1
|
Hariri A, Mirian M, Khosravi A, Zarepour A, Iravani S, Zarrabi A. Intersecting pathways: The role of hybrid E/M cells and circulating tumor cells in cancer metastasis and drug resistance. Drug Resist Updat 2024; 76:101119. [PMID: 39111134 DOI: 10.1016/j.drup.2024.101119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 06/30/2024] [Accepted: 07/09/2024] [Indexed: 08/17/2024]
Abstract
Cancer metastasis and therapy resistance are intricately linked with the dynamics of Epithelial-Mesenchymal Transition (EMT) and Circulating Tumor Cells (CTCs). EMT hybrid cells, characterized by a blend of epithelial and mesenchymal traits, have emerged as pivotal in metastasis and demonstrate remarkable plasticity, enabling transitions across cellular states crucial for intravasation, survival in circulation, and extravasation at distal sites. Concurrently, CTCs, which are detached from primary tumors and travel through the bloodstream, are crucial as potential biomarkers for cancer prognosis and therapeutic response. There is a significant interplay between EMT hybrid cells and CTCs, revealing a complex, bidirectional relationship that significantly influences metastatic progression and has a critical role in cancer drug resistance. This resistance is further influenced by the tumor microenvironment, with factors such as tumor-associated macrophages, cancer-associated fibroblasts, and hypoxic conditions driving EMT and contributing to therapeutic resistance. It is important to understand the molecular mechanisms of EMT, characteristics of EMT hybrid cells and CTCs, and their roles in both metastasis and drug resistance. This comprehensive understanding sheds light on the complexities of cancer metastasis and opens avenues for novel diagnostic approaches and targeted therapies and has significant advancements in combating cancer metastasis and overcoming drug resistance.
Collapse
Affiliation(s)
- Amirali Hariri
- Department of Pharmaceutical Biotechnology, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan 8174673461, Iran
| | - Mina Mirian
- Department of Pharmaceutical Biotechnology, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan 8174673461, Iran.
| | - Arezoo Khosravi
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, Istanbul Okan University, Istanbul 34959, Turkiye
| | - Atefeh Zarepour
- Department of Research Analytics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600 077, India
| | - Siavash Iravani
- Independent Researcher, W Nazar ST, Boostan Ave, Isfahan, Iran.
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul 34396, Turkiye; Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan 320315, Taiwan.
| |
Collapse
|
2
|
Yousefi T, Mohammadi Jobani B, Taebi R, Qujeq D. Innovating Cancer Treatment Through Cell Cycle, Telomerase, Angiogenesis, and Metastasis. DNA Cell Biol 2024; 43:438-451. [PMID: 39018567 DOI: 10.1089/dna.2024.0109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/19/2024] Open
Abstract
Cancer remains a formidable challenge in the field of medicine, necessitating innovative therapeutic strategies to combat its relentless progression. The cell cycle, a tightly regulated process governing cell growth and division, plays a pivotal role in cancer development. Dysregulation of the cell cycle allows cancer cells to proliferate uncontrollably. Therapeutic interventions designed to disrupt the cell cycle offer promise in restraining tumor growth and progression. Telomerase, an enzyme responsible for maintaining telomere length, is often overactive in cancer cells, conferring them with immortality. Targeting telomerase presents an opportunity to limit the replicative potential of cancer cells and hinder tumor growth. Angiogenesis, the formation of new blood vessels, is essential for tumor growth and metastasis. Strategies aimed at inhibiting angiogenesis seek to deprive tumors of their vital blood supply, thereby impeding their progression. Metastasis, the spread of cancer cells from the primary tumor to distant sites, is a major challenge in cancer therapy. Research efforts are focused on understanding the underlying mechanisms of metastasis and developing interventions to disrupt this deadly process. This review provides a glimpse into the multifaceted approach to cancer therapy, addressing critical aspects of cancer biology-cell cycle regulation, telomerase activity, angiogenesis, and metastasis. Through ongoing research and innovative strategies, the field of oncology continues to advance, offering new hope for improved treatment outcomes and enhanced quality of life for cancer patients.
