibly through targeting ER alpha K63-linked ubiquitination. In conclusion, our study reveals an interesting posttranslational mechanism between TRIM56 and ER alpha in breast cancer progression. Targeting TRIM56 could be a promising approach for ER-alpha-positive breast cancer.
Collapse
Affiliation(s)
- Min Xue
- Henan Key Laboratory of Immunology and Targeted Therapy, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, 453003, Xinxiang, Henan, P.R. China
| | - Kai Zhang
- Department of Breast Surgery, Qilu Hospital of Shandong University, 107 West Wenhua Road, 250012, Jinan, Shandong, P.R. China
| | - Kun Mu
- Department of Pathology, School of Basic Medical Sciences, Shandong University, 250012, Jinan, Shandong, P.R. China
| | - Juntao Xu
- Rhil Rivers Technology (Beijing) Ltd, Beijing, P.R. China.,Department of Cancer Genomics, LemonData Biotech (Shenzhen), Shenzhen, P.R. China
| | - Huijie Yang
- Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, 22 Qixiangtai Road, Heping District, 300070, Tianjin, P.R. China
| | - Yun Liu
- Henan Key Laboratory of Immunology and Targeted Therapy, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, 453003, Xinxiang, Henan, P.R. China
| | - Beibei Wang
- Henan Key Laboratory of Immunology and Targeted Therapy, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, 453003, Xinxiang, Henan, P.R. China
| | - Zhonghao Wang
- School of Stomatology, Xinxiang Medical University, 453003, Xinxiang, Henan, P.R. China
| | - Zhongbo Li
- Henan Key Laboratory of Immunology and Targeted Therapy, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, 453003, Xinxiang, Henan, P.R. China
| | - Qiong Kong
- School of International Education, Xinxiang Medical University, 453003, Xinxiang, Henan, P.R. China
| | - Xiumin Li
- Department of Gastroenterology, The Third Affiliated Hospital of Xinxiang Medical University, 453003, Xinxiang, Henan, P.R. China
| | - Hui Wang
- Henan Key Laboratory of Immunology and Targeted Therapy, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, 453003, Xinxiang, Henan, P.R. China.
| | - Jian Zhu
- Henan Key Laboratory of Immunology and Targeted Therapy, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, 453003, Xinxiang, Henan, P.R. China. .,Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| | - Ting Zhuang
- Henan Key Laboratory of Immunology and Targeted Therapy, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, 453003, Xinxiang, Henan, P.R. China.
| |
Collapse
|
20
|
Du Y, Song W, Chen J, Chen H, Xuan Z, Zhao L, Chen J, Jin C, Zhou M, Tuo B, Zhao Y, Zheng S, Song P. The potassium channel KCa3.1 promotes cell proliferation by activating SKP2 and metastasis through the EMT pathway in hepatocellular carcinoma. Int J Cancer 2019; 145:503-516. [PMID: 30628729 DOI: 10.1002/ijc.32121] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Revised: 11/17/2018] [Accepted: 12/19/2018] [Indexed: 12/19/2022]
Abstract
The intermediate conductance calcium-activated potassium channel (KCa3.1) plays an important role in maintaining intracellular calcium homeostasis and is involved in the tumorigenesis of many human cancers. However, it is unknown whether KCa3.1 plays a role in the genesis of hepatocellular carcinoma (HCC), one of the most common malignant tumors worldwide with a very poor prognosis. In our study, we found that the expression of KCa3.1 was significantly elevated in poorly differentiated HCC tissues compared to adjacent noncancerous tissues. In vitro and in vivo experiments showed that KCa3.1 could promote cell proliferation, migration, and invasion of HCC. Mechanistically, KCa3.1 promoted cell cycle progression and migration and invasion of HCC cells by activating S-phase protein kinase 2 (SKP2) to trigger the degradation of p21 and p27 and targeting Reelin (RELN) to induce epithelial-mesenchymal transition (EMT), respectively. Taken together, our results demonstrate that KCa3.1 plays an important role in the genesis and progression of HCC, implying that it might be a promising therapeutic target in HCC.
Collapse
Affiliation(s)
- Yehui Du
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University.,NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China.,Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou, China
| | - Wenfeng Song
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University.,NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China.,Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou, China
| | - Jian Chen
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University.,NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China.,Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou, China
| | - Hao Chen
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University.,NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China.,Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou, China
| | - Zefeng Xuan
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University.,NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China.,Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou, China
| | - Long Zhao
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University.,NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China.,Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou, China
| | - Jun Chen
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University.,NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China.,Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou, China
| | - Cheng Jin
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University.,NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China.,Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou, China
| | - Mengqiao Zhou
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University.,NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China.,Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou, China
| | - Biguang Tuo
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical College, Zunyi, China
| | - Yongchao Zhao
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University.,NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University.,NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China.,Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou, China
| | - Penghong Song
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University.,NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China.,Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou, China
| |
Collapse
|
21
|
Simpkins F, Jang K, Yoon H, Hew KE, Kim M, Azzam DJ, Sun J, Zhao D, Ince TA, Liu W, Guo W, Wei Z, Zhang G, Mills GB, Slingerland JM. Dual Src and MEK Inhibition Decreases Ovarian Cancer Growth and Targets Tumor Initiating Stem-Like Cells. Clin Cancer Res 2018; 24:4874-4886. [PMID: 29959144 PMCID: PMC6557165 DOI: 10.1158/1078-0432.ccr-17-3697] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 04/06/2018] [Accepted: 06/28/2018] [Indexed: 01/06/2023]
Abstract
Purpose: Rational targeted therapies are needed for treatment of ovarian cancers. Signaling kinases Src and MAPK are activated in high-grade serous ovarian cancer (HGSOC). Here, we tested the frequency of activation of both kinases in HGSOC and the therapeutic potential of dual kinase inhibition.Experimental Design: MEK and Src activation was assayed in primary HGSOC from The Cancer Genome Atlas (TGGA). Effects of dual kinase inhibition were assayed on cell-cycle, apoptosis, gene, and proteomic analysis; cancer stem cells; and xenografts.Results: Both Src and MAPK are coactivated in 31% of HGSOC, and this associates with worse overall survival on multivariate analysis. Frequent dual kinase activation in HGSOC led us to assay the efficacy of combined Src and MEK inhibition. Treatment of established lines and primary ovarian cancer cultures with Src and MEK inhibitors saracatinib and selumetinib, respectively, showed target kinase inhibition and synergistic induction of apoptosis and cell-cycle arrest in vitro, and tumor inhibition in xenografts. Gene expression and proteomic analysis confirmed cell-cycle inhibition and autophagy. Dual therapy also potently inhibited tumor-initiating cells. Src and MAPK were both activated in tumor-initiating populations. Combination treatment followed by drug washout decreased sphere formation and ALDH1+ cells. In vivo, tumors dissociated after dual therapy showed a marked decrease in ALDH1 staining, sphere formation, and loss of tumor-initiating cells upon serial xenografting.Conclusions: Selumetinib added to saracatinib overcomes EGFR/HER2/ERBB2-mediated bypass activation of MEK/MAPK observed with saracatinib alone and targets tumor-initiating ovarian cancer populations, supporting further evaluation of combined Src-MEK inhibition in clinical trials. Clin Cancer Res; 24(19); 4874-86. ©2018 AACR.
Collapse
Affiliation(s)
- Fiona Simpkins
- Braman Family Breast Cancer Institute at Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida.
- Department of Obstetrics & Gynecology, University of Miami, Miami, Florida
- Ovarian Cancer Research Center, Division of Gynecology Oncology, Department of Obstetrics & Gynecology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kibeom Jang
- Braman Family Breast Cancer Institute at Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
- Department of Biochemistry and Molecular Biology, University of Miami, Miami, Florida
| | - Hyunho Yoon
- Braman Family Breast Cancer Institute at Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| | - Karina E Hew
- Braman Family Breast Cancer Institute at Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
- Department of Obstetrics & Gynecology, University of Miami, Miami, Florida
| | - Minsoon Kim
- Braman Family Breast Cancer Institute at Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
- Department of Biochemistry and Molecular Biology, University of Miami, Miami, Florida
| | - Diana J Azzam
- Braman Family Breast Cancer Institute at Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
- Department of Biochemistry and Molecular Biology, University of Miami, Miami, Florida
| | - Jun Sun
- Braman Family Breast Cancer Institute at Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| | - Dekuang Zhao
- Braman Family Breast Cancer Institute at Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| | - Tan A Ince
- Braman Family Breast Cancer Institute at Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
- Department of Pathology and Laboratory Medicine, University of Miami, Miami, Florida
- Interdisciplinary Stem Cell Institute, University of Miami, Miami, Florida
| | - Wenbin Liu
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Wei Guo
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Zhi Wei
- Department of Computer Science, New Jersey Institute of Technology, Newark, New Jersey
| | - Gao Zhang
- Wistar Institute, Philadelphia, Pennsylvania
| | - Gordon B Mills
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Joyce M Slingerland
- Braman Family Breast Cancer Institute at Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida.
- Department of Biochemistry and Molecular Biology, University of Miami, Miami, Florida
- Department of Medicine, University of Miami, Miami, Florida
| |
Collapse
|
22
|
A dual role of miR-22 modulated by RelA/p65 in resensitizing fulvestrant-resistant breast cancer cells to fulvestrant by targeting FOXP1 and HDAC4 and constitutive acetylation of p53 at Lys382. Oncogenesis 2018; 7:54. [PMID: 30057418 PMCID: PMC6064715 DOI: 10.1038/s41389-018-0063-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 05/21/2018] [Indexed: 12/11/2022] Open
Abstract
Antiestrogen resistance is a major challenge encountered during the treatment of estrogen receptor alpha positive (ERα+) breast cancer. A better understanding of signaling pathways and downstream transcription factors and their targets may identify key molecules that can overcome antiestrogen resistance in breast cancer. An aberrant expression of miR-22 has been demonstrated in breast cancer; however, its contribution to breast cancer resistance to fulvestrant, an antiestrogen drug, remains unknown. In this study, we demonstrated a moderate elevation in miR-22 expression in the 182R-6 fulvestrant-resistant breast cancer line we used as a model system, and this elevation was positively correlated with the expression of the miRNA biogenesis enzymes AGO2 and Dicer. The level of phosphorylated HER2/neu at Tyr877 was also upregulated in these cells, whereas the level of RelA/p65 phosphorylated at Ser536 (p-p65) was downregulated. Knockdown of HER2/neu led to an induction of p-p65 and a reduction in miR-22 levels. Luciferase assays identified two NF-κB binding motifs in the miR-22 promoter that contributed to transcriptional repression of miR-22. Activation of RelA/p65, triggered by LPS, attenuated miR-22 expression, but this expression was restored by sc-514, a selective IKKβ inhibitor. Inhibition of miR-22 suppressed cell proliferation, induced apoptosis and caused cell cycle S-phase arrest, whereas enhancing expression of p21Cip1/Waf1 and p27Kip1. Surprisingly, ectopic expression of miR-22 also suppressed cell proliferation, induced apoptosis, caused S-phase arrest, and promoted the expression of p21Cip1/Waf1 and p27Kip1. Ectopic overexpression of miR-22 repressed the expression of FOXP1 and HDAC4, leading to a marked induction of acetylation of HDAC4 target histones. Conversely, inhibition of miR-22 promoted the expression of both FOXP1 and HDAC4, without the expected attenuation of histone acetylation. Instead, p53 acetylation at lysine 382 was unexpectedly upregulated. Taken together, our findings demonstrated, for the first time, that HER2 activation dephosphorylates RelA/p65 at Ser536. This dephosphoryalted p65 may be pivotal in transactivation of miR-22. Both increased and decreased miR-22 expression cause resensitization of fulvestrant-resistant breast cancer cells to fulvestrant. HER2/NF-κB (p65)/miR-22/HDAC4/p21 and HER2/NF-κB (p65)/miR-22/Ac-p53/p21 signaling circuits may therefore confer this dual role on miR-22 through constitutive transactivation of p21.
Collapse
|
23
|
Liu Z, Zhang J, Xu J, Yang H, Li X, Hou Y, Zhao Y, Xue M, Wang B, Yu N, Yu S, Niu G, Wu G, Li X, Wang H, Zhu J, Zhuang T. RNF168 facilitates oestrogen receptor ɑ transcription and drives breast cancer proliferation. J Cell Mol Med 2018; 22:4161-4170. [PMID: 29974997 PMCID: PMC6111850 DOI: 10.1111/jcmm.13694] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 04/23/2018] [Indexed: 01/13/2023] Open
Abstract
Oestrogen receptor ɑ (ERɑ) is overexpressed in two-thirds of all breast cancers and involves in development and breast cancer progression. Although ERɑ-positive breast cancer could be effective treated by endocrine therapy, the endocrine resistance is still an urgent clinical problem. Thus, further understanding of the underlying mechanisms ERɑ signalling is critical in dealing with endocrine resistance in breast cancer patients. MCF-7 and T47D breast cancer cell lines are used to carry out the molecular biological experiments. Western blot is used to assess the relative protein level of ERɑ, RNF168 and actin. Real-time PCR is used the measure the relative ERɑ-related gene mRNA level. Luciferase assay is used to measure the relative ERɑ signalling activity. Chromatin immunoprecipitation is used to measure the RNF168 binding affinity to ERɑ promoter regions. WST assay and flow cytometry are used to measure the cell proliferation capacity. We use Student's t test and one-way ANOVA test for statistical data analysis. Here, we report an important role in ERɑ-positive breast cancer cells for RNF168 protein in supporting cell proliferation by driving the transcription of ERɑ. RNF168 is highly expressed in breast cancer samples, compared with normal breast tissue. In patients with breast cancer, RNF168 expression level is correlated with poor endocrine treatment outcome. Depletion of RNF168 causes decreased cell proliferation in MCF-7 and T47D cells. Besides, depletion RNF168 reduced mRNA level of ERɑ and its target genes, such as PS2 and GREB1. Chromatin immunoprecipitation revealed that ERɑ transcription is associated with RNF168 recruitment to ERɑ promoter region, suggesting that transcriptional regulation is one mechanism by which RNF168 regulates ERɑ mRNA level and ERɑ signalling in breast cancer cells. RNF168 is required for ERɑ-positive breast cancer cell proliferation and facilitate ERɑ signalling activity possibly through promoting transcription of ERɑ.
Collapse
Affiliation(s)
- Zhenhua Liu
- Laboratory of Molecular Oncology, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of laboratory Medicine, Xinxiang Medical University, Xinxiang, China.,Henan Key Laboratory of immunology and targeted therapy, Xinxiang Medical University, Xinxiang, China.,Institute of Lung and Molecular Therapy (ILMT), Xinxiang Medical University, Xinxiang, China.,Synthetic Biology Engineering Lab of Henan Province, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, China
| | - Jinghang Zhang
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Juntao Xu
- Rhil Rivers Technology (Beijing) Ltd., Beijing, China.,Department of Cancer Genomics, LemonData Biotech (Shenzhen), Shenzhen, China
| | - Huijie Yang
- Laboratory of Molecular Oncology, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of laboratory Medicine, Xinxiang Medical University, Xinxiang, China.,Henan Key Laboratory of immunology and targeted therapy, Xinxiang Medical University, Xinxiang, China.,Institute of Lung and Molecular Therapy (ILMT), Xinxiang Medical University, Xinxiang, China
| | - Xin Li
- Laboratory of Molecular Oncology, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of laboratory Medicine, Xinxiang Medical University, Xinxiang, China.,Henan Key Laboratory of immunology and targeted therapy, Xinxiang Medical University, Xinxiang, China.,Institute of Lung and Molecular Therapy (ILMT), Xinxiang Medical University, Xinxiang, China
| | - Yingxiang Hou
- Laboratory of Molecular Oncology, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of laboratory Medicine, Xinxiang Medical University, Xinxiang, China.,Henan Key Laboratory of immunology and targeted therapy, Xinxiang Medical University, Xinxiang, China.,Institute of Lung and Molecular Therapy (ILMT), Xinxiang Medical University, Xinxiang, China
| | - Yan Zhao
- Laboratory of Molecular Oncology, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of laboratory Medicine, Xinxiang Medical University, Xinxiang, China.,Henan Key Laboratory of immunology and targeted therapy, Xinxiang Medical University, Xinxiang, China.,Institute of Lung and Molecular Therapy (ILMT), Xinxiang Medical University, Xinxiang, China
| | - Min Xue
- Laboratory of Molecular Oncology, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of laboratory Medicine, Xinxiang Medical University, Xinxiang, China.,Henan Key Laboratory of immunology and targeted therapy, Xinxiang Medical University, Xinxiang, China.,Institute of Lung and Molecular Therapy (ILMT), Xinxiang Medical University, Xinxiang, China
| | - Beibei Wang
- Laboratory of Molecular Oncology, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of laboratory Medicine, Xinxiang Medical University, Xinxiang, China.,Henan Key Laboratory of immunology and targeted therapy, Xinxiang Medical University, Xinxiang, China.,Institute of Lung and Molecular Therapy (ILMT), Xinxiang Medical University, Xinxiang, China
| | - Na Yu
- Institute of Lung and Molecular Therapy (ILMT), Xinxiang Medical University, Xinxiang, China.,Center for Cancer Research, Xinxiang Medical University, Xinxiang, China
| | - Sifan Yu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Renal cancer and Melanoma, Beijing Cancer Hospital and Institute, Peking University School of Oncology, Beijing, China
| | - Gang Niu
- Rhil Rivers Technology (Beijing) Ltd., Beijing, China.,Department of Cancer Genomics, LemonData Biotech (Shenzhen), Shenzhen, China
| | - Gaosong Wu
- Department of Thyroid and Breast Surgery, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Xiumin Li
- Institute of Lung and Molecular Therapy (ILMT), Xinxiang Medical University, Xinxiang, China.,Center for Cancer Research, Xinxiang Medical University, Xinxiang, China
| | - Hui Wang
- Laboratory of Molecular Oncology, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of laboratory Medicine, Xinxiang Medical University, Xinxiang, China.,Henan Key Laboratory of immunology and targeted therapy, Xinxiang Medical University, Xinxiang, China
| | - Jian Zhu
- Laboratory of Molecular Oncology, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of laboratory Medicine, Xinxiang Medical University, Xinxiang, China.,Henan Key Laboratory of immunology and targeted therapy, Xinxiang Medical University, Xinxiang, China.,Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ting Zhuang
- Laboratory of Molecular Oncology, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of laboratory Medicine, Xinxiang Medical University, Xinxiang, China.,Henan Key Laboratory of immunology and targeted therapy, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
24
|
Schedin TB, Borges VF, Shagisultanova E. Overcoming Therapeutic Resistance of Triple Positive Breast Cancer with CDK4/6 Inhibition. Int J Breast Cancer 2018; 2018:7835095. [PMID: 30018827 PMCID: PMC6029445 DOI: 10.1155/2018/7835095] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 03/20/2018] [Accepted: 05/08/2018] [Indexed: 01/30/2023] Open
Abstract
Triple positive breast cancers overexpress both the human epidermal growth factor receptor 2 (HER2) oncogene and the hormonal receptors (HR) to estrogen and progesterone. These cancers represent a unique therapeutic challenge because of a bidirectional cross-talk between the estrogen receptor alpha (ERα) and HER2 pathways leading to tumor progression and resistance to targeted therapy. Attempts to combine standard of care HER2-targeted drugs with antihormonal agents for the treatment of HR+/HER2+ breast cancer yielded encouraging results in preclinical experiments but did improve overall survival in clinical trial. In this review, we dissect multiple mechanisms of therapeutic resistance typical of HR+/HER2+ breast cancer, summarize prior clinical trials of targeted agents, and describe novel rational drug combinations that include antihormonal agents, HER2-targeted drugs, and CDK4/6 inhibitors for treatment of the HR+/HER2+ breast cancer subtype.