Collapse
Affiliation(s)
- Tooba Yousefi
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bahareh Mohammadi Jobani
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reyhaneh Taebi
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Durdi Qujeq
- Department of Clinical Biochemistry, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
3
|
Jiang F, Yu M, Liang Y, Ding K, Wang Y. Discovery of Novel Diaryl-Substituted Fused Heterocycles Targeting Katanin and Tubulin with Potent Antitumor and Antimultidrug Resistance Efficacy. J Med Chem 2024; 67:12118-12142. [PMID: 38996194 DOI: 10.1021/acs.jmedchem.4c00878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/14/2024]
Abstract
Disrupting microtubule dynamics has emerged as a promising strategy for cancer treatment. However, drug resistance remains a challenge hindering the development of microtubule-targeting agents. In this work, a novel class of diaryl substituted fused heterocycles were designed, synthesized, and evaluated, which were demonstrated as effective dual katanin and tubulin regulators with antitumor activity. Following three rounds of stepwise optimization, compound 21b, featuring a 3H-imidazo[4,5-b]pyridine core, displayed excellent targeting capabilities on katanin and tubulin, along with notable antiproliferative and antimetastatic effects. Mechanistic studies revealed that 21b disrupts the microtubule network in tumor cells, leading to G2/M cell cycle arrest and apoptosis induction. Importantly, 21b exhibited significant inhibition of tumor growth in MDA-MB-231 and A549/T xenograft tumor models without evident toxicity and side effects. In conclusion, compound 21b presents a novel mechanism for disrupting microtubule dynamics, warranting further investigation as a dual-targeted antitumor agent with potential antimultidrug resistance properties.
Collapse
Affiliation(s)
- Fuhao Jiang
- School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Min Yu
- School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yuru Liang
- State Key Laboratory of Organometallic Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China
| | - Kuiling Ding
- State Key Laboratory of Organometallic Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China
| | - Yang Wang
- School of Pharmacy, Fudan University, Shanghai 201203, China
- School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang 453007, China
| |
Collapse
|
4
|
Thakur C, Qiu Y, Pawar A, Chen F. Epigenetic regulation of breast cancer metastasis. Cancer Metastasis Rev 2024; 43:597-619. [PMID: 37857941 DOI: 10.1007/s10555-023-10146-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 10/02/2023] [Indexed: 10/21/2023]
Abstract
Breast cancer is the most frequently diagnosed malignancy and the second leading cause of cancer-related mortality among women worldwide. Recurrent metastasis is associated with poor patient outcomes and poses a significant challenge in breast cancer therapies. Cancer cells adapting to a new tissue microenvironment is the key event in distant metastasis development, where the disseminating tumor cells are likely to acquire genetic and epigenetic alterations during the process of metastatic colonization. Despite several decades of research in this field, the exact mechanisms governing metastasis are not fully understood. However, emerging body of evidence indicates that in addition to genetic changes, epigenetic reprogramming of cancer cells and the metastatic niche are paramount toward successful metastasis. Here, we review and discuss the latest knowledge about the salient attributes of metastasis and epigenetic regulation in breast cancer and crucial research domains that need further investigation.
Collapse
Affiliation(s)
- Chitra Thakur
- Stony Brook Cancer Center, Renaissance School of Medicine, Stony Brook University, Lauterbur Drive, Stony Brook, NY, 11794, USA.
| | - Yiran Qiu
- Stony Brook Cancer Center, Renaissance School of Medicine, Stony Brook University, Lauterbur Drive, Stony Brook, NY, 11794, USA
| | - Aashna Pawar
- Stony Brook Cancer Center, Renaissance School of Medicine, Stony Brook University, Lauterbur Drive, Stony Brook, NY, 11794, USA
| | - Fei Chen
- Stony Brook Cancer Center, Renaissance School of Medicine, Stony Brook University, Lauterbur Drive, Stony Brook, NY, 11794, USA.
| |
Collapse
|
5
|
Inayatullah M, Mahesh A, Turnbull AK, Dixon JM, Natrajan R, Tiwari VK. Basal-epithelial subpopulations underlie and predict chemotherapy resistance in triple-negative breast cancer. EMBO Mol Med 2024; 16:823-853. [PMID: 38480932 PMCID: PMC11018633 DOI: 10.1038/s44321-024-00050-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 02/07/2024] [Accepted: 02/14/2024] [Indexed: 03/18/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive breast cancer subtype, characterized by extensive intratumoral heterogeneity, high