Collapse
Affiliation(s)
- Troy B. Schedin
- Young Women's Breast Cancer Translational Program, University of Colorado Denver, 13001 E 17th Pl, Aurora, CO 80045, USA
| | - Virginia F. Borges
- Young Women's Breast Cancer Translational Program, University of Colorado Denver, 13001 E 17th Pl, Aurora, CO 80045, USA
| | - Elena Shagisultanova
- Young Women's Breast Cancer Translational Program, University of Colorado Denver, 13001 E 17th Pl, Aurora, CO 80045, USA
| |
Collapse
|
25
|
Park JW, Lee SH, Woo GH, Kwon HJ, Kim DY. Downregulation of TXNIP leads to high proliferative activity and estrogen-dependent cell growth in breast cancer. Biochem Biophys Res Commun 2018. [DOI: 10.1016/j.bbrc.2018.03.020] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
26
|
Chen KHE, Bustamante K, Nguyen V, Walker AM. Involvement of miR-106b in tumorigenic actions of both prolactin and estradiol. Oncotarget 2018; 8:36368-36382. [PMID: 28422740 PMCID: PMC5482661 DOI: 10.18632/oncotarget.16755] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Accepted: 03/21/2017] [Indexed: 02/07/2023] Open
Abstract
Prolactin promotes a variety of cancers by an array of different mechanisms. Here, we have investigated prolactin's inhibitory effect on expression of the cell cycle-regulating protein, p21. Using a miRNA array, we identified a number of miRNAs upregulated by prolactin treatment, but one in particular that was strongly induced by prolactin and predicted to bind to the 3′UTR of p21 mRNA, miR-106b. By creating a p21 mRNA 3′UTR-luciferase mRNA construct, we demonstrated degradation of the construct in response to prolactin in human breast, prostate and ovarian cancer cell lines. Increased expression of miR-106b replicated, and anti-miR-106b counteracted, the effects of prolactin on degradation of the 3′UTR construct, p21 mRNA levels, and cell proliferation in breast (T47D) and prostate (PC3) cancer cells. Increased expression of miR-106b also stimulated migration of the very epithelioid T47D cell line. By contrast, anti-miR-106b dramatically decreased expression of the mesenchymal markers, SNAIL-2, TWIST-2, VIMENTIN, and FIBRONECTIN. Using signaling pathway inhibitors and the 3′UTR construct, induction of miR-106b by prolactin was determined to be mediated through the MAPK/ERK and PI3K/Akt pathways and not through Jak2/Stat5 in both T47D and PC3 cells. Prolactin activation of MAPK/ERK and PI3K/Akt also activates ERα in the absence of an ERα ligand. 17β-estradiol promoted degradation of the construct in both cell lines and pre-incubation in the estrogen antagonist, Fulvestrant, blocked the ability of both prolactin and 17β-estradiol to induce the construct-degrading activity. Together, these data support a convergence of the prolactin and 17β-estradiol miR-106b-elevating signaling pathways at ERα.
Collapse
Affiliation(s)
- Kuan-Hui Ethan Chen
- Division of Biomedical Sciences, University of California, Riverside, CA 92521, USA
| | - Karissa Bustamante
- Division of Biomedical Sciences, University of California, Riverside, CA 92521, USA
| | - Vi Nguyen
- Division of Biomedical Sciences, University of California, Riverside, CA 92521, USA
| | - Ameae M Walker
- Division of Biomedical Sciences, University of California, Riverside, CA 92521, USA
| |
Collapse
|
27
|
Chow LWC, Morita S, Chow CYC, Ng WK, Toi M. Neoadjuvant palbociclib on ER+ breast cancer (N007): clinical response and EndoPredict's value. Endocr Relat Cancer 2018; 25:123-130. [PMID: 29158285 PMCID: PMC5763422 DOI: 10.1530/erc-17-0396] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Accepted: 11/20/2017] [Indexed: 12/25/2022]
Abstract
The purpose of the study was to test the efficacy of neoadjuvant palbociclib therapy and to evaluate its impact on cell cycle arrest and changes in EndoPredict (EP) scores before and after treatment. Postmenopausal women with histologically proven ER+ve, HER2-ve invasive breast cancer, 2 cm or greater, were enrolled in an open-label, single-arm study. Twenty eligible patients were given letrozole 2.5 mg per day together with palbociclib 125 mg per day for 3 out of 4 weeks in repeated cycles for 16 weeks (4 cycles) before surgery. The primary end points were clinical response rates (cRR) and preoperative endocrine prognostic index (PEPI). The secondary end points were pathologic response and gene expression testing with EP test on collected tumor samples. The following results were obtained. 17 patients showed a clinical response of 50% or more, including 8 complete responses and 9 partial responses. There was significant reduction in area (P < 0.0001) and volume (P = 0.017) of the cancer. Pathologic complete response (pCR) was achieved in one patient; all cancers were downgraded after treatment. Ki67 (P = 0.044) and EP scores (P < 0.0001) were significantly reduced after treatment. Analysis of the relative gene expression levels showed that all proliferative genes, IL6ST and RBBP8 were decreased after palbociclib treatment. 6 patients with intermediate and three patients with high PEPI risk scores were found to have low EPclin scores. All patients with high PEPI relapse risk score had high EPclin score. In conclusion, effective clinical response was demonstrated by neoadjuvant letrozole in combination with palbociclib. Compared with PEPI, EPclin might be a better parameter to estimate prognosis after neoadjuvant therapy.
Collapse
Affiliation(s)
- Louis W C Chow
- State Key Laboratory of Quality Research in Chinese MedicinesMacau University of Science and Technology, Macau SAR, Macao
- Organisation for Oncology and Translational ResearchHong Kong SAR, China
- UNIMED Medical InstituteHong Kong SAR, China
| | - Satoshi Morita
- Department of Biomedical Statistics and BioinformaticsKyoto University Graduate School of Medicine, Kyoto, Japan
| | | | - Wai-Kuen Ng
- Department of PathologyPrecious Blood Hospital, Hong Kong SAR, China
| | - Masakazu Toi
- Organisation for Oncology and Translational ResearchHong Kong SAR, China
- Breast SurgeryKyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
28
|
Menendez JA, Lupu R. Fatty acid synthase regulates estrogen receptor-α signaling in breast cancer cells. Oncogenesis 2017; 6:e299. [PMID: 28240737 PMCID: PMC5337623 DOI: 10.1038/oncsis.2017.4] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 06/29/2016] [Accepted: 07/08/2016] [Indexed: 02/06/2023] Open
Abstract
Fatty acid synthase (FASN), the key enzyme for endogenous synthesis of fatty acids, is overexpressed and hyperactivated in a biologically aggressive subset of sex steroid-related tumors, including breast carcinomas. Using pharmacological and genetic approaches, we assessed the molecular relationship between FASN signaling and estrogen receptor alpha (ERα) signaling in breast cancer. The small compound C75, a synthetic slow-binding inhibitor of FASN activity, induced a dramatic augmentation of estradiol (E2)-stimulated, ERα-driven transcription. FASN and ERα were both necessary for the synergistic activation of ERα transcriptional activity that occurred following co-exposure to C75 and E2: first, knockdown of FASN expression using RNAi (RNA interference) drastically lowered (>100 fold) the amount of E2 required for optimal activation of ERα-mediated transcriptional activity; second, FASN blockade synergistically increased E2-stimulated ERα-mediated transcriptional activity in ERα-negative breast cancer cells stably transfected with ERα, but not in ERα-negative parental cells. Non-genomic, E2-regulated cross-talk between the ERα and MAPK pathways participated in these phenomena. Thus, treatment with the pure antiestrogen ICI 182 780 or the potent and specific inhibitor of MEK/ERK, U0126, was sufficient to abolish the synergistic nature of the interaction between FASN blockade and E2-stimulated ERα transactivation. FASN inhibition suppressed E2-stimulated breast cancer cell proliferation and anchorage-independent colony formation while promoting the reduction of ERα protein. FASN blockade resulted in the increased expression and nuclear accumulation of the cyclin-dependent kinase inhibitors p21WAF1/CIP1 and p27Kip1, two critical mediators of the therapeutic effects of antiestrogen in breast cancer, while inactivating AKT, a key mediator of E2-promoted anchorage-independent growth. The ability of FASN to regulate E2/ERα signaling may represent a promising strategy for anticancer treatment involving a new generation of FASN inhibitors.
Collapse
Affiliation(s)
- J A Menendez
- Program Against Cancer Therapeutic Resistance (ProCURE), Metabolism and Cancer Group, Catalan Institute of Oncology, Girona, Catalonia, Spain.,Molecular Oncology Group, Girona Biomedical Research Institute (IDIBGI), Girona, Catalonia, Spain
| | - R Lupu
- Mayo Clinic, Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, Rochester, MN, USA.,Mayo Clinic Cancer Center, Rochester, MN, USA
| |
Collapse
|
29
|
Zhang N, Pati D. Biology and insights into the role of cohesin protease separase in human malignancies. Biol Rev Camb Philos Soc 2017; 92:2070-2083. [PMID: 28177203 DOI: 10.1111/brv.12321] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 01/05/2017] [Accepted: 01/12/2017] [Indexed: 12/11/2022]
Abstract
Separase, an enzyme that resolves sister chromatid cohesion during the metaphase-to-anaphase transition, plays a pivotal role in chromosomal segregation and cell division. Separase protein, encoded by the extra spindle pole bodies like 1 (ESPL1) gene, is overexpressed in numerous human cancers including breast, bone, brain, and prostate. Separase is oncogenic, and its overexpression is sufficient to induce mammary tumours in mice. Either acute or chronic overexpression of separase in mouse mammary glands leads to aneuploidy and tumorigenesis, and inhibition of separase enzymatic activity decreases the growth of human breast tumour xenografts in mice. This review focuses on the biology of and insights into the molecular mechanisms of separase as an oncogene, and its significance and implications for human cancers.
Collapse
Affiliation(s)
- Nenggang Zhang
- Departments of Pediatrics and Molecular and Cellular Biology, Texas Children's Cancer Center, Baylor College of Medicine, 1102 Bates St., FC1220, Houston, TX 77030, U.S.A
| | - Debananda Pati
- Departments of Pediatrics and Molecular and Cellular Biology, Texas Children's Cancer Center, Baylor College of Medicine, 1102 Bates St., FC1220, Houston, TX 77030, U.S.A
| |
Collapse
|
30
|
Chuffa LGDA, Lupi-Júnior LA, Costa AB, Amorim JPDA, Seiva FRF. The role of sex hormones and steroid receptors on female reproductive cancers. Steroids 2017; 118:93-108. [PMID: 28041951 DOI: 10.1016/j.steroids.2016.12.011] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 12/10/2016] [Accepted: 12/24/2016] [Indexed: 02/08/2023]
Abstract
Sex steroids have been widely described to be associated with a number of human diseases, including hormone-dependent tumors. Several studies have been concerned about the factors regulating the availability of sex steroids and its importance in the pathophysiological aspects of the reproductive cancers in women. In premenopausal women, large fluctuations in the concentration of circulating estradiol (E2) and progesterone (P4) orchestrate many events across the menstrual cycle. After menopause, the levels of circulating E2 and P4 decline but remain at high concentration in the peripheral tissues. Notably, there is a strong relationship between circulating sex hormones and female reproductive cancers (e.g. ovarian, breast, and endometrial cancers). These hormones activate a number of specific signaling pathways after binding either to estrogen receptors (ERs), especially ERα, ERα36, and ERβ or progesterone receptors (PRs). Importantly, the course of the disease will depend on particular transactivation pathway. Identifying ER- or PR-positive tumors will benefit patients in terms of proper endocrine therapy. Based on hormonal responsiveness, effective prevention methods for ovarian, breast, and endometrial cancers represent a special opportunity for women at risk of malignancies. Hormone replacement therapy (HRT) might significantly increase the risk of these cancer types, and endocrine treatments targeting ER signaling may be helpful against E2-dependent tumors. This review will present the role of sex steroids and their receptors associated with the risk of developing female reproductive cancers, with emphasis on E2 levels in pre and postmenopausal women. In addition, new therapeutic strategies for improving the survival rate outcomes in women will be addressed.
Collapse
Affiliation(s)
| | - Luiz Antonio Lupi-Júnior
- Department of Anatomy, IBB/UNESP, Institute of Biosciences of Botucatu, Univ. Estadual Paulista, SP, Brazil
| | - Aline Balandis Costa
- Department of Nursing, UENP/CLM - Universidade Estadual do Norte do Paraná, PR, Brazil
| | | | | |
Collapse
|
31
|
Acquired CDK6 amplification promotes breast cancer resistance to CDK4/6 inhibitors and loss of ER signaling and dependence. Oncogene 2016; 36:2255-2264. [PMID: 27748766 PMCID: PMC5393973 DOI: 10.1038/onc.2016.379] [Citation(s) in RCA: 306] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Revised: 08/24/2016] [Accepted: 08/31/2016] [Indexed: 12/24/2022]
Abstract
Dysregulated activation of the CDK4/6 kinases is a hallmark of most mammary-derived carcinomas. ATP-competitive inhibitors against this complex have been recently advanced in the clinic and have shown significant activity, particularly against tumors driven by the estrogen receptor (ER). However, resistance to these compounds has begun to emerge often months to years after their initiation. We investigated potential mechanisms of resistance using cell line models that are highly sensitive to this class of drugs. After prolonged exposure to the selective and potent CDK4/6 inhibitor LY2835219, clones emerged and several were found to harbor amplification of the CDK6 kinase. Amplification of CDK6 resulted in a marked increase in CDK6 expression and reduced response of the CDK4/6 target, phospho-Rb (pRb), to CDK4/6 inhibitors. Knockdown of CDK6 restored drug sensitivity, while enforced overexpression of CDK6 was sufficient to mediate drug resistance. Not only did CDK6 overexpression mediate resistance to CDK4/6 inhibitors but it also led to reduced expression of the ER and progesterone receptor (PR), and diminished responsiveness to ER antagonism. The reduced ER/PR expression after CDK4/6 inhibitor resistance was additionally observed in tumor biopsy specimens from patients treated with these drugs. Alternative mechanisms of resistance to CDK4/6 inhibitors such as loss of pRb and cyclin E1 overexpression also exhibited decreased hormone responsiveness, suggesting that the clinical paradigm of sequential endocrine-based therapy may be ineffective in some settings of acquired CDK4/6 resistance.
Collapse
|
32
|
Luo A, Su D, Zhang X, Qi L, Fu L, Dong JT. Estrogen-estrogen receptor signaling suppresses the transcription of ERRF in breast cancer cells. J Genet Genomics 2016; 43:565-567. [DOI: 10.1016/j.jgg.2016.06.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 04/13/2016] [Accepted: 06/20/2016] [Indexed: 10/21/2022]
|
33
|
Xu S, Huang H, Chen YN, Deng YT, Zhang B, Xiong XD, Yuan Y, Zhu Y, Huang H, Xie L, Liu X. DNA damage responsive miR-33b-3p promoted lung cancer cells survival and cisplatin resistance by targeting p21 WAF1/CIP1. Cell Cycle 2016; 15:2920-2930. [PMID: 27559850 DOI: 10.1080/15384101.2016.1224043] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Cisplatin is the most potent and widespread used chemotherapy drug for lung cancer treatment. However, the development of resistance to cisplatin is a major obstacle in clinical therapy. The principal mechanism of cisplatin is the induction of DNA damage, thus the capability of DNA damage response (DDR) is a key factor that influences the cisplatin sensitivity of cancer cells. Recent advances have demonstrated that miRNAs (microRNAs) exerted critical roles in DNA damage response; nonetheless, the association between DNA damage responsive miRNAs and cisplatin resistance and its underlying molecular mechanism still require further investigation. The present study has attempted to identify differentially expressed miRNAs in cisplatin induced DNA damage response in lung cancer cells, and probe into the effects of the misexpressed miRNAs on cisplatin sensitivity. Deep sequencing showed that miR-33b-3p was dramatically down-regulated in cisplatin-induced DNA damage response in A549 cells; and ectopic expression of miR-33b-3p endowed the lung cancer cells with enhanced survival and decreased γH2A.X expression level under cisplatin treatment. Consistently, silencing of miR-33b-3p in the cisplatin-resistant A549/DDP cells evidently sensitized the cells to cisplatin. Furthermore, we identified CDKN1A (p21) as a functional target of miR-33b-3p, a critical regulator of G1/S checkpoint, which potentially mediated the protection effects of miR-33b-3p against cisplatin. In aggregate, our results suggested that miR-33b-3p modulated the cisplatin sensitivity of cancer cells might probably through impairing the DNA damage response. And the knowledge of the drug resistance conferred by miR-33b-3p has great clinical implications for improving the efficacy of chemotherapies for treating lung cancers.
Collapse
Affiliation(s)
- Shun Xu
- a Institute of Aging Research, Guangdong Medical University , Dongguan , P.R. China.,b Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics , Dongguan , P.R. China.,c Institute of Biochemistry & Molecular Biology, Guangdong Medical University , Zhanjiang , P.R. China
| | - Haijiao Huang
- a Institute of Aging Research, Guangdong Medical University , Dongguan , P.R. China.,b Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics , Dongguan , P.R. China.,c Institute of Biochemistry & Molecular Biology, Guangdong Medical University , Zhanjiang , P.R. China
| | - Yu-Ning Chen
- a Institute of Aging Research, Guangdong Medical University , Dongguan , P.R. China.,b Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics , Dongguan , P.R. China.,c Institute of Biochemistry & Molecular Biology, Guangdong Medical University , Zhanjiang , P.R. China
| | - Yun-Ting Deng
- a Institute of Aging Research, Guangdong Medical University , Dongguan , P.R. China.,b Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics , Dongguan , P.R. China.,c Institute of Biochemistry & Molecular Biology, Guangdong Medical University , Zhanjiang , P.R. China
| | - Bing Zhang
- a Institute of Aging Research, Guangdong Medical University , Dongguan , P.R. China.,b Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics , Dongguan , P.R. China.,c Institute of Biochemistry & Molecular Biology, Guangdong Medical University , Zhanjiang , P.R. China
| | - Xing-Dong Xiong
- a Institute of Aging Research, Guangdong Medical University , Dongguan , P.R. China.,b Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics , Dongguan , P.R. China.,c Institute of Biochemistry & Molecular Biology, Guangdong Medical University , Zhanjiang , P.R. China
| | - Yuan Yuan
- a Institute of Aging Research, Guangdong Medical University , Dongguan , P.R. China.,b Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics , Dongguan , P.R. China.,c Institute of Biochemistry & Molecular Biology, Guangdong Medical University , Zhanjiang , P.R. China
| | - Yanmei Zhu
- a Institute of Aging Research, Guangdong Medical University , Dongguan , P.R. China.,b Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics , Dongguan , P.R. China.,c Institute of Biochemistry & Molecular Biology, Guangdong Medical University , Zhanjiang , P.R. China
| | - Haiyong Huang
- a Institute of Aging Research, Guangdong Medical University , Dongguan , P.R. China.,b Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics , Dongguan , P.R. China.,c Institute of Biochemistry & Molecular Biology, Guangdong Medical University , Zhanjiang , P.R. China
| | - Luoyijun Xie
- a Institute of Aging Research, Guangdong Medical University , Dongguan , P.R. China.,b Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics , Dongguan , P.R. China.,c Institute of Biochemistry & Molecular Biology, Guangdong Medical University , Zhanjiang , P.R. China
| | - Xinguang Liu
- a Institute of Aging Research, Guangdong Medical University , Dongguan , P.R. China.,b Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics , Dongguan , P.R. China.,c Institute of Biochemistry & Molecular Biology, Guangdong Medical University , Zhanjiang , P.R. China
| |
Collapse
|
34
|
The alpha-fetoprotein (AFP) third domain: a search for AFP interaction sites of cell cycle proteins. Tumour Biol 2016; 37:12697-12711. [DOI: 10.1007/s13277-016-5131-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 06/29/2016] [Indexed: 01/28/2023] Open
|
35
|
Grassi TF, da Silva GN, Bidinotto LT, Rossi BF, Quinalha MM, Kass L, Muñoz-de-Toro M, Barbisan LF. Global gene expression and morphological alterations in the mammary gland after gestational exposure to bisphenol A, genistein and indole-3-carbinol in female Sprague-Dawley offspring. Toxicol Appl Pharmacol 2016; 303:101-109. [DOI: 10.1016/j.taap.2016.05.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 05/06/2016] [Accepted: 05/07/2016] [Indexed: 12/27/2022]
|
36
|
Maynadier M, Basile I, Gallud A, Gary-Bobo M, Garcia M. Combination treatment with proteasome inhibitors and antiestrogens has a synergistic effect mediated by p21WAF1 in estrogen receptor-positive breast cancer. Oncol Rep 2016; 36:1127-34. [PMID: 27373750 DOI: 10.3892/or.2016.4873] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 05/11/2015] [Indexed: 11/06/2022] Open
Abstract
Although antiestrogens significantly improve the survival of patients with ER-positive breast cancer, therapeutic resistance remains a major limitation. The combinatorial use of antiestrogen with other therapies was proposed to increase their efficiency and more importantly, to prevent or delay the resistance phenomenon. In the present study, we addressed their combined effects with proteasome inhibitors (PIs). The effects of antiestrogens (hydroxyl-tamoxifen, raloxifen and fulvestrant) currently used in endocrine therapy were tested in combination with PIs, bortezomib or MG132, on the growth of three ER-positive breast cancer cell lines and in two cellular models of acquired antiestrogen resistance. When compared to single treatments, these combined treatments were significantly more effective in preventing the growth of the cell lines. The regulation of key cell cycle proteins, the cyclin-dependent kinase inhibitors, p21WAF1 and p27KIP1, were also studied. Bortezomib and MG132 drastically increased p21WAF1 expression through elevation of its mRNA concentration. Notably, p27KIP1 regulation was quite different from that of p21WAF1. Furthermore, the effect of bortezomib in combination with antiestrogen was evaluated on antiestrogen-resistant cell lines. The growth of two antiestrogen-resistant cell lines appeared responsive to proteasome inhibition and was strongly decreased by a combined therapy with an antiestrogen. Collectively, these findings provide new perspectives for the use of PIs in combination with endocrine therapies for breast cancer and possibly to overcome acquired hormonal resistance.
Collapse
Affiliation(s)
- Marie Maynadier
- Institut des Biomolécules Max Mousseron (IBMM), CNRS, Université Montpellier, ENSCM, Faculté de Montpellier, 34093 Montpellier Cedex 5, France
| | - Ilaria Basile
- Institut des Biomolécules Max Mousseron (IBMM), CNRS, Université Montpellier, ENSCM, Faculté de Montpellier, 34093 Montpellier Cedex 5, France
| | - Audrey Gallud
- Institut des Biomolécules Max Mousseron (IBMM), CNRS, Université Montpellier, ENSCM, Faculté de Montpellier, 34093 Montpellier Cedex 5, France
| | - Magali Gary-Bobo
- Institut des Biomolécules Max Mousseron (IBMM), CNRS, Université Montpellier, ENSCM, Faculté de Montpellier, 34093 Montpellier Cedex 5, France
| | - Marcel Garcia
- Institut des Biomolécules Max Mousseron (IBMM), CNRS, Université Montpellier, ENSCM, Faculté de Montpellier, 34093 Montpellier Cedex 5, France
| |
Collapse
|
37
|
ERRF is essential for Estrogen-Estrogen Receptor alpha signaling pathway in ER positive breast cancer cells. Biochem Biophys Res Commun 2016; 474:400-405. [PMID: 27125460 DOI: 10.1016/j.bbrc.2016.04.132] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 04/25/2016] [Indexed: 12/28/2022]
Abstract
Estrogen-Estrogen Receptor alpha (ERα) belongs to one of the most important signaling pathways controlling breast tissue development and progression of breast cancer. ERRF was recently identified as a candidate breast cancer associated protein and showed positive association with ERα status in clinical samples and cell lines. To further explore the relationship between ERRF and ERα, we studied whether ERRF plays any roles in estrogen-ERα pathway. Knockdown of ERRF in ER positive breast cancer cells T-47D and BT-474 reduced the level of p-AKT, p-MAPK, and phosphorylation of ERα at Ser 118 and Ser 167, and the transcriptional activity of ERα was inhibited as well. Further mechanism study proved ERRF to be an interacting partner of ERα. In total, these data revealed that ERRF is essential for the activity of E2-ERα pathway.
Collapse
|
38
|
Chaudhary S, Madhukrishna B, Adhya AK, Keshari S, Mishra SK. Overexpression of caspase 7 is ERα dependent to affect proliferation and cell growth in breast cancer cells by targeting p21(Cip). Oncogenesis 2016; 5:e219. [PMID: 27089142 PMCID: PMC4848833 DOI: 10.1038/oncsis.2016.12] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Revised: 12/01/2015] [Accepted: 12/17/2015] [Indexed: 12/20/2022] Open
Abstract
Caspase 7 (CASP7) expression has important function during cell cycle progression and cell growth in certain cancer cells and is also involved in the development and differentiation of dental tissues. However, the function of CASP7 in breast cancer cells is unclear. The aim of this study was to analyze the expression of CASP7 in breast carcinoma patients and determine the role of CASP7 in regulating tumorigenicity in breast cancer cells. In this study, we show that the CASP7 expression is high in breast carcinoma tissues compared with normal counterpart. The ectopic expression of CASP7 is significantly associated with ERα expression status and persistently elevated in different stages of the breast tumor grades. High level of CASP7 expression showed better prognosis in breast cancer patients with systemic endocrine therapy as observed from Kaplan–Meier analysis. S3 and S4, estrogen responsive element (ERE) in the CASP7 promoter, is important for estrogen-ERα-mediated CASP7 overexpression. Increased recruitment of p300, acetylated H3 and pol II in the ERE region of CASP7 promoter is observed after hormone stimulation. Ectopic expression of CASP7 in breast cancer cells results in cell growth and proliferation inhibition via p21Cip reduction, whereas small interfering RNA (siRNA) mediated reduction of CASP7 rescued p21Cip levels. We also show that pro- and active forms of CASP7 is located in the nucleus apart from cytoplasmic region of breast cancer cells. The proliferation and growth of breast cancer cells is significantly reduced by broad-spectrum peptide inhibitors and siRNA of CASP7. Taken together, our findings show that CASP7 is aberrantly expressed in breast cancer and contributes to cell growth and proliferation by downregulating p21Cip protein, suggesting that targeting CASP7-positive breast cancer could be one of the potential therapeutic strategies.
Collapse
Affiliation(s)
- S Chaudhary
- Cancer Biology Laboratory, Gene Function and Regulation group, Institute of Life Sciences, Nalco Square, Chandrasekharpur, Bhubaneswar, Odisha, India
| | - B Madhukrishna
- Cancer Biology Laboratory, Gene Function and Regulation group, Institute of Life Sciences, Nalco Square, Chandrasekharpur, Bhubaneswar, Odisha, India
| | - A K Adhya
- Department of Pathology, Kalinga Institute of Medical Sciences, KIIT Rd, Chandaka Industrial Estate, Patia, Bhubaneshwar, Odisha, India
| | - S Keshari
- Cancer Biology Laboratory, Gene Function and Regulation group, Institute of Life Sciences, Nalco Square, Chandrasekharpur, Bhubaneswar, Odisha, India
| | - S K Mishra
- Cancer Biology Laboratory, Gene Function and Regulation group, Institute of Life Sciences, Nalco Square, Chandrasekharpur, Bhubaneswar, Odisha, India
| |
Collapse
|
39
|
Żuryń A, Litwiniec A, Safiejko-Mroczka B, Klimaszewska-Wiśniewska A, Gagat M, Krajewski A, Gackowska L, Grzanka D. The effect of sulforaphane on the cell cycle, apoptosis and expression of cyclin D1 and p21 in the A549 non-small cell lung cancer cell line. Int J Oncol 2016; 48:2521-33. [PMID: 27035641 DOI: 10.3892/ijo.2016.3444] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 02/20/2016] [Indexed: 11/05/2022] Open
Abstract
Sulforaphane (SFN) is present in plants belonging to Cruciferae family and was first isolated from broccoli sprouts. Chemotherapeutic and anticarcinogenic properties of sulforaphane were demonstrated, however, the underlying mechanisms are not fully understood. In this study we evaluated the expression of cyclin D1 and p21 protein in SFN-treated A549 cells and correlated these results with the extent of cell death and/or cell cycle alterations, as well as determined a potential contribution of cyclin D1 to cell death. A549 cells were treated with increasing concentrations of SFN (30, 60 and 90 µM) for 24 h. Morphological and ultrastructural changes were observed using light, transmission electron microscope and videomicroscopy. Image-based cytometry was applied to evaluate the effect of SFN on apoptosis and the cell cycle. Cyclin D1 and p21 expression was determined by flow cytometry, RT-qPCR and immunofluorescence. siRNA was used to evaluate the role of cyclin D1 in the process of suforaphane-induced cell death. We found that the percentage of cyclin D1-positive cells decreased after the treatment with SFN, but at the same time mean fluorescence intensity reflecting cyclin D1 content was increased at 30 µM SFN and decreased at 60 and 90 µM SFN. Percentage of p21-positive cells increased following the treatment, with the highest increase at 60 µM SFN, at which concentration mean fluorescence intensity of this protein was also significantly increased. The 30-µM dose of SFN induced an increased G2/M phase population along with a decreased polyploid fraction of cells, which implies a functional G2/M arrest. The major mode of cell death induced by SFN was necrosis and, to a lower degree apoptosis. Transfection with cyclin D1-siRNA resulted in significantly compromised fraction of apoptotic and necrotic cells, which suggests that cyclin D1 is an important determinant of the therapeutic efficiency of SFN in the A549 cells.
Collapse
Affiliation(s)
- Agnieszka Żuryń
- Department of Histology and Embryology, Nicolaus Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, Faculty of Medicine, 85-092 Bydgoszcz, Poland
| | - Anna Litwiniec
- Plant Breeding and Acclimatization Institute - National Research Institute, Bydgoszcz Research Center, Department of Genetics and Breeding of Root Crops, Laboratory of Biotechnology, 85-090 Bydgoszcz, Poland
| | | | - Anna Klimaszewska-Wiśniewska
- Department of Histology and Embryology, Nicolaus Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, Faculty of Medicine, 85-092 Bydgoszcz, Poland
| | - Maciej Gagat
- Department of Histology and Embryology, Nicolaus Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, Faculty of Medicine, 85-092 Bydgoszcz, Poland
| | - Adrian Krajewski
- Department of Histology and Embryology, Nicolaus Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, Faculty of Medicine, 85-092 Bydgoszcz, Poland
| | - Lidia Gackowska
- Department of Immunology, Sexually Transmitted Diseases and Immunodermatology, Nicolaus Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, Faculty of Medicine, 85-094 Bydgoszcz, Poland
| | - Dariusz Grzanka
- Department of Dermatology, Sexually Transmitted Diseases and Immunodermatology, Nicolaus Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, Faculty of Medicine, 85-094 Bydgoszcz, Poland
| |
Collapse
|
40
|
Loss of DAB2IP in RCC cells enhances their growth and resistance to mTOR-targeted therapies. Oncogene 2016; 35:4663-74. [DOI: 10.1038/onc.2016.4] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Revised: 12/16/2015] [Accepted: 12/18/2015] [Indexed: 12/20/2022]
|
41
|
Finn RS, Aleshin A, Slamon DJ. Targeting the cyclin-dependent kinases (CDK) 4/6 in estrogen receptor-positive breast cancers. Breast Cancer Res 2016; 18:17. [PMID: 26857361 PMCID: PMC4746893 DOI: 10.1186/s13058-015-0661-5] [Citation(s) in RCA: 259] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 12/10/2015] [Indexed: 12/18/2022] Open
Abstract
Despite significant advances in early detection and treatment, breast cancer still remains a major cause of morbidity and mortality for women. Our understanding of the molecular heterogeneity of the disease has significantly expanded over the past decade and the role of cell cycle signaling in both breast cancer oncogenesis and anti-estrogen resistance has gained increasing attention. The mammalian cell cycle is driven by a complex interplay between cyclins and their associated cyclin-dependent kinase (CDK) partners, and dysregulation of this process is one of the hallmarks of cancer. Despite this, initial results with broadly acting CDK inhibitors were largely disappointing. However, recent preclinical and phase I/II clinical studies using a novel, oral, reversible CDK4/6 inhibitor, palbociclib (PD-0332991), have validated the role of CDK4/6 as a potential target in estrogen receptor-positive (ER+) breast cancers. This review highlights our current understanding of CDK signaling in both normal and malignant breast tissues, with special attention placed on recent clinical advances in inhibition of CDK4/6 in ER+ disease.
Collapse
Affiliation(s)
- Richard S Finn
- Geffen School of Medicine at UCLA, Department of Medicine, Division of Hematology Oncology, 2825 Santa Monica Blvd, Santa Monica, CA, 90404, USA.
| | - Alexey Aleshin
- Geffen School of Medicine at UCLA, Department of Medicine, Division of Hematology Oncology, 2825 Santa Monica Blvd, Santa Monica, CA, 90404, USA
| | - Dennis J Slamon
- Stanford School of Medicine, 291 Campus Drive, Stanford, CA, 94305, USA
| |
Collapse
|
42
|
Hew KE, Miller PC, El-Ashry D, Sun J, Besser AH, Ince TA, Gu M, Wei Z, Zhang G, Brafford P, Gao W, Lu Y, Mills GB, Slingerland JM, Simpkins F. MAPK Activation Predicts Poor Outcome and the MEK Inhibitor, Selumetinib, Reverses Antiestrogen Resistance in ER-Positive High-Grade Serous Ovarian Cancer. Clin Cancer Res 2015; 22:935-47. [PMID: 26482043 DOI: 10.1158/1078-0432.ccr-15-0534] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 09/20/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE Although 67% of high-grade serous ovarian cancers (HGSOC) express the estrogen receptor (ER), most fail antiestrogen therapy. Because MAPK activation is frequent in ovarian cancer, we investigated if estrogen regulates MAPK and if MEK inhibition (MEKi) reverses antiestrogen resistance. EXPERIMENTAL DESIGN Effects of MEKi (selumetinib), antiestrogen (fulvestrant), or both were assayed in ER-positive HGSOC in vitro and in xenografts. Response biomarkers were investigated by gene expression microarray and reverse phase protein array (RPPA). Genes differentially expressed in two independent primary HGSOC datasets with high versus low pMAPK by RPPA were used to generate a "MAPK-activated gene signature." Gene signature components that were reversed by MEKi were then identified. RESULTS High intratumor pMAPK independently predicts decreased survival (HR, 1.7; CI > 95%,1.3-2.2; P = 0.0009) in 408 HGSOC from The Cancer Genome Atlas. A differentially expressed "MAPK-activated" gene subset was also prognostic. "MAPK-activated genes" in HGSOC differ from those in breast cancer. Combined MEK and ER blockade showed greater antitumor effects in xenografts than monotherapy. Gene set enrichment analysis and RPPA showed that dual therapy downregulated DNA replication and cell-cycle drivers, and upregulated lysosomal gene sets. Selumetinib reversed expression of a subset of "MAPK-activated genes" in vitro and/or in xenografts. Three of these genes were prognostic for poor survival (P = 0.000265) and warrant testing as a signature predictive of MEKi response. CONCLUSIONS High pMAPK is independently prognostic and may underlie antiestrogen failure. Data support further evaluation of fulvestrant and selumetinib in ER-positive HGSOC. The MAPK-activated HGSOC signature may help identify MEK inhibitor responsive tumors.
Collapse
Affiliation(s)
- Karina E Hew
- Department of Obstetrics and Gynecology, University of Miami Miller School of Medicine, Miami, Florida. Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
| | - Philip C Miller
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida. Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida
| | - Dorraya El-Ashry
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida. Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida
| | - Jun Sun
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
| | - Alexandra H Besser
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
| | - Tan A Ince
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida. Department of Pathology, University of Miami Miller School of Medicine, Miami, Florida
| | - Mengnan Gu
- Department of Computer Science, New Jersey Institute of Technology, Newark, New Jersey
| | - Zhi Wei
- Department of Computer Science, New Jersey Institute of Technology, Newark, New Jersey
| | - Gao Zhang
- Wistar Institute, Philadelphia, Pennsylvania
| | | | - Wei Gao
- Department of Systems Biology, MD Anderson Cancer Center, Houston, Texas
| | - Yiling Lu
- Department of Systems Biology, MD Anderson Cancer Center, Houston, Texas
| | - Gordon B Mills
- Department of Systems Biology, MD Anderson Cancer Center, Houston, Texas
| | - Joyce M Slingerland
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida. Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida.
| | - Fiona Simpkins
- Department of Obstetrics and Gynecology, University of Miami Miller School of Medicine, Miami, Florida. Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
43
|
Atmospheric pressure gas plasma-induced colorectal cancer cell death is mediated by Nox2–ASK1 apoptosis pathways and oxidative stress is mitigated by Srx–Nrf2 anti-oxidant system. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1843:2827-37. [DOI: 10.1016/j.bbamcr.2014.08.011] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 08/19/2014] [Accepted: 08/20/2014] [Indexed: 11/24/2022]
|
44
|
The differential susceptibilities of MCF-7 and MDA-MB-231 cells to the cytotoxic effects of curcumin are associated with the PI3K/Akt-SKP2-Cip/Kips pathway. Cancer Cell Int 2014; 14:126. [PMID: 25530715 PMCID: PMC4272549 DOI: 10.1186/s12935-014-0126-4] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 11/10/2014] [Indexed: 01/08/2023] Open
Abstract
Background The mechanism underlying the differential cytotoxicity of curcumin in various cancer types, however, remains largely unclear. The aims of this study is to examine the concentration- and time-related effects of curcumin on two different breast cancer cells, MCF-7 and MDA-MB-231, and investigated the functional changes induced by curcumin treatment, as well as their relationship to the PI3K/Akt-SKP2-Cip/Kips pathway. Methods First, WST-1 and clonogenic assay were performed to determine the cytotoxicity of curcumin in MCF-7 and MDA-MB-231 cells. Then, the expression of CDK interacting protein/Kinase inhibitory protein (Cip/Kips) members (p27, p21 and p57) and S-phase kinase-associated protein-2 (SKP2) was investigated by QRT PCR and Western Blotting. Curcumin’s effect on PI3K (phosphatidylinositol 3-kinase) /Akt and its substrates Foxo1 and Foxo3a were then studied by Western Blotting. Small interfering RNAs (siRNAs) targeting SKP2 was used to explore the relationship between SKP2 and Cip/Kips members. Finally, WST-1 assay was tested to explore the concomitant treatment with curcumin and the inhibition of PKB or SKP2 signaling on curcumin sensitivity in MCF-7 and MDA-MB-231 cells. Results We demonstrated MCF-7 and MDA-MB-231 cells exhibited differential responses to curcumin by WST-1 and clonogenic assay (MDA-MB-231 cells was sensitive, and MCF-7 cells was resistant), which were found to be related to the differential curcumin-mediated regulation of SKP2-Cip/Kips (p21 and p27 but not p57) signaling. The differential cellular responses were further linked to the converse effects of curcumin on PI3K/Akt and its substrates Foxo1 and Foxo3a. Importantly, PI3K inhibitor wortmannin could counteract both curcumin-induced phosphorylation of Akt and up-regulation of SKP2 in MCF-7 cells. Subsequent WST-1 assay demonstrated concomitant treatment with curcumin and wortmannin or SKP2 siRNA not only further augmented curcumin sensitivity in MDA-MB-231 cells but also overcame curcumin resistance in MCF-7 cells. Conclusions Our study established PI3K/Akt-SKP2-Cip/Kips signaling pathway is involved in the mechanism of action of curcumin and revealed that the discrepant modulation of this pathway by curcumin is responsible for the differential susceptibilities of these two cell types to curcumin.
Collapse
|
45
|
Zhao YF, Zhao JY, Yue H, Hu KS, Shen H, Guo ZG, Su XJ. FOXD1 promotes breast cancer proliferation and chemotherapeutic drug resistance by targeting p27. Biochem Biophys Res Commun 2014; 456:232-7. [PMID: 25462566 DOI: 10.1016/j.bbrc.2014.11.064] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 11/14/2014] [Indexed: 12/27/2022]
Abstract
Forkhead transcription factors are essential for diverse processes in early embryonic development and organogenesis. As a member of the forkhead family, FOXD1 is required during kidney development and its inactivation results in failure of nephron progenitor cells. However, the role of FOXD1 in carcinogenesis and progression is still limited. Here, we reported that FOXD1 is a potential oncogene in breast cancer. We found that FOXD1 is up-regulated in breast cancer tissues. Depletion of FOXD1 expression decreases the ability of cell proliferation and chemoresistance in MDA-MB-231 cells, whereas overexpression of FOXD1 increases the ability of cell proliferation and chemoresistance in MCF-7 cells. Furthermore, we observed that FOXD1 induces G1 to S phase transition by targeting p27 expression. Our results suggest that FOXD1 may be a potential therapy target for patients with breast cancer.
Collapse
Affiliation(s)
- Yi-Fan Zhao
- Department of Anesthesiology, The First Affiliated Hospital of the General Hospital of CPLA, Beijing 100048, China
| | - Jing-Yu Zhao
- Department of Anesthesiology, The First Affiliated Hospital of the General Hospital of CPLA, Beijing 100048, China
| | - Hong Yue
- Department of Anesthesiology, The First Affiliated Hospital of the General Hospital of CPLA, Beijing 100048, China
| | - Ke-Shi Hu
- Department of Anesthesiology, The General Hospital of CPLA, Beijing 100853, China
| | - Hao Shen
- Department of Anesthesiology, The General Hospital of CPLA, Beijing 100853, China
| | - Zheng-Gang Guo
- Department of Anesthesiology, The First Affiliated Hospital of the General Hospital of CPLA, Beijing 100048, China.
| | - Xiao-Jun Su
- Department of Anesthesiology, The First Affiliated Hospital of the General Hospital of CPLA, Beijing 100048, China.
| |
Collapse
|
46
|
Abstract
Around 70% of all breast cancers are estrogen receptor alpha positive and hence their development is highly dependent on estradiol. While the invention of endocrine therapies has revolusioned the treatment of the disease, resistance to therapy eventually occurs in a large number of patients. This paper seeks to illustrate and discuss the complexity and heterogeneity of the mechanisms which underlie resistance and the approaches proposed to combat them. It will also focus on the use and development of methods for predicting which patients are likely to develop resistance.
Collapse
|
47
|
Viedma-Rodríguez R, Baiza-Gutman L, Salamanca-Gómez F, Diaz-Zaragoza M, Martínez-Hernández G, Ruiz Esparza-Garrido R, Velázquez-Flores MA, Arenas-Aranda D. Mechanisms associated with resistance to tamoxifen in estrogen receptor-positive breast cancer (review). Oncol Rep 2014; 32:3-15. [PMID: 24841429 DOI: 10.3892/or.2014.3190] [Citation(s) in RCA: 121] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 04/03/2014] [Indexed: 11/06/2022] Open
Abstract
Anti-estrogens such as tamoxifen are widely used in the clinic to treat estrogen receptor-positive breast tumors. Patients with estrogen receptor-positive breast cancer initially respond to treatment with anti-hormonal agents such as tamoxifen, but remissions are often followed by the acquisition of resistance and, ultimately, disease relapse. The development of a rationale for the effective treatment of tamoxifen-resistant breast cancer requires an understanding of the complex signal transduction mechanisms. In the present study, we explored some mechanisms associated with resistance to tamoxifen, such as pharmacologic mechanisms, loss or modification in estrogen receptor expression, alterations in co-regulatory proteins and the regulation of the different signaling pathways that participate in different cellular processes such as survival, proliferation, stress, cell cycle, inhibition of apoptosis regulated by the Bcl-2 family, autophagy, altered expression of microRNA, and signaling pathways that regulate the epithelial-mesenchymal transition in the tumor microenvironment. Delineation of the molecular mechanisms underlying the development of resistance may aid in the development of treatment strategies to enhance response and compromise resistance.
Collapse
Affiliation(s)
- Rubí Viedma-Rodríguez
- Molecular Genetics Laboratory, Medical Research Unit in Human Genetics, Pediatric Hospital, National Medical Center Century XXI (CMN-SXXI), Mexican Social Security Institute (IMSS), Mexico City, Mexico
| | - Luis Baiza-Gutman
- Unit of Morphology and Function, Faculty of Higher Studies (FES) Iztacala, National Autonomous University of Mexico (UNAM), Los Reyes Iztacala, State of Mexico, Mexico
| | - Fabio Salamanca-Gómez
- Molecular Genetics Laboratory, Medical Research Unit in Human Genetics, Pediatric Hospital, National Medical Center Century XXI (CMN-SXXI), Mexican Social Security Institute (IMSS), Mexico City, Mexico
| | | | - Guadalupe Martínez-Hernández
- Unit of Morphology and Function, Faculty of Higher Studies (FES) Iztacala, National Autonomous University of Mexico (UNAM), Los Reyes Iztacala, State of Mexico, Mexico
| | - Ruth Ruiz Esparza-Garrido
- Molecular Genetics Laboratory, Medical Research Unit in Human Genetics, Pediatric Hospital, National Medical Center Century XXI (CMN-SXXI), Mexican Social Security Institute (IMSS), Mexico City, Mexico
| | - Miguel Angel Velázquez-Flores
- Molecular Genetics Laboratory, Medical Research Unit in Human Genetics, Pediatric Hospital, National Medical Center Century XXI (CMN-SXXI), Mexican Social Security Institute (IMSS), Mexico City, Mexico
| | - Diego Arenas-Aranda
- Molecular Genetics Laboratory, Medical Research Unit in Human Genetics, Pediatric Hospital, National Medical Center Century XXI (CMN-SXXI), Mexican Social Security Institute (IMSS), Mexico City, Mexico
| |
Collapse
|
48
|
Saldanha SN, Tollefsbol TO. Pathway modulations and epigenetic alterations in ovarian tumorbiogenesis. J Cell Physiol 2014; 229:393-406. [PMID: 24105793 DOI: 10.1002/jcp.24466] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2013] [Accepted: 09/06/2013] [Indexed: 12/23/2022]
Abstract
Cellular pathways are numerous and are highly integrated in function in the control of cellular systems. They collectively regulate cell division, proliferation, survival and apoptosis of cells and mutagenesis of key genes that control these pathways can initiate neoplastic transformations. Understanding these pathways is crucial to future therapeutic and preventive strategies of the disease. Ovarian cancers are of three major types; epithelial, germ-cell, and stromal. However, ovarian cancers of epithelial origin, arising from the mesothelium, are the predominant form. Of the subtypes of ovarian cancer, the high-grade serous tumors are fatal, with low survival rate due to late detection and poor response to treatments. Close examination of preserved ovarian tissues and in vitro studies have provided insights into the mechanistic changes occurring in cells mediated by a few key genes. This review will focus on pathways and key genes of the pathways that are mutated or have aberrant functions in the pathology of ovarian cancer. Non-genetic mechanisms that are gaining prominence in the pathology of ovarian cancer, miRNAs and epigenetics, will also be discussed in the review.
Collapse
Affiliation(s)
- Sabita N Saldanha
- Department of Biology, University of Alabama at Birmingham, Birmingham, Alabama; Department of Biological Sciences, Alabama State University, Montgomery, Alabama
| | | |
Collapse
|
49
|
Zhu J, Zhao C, Kharman-Biz A, Zhuang T, Jonsson P, Liang N, Williams C, Lin CY, Qiao Y, Zendehdel K, Strömblad S, Treuter E, Dahlman-Wright K. The atypical ubiquitin ligase RNF31 stabilizes estrogen receptor α and modulates estrogen-stimulated breast cancer cell proliferation. Oncogene 2014; 33:4340-51. [PMID: 24441041 PMCID: PMC4141304 DOI: 10.1038/onc.2013.573] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Revised: 11/29/2013] [Accepted: 12/10/2013] [Indexed: 12/28/2022]
Abstract
Estrogen receptor α (ERα) is initially expressed in the majority of breast cancers and promotes estrogen-dependent cancer progression by regulating the transcription of genes linked to cell proliferation. ERα status is of clinical importance, as ERα-positive breast cancers can be successfully treated by adjuvant therapy with antiestrogens or aromatase inhibitors. Complications arise from the frequent development of drug resistance that might be caused by multiple alterations, including components of ERα signaling, during tumor progression and metastasis. Therefore, insights into the molecular mechanisms that control ERα expression and stability are of utmost importance to improve breast cancer diagnostics and therapeutics. Here we report that the atypical E3 ubiquitin ligase RNF31 stabilizes ERα and facilitates ERα-stimulated proliferation in breast cancer cell lines. We show that depletion of RNF31 decreases the number of cells in the S phase and reduces the levels of ERα and its downstream target genes, including cyclin D1 and c-myc. Analysis of data from clinical samples confirms correlation between RNF31 expression and the expression of ERα target genes. Immunoprecipitation indicates that RNF31 associates with ERα and increases its stability and mono-ubiquitination, dependent on the ubiquitin ligase activity of RNF31. Our data suggest that association of RNF31 and ERα occurs mainly in the cytosol, consistent with the lack of RNF31 recruitment to ERα-occupied promoters. In conclusion, our study establishes a non-genomic mechanism by which RNF31 via stabilizing ERα levels controls the transcription of estrogen-dependent genes linked to breast cancer cell proliferation.
Collapse
Affiliation(s)
- J Zhu
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - C Zhao
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - A Kharman-Biz
- 1] Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden [2] Cancer Research Center, Cancer institute of Iran, Tehran University of Medical Sciences, Tehran, Iran
| | - T Zhuang
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - P Jonsson
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - N Liang
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - C Williams
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - C-Y Lin
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Y Qiao
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - K Zendehdel
- Cancer Research Center, Cancer institute of Iran, Tehran University of Medical Sciences, Tehran, Iran
| | - S Strömblad
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - E Treuter
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - K Dahlman-Wright
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| |
Collapse
|
50
|
El Touny LH, Vieira A, Mendoza A, Khanna C, Hoenerhoff MJ, Green JE. Combined SFK/MEK inhibition prevents metastatic outgrowth of dormant tumor cells. J Clin Invest 2013; 124:156-68. [PMID: 24316974 DOI: 10.1172/jci70259] [Citation(s) in RCA: 113] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Accepted: 10/03/2013] [Indexed: 12/27/2022] Open
Abstract
Breast cancer (BC) can recur as metastatic disease many years after primary tumor removal, suggesting that disseminated tumor cells survive for extended periods in a dormant state that is refractory to conventional therapies. We have previously shown that altering the tumor microenvironment through fibrosis with collagen and fibronectin deposition can trigger tumor cells to switch from a dormant to a proliferative state. Here, we used an in vivo preclinical model and a 3D in vitro model of dormancy to evaluate the role of Src family kinase (SFK) in regulating this dormant-to-proliferative switch. We found that pharmacological inhibition of SFK signaling or Src knockdown results in the nuclear localization of cyclin-dependent kinase inhibitor p27 and prevents the proliferative outbreak of dormant BC cells and metastatic lesion formation; however, SFK inhibition did not kill dormant cells. Dormant cell proliferation also required ERK1/2 activation. Combination treatment of cells undergoing the dormant-to-proliferative switch with the Src inhibitor (AZD0530) and MEK1/2 inhibitor (AZD6244) induced apoptosis in a large fraction of the dormant cells and delayed metastatic outgrowth, neither of which was observed with either inhibitor alone. Thus, targeting Src prevents the proliferative response of dormant cells to external stimuli, but requires MEK1/2 inhibition to suppress their survival. These data indicate that treatments targeting Src in combination with MEK1/2 may prevent BC recurrence.
Collapse
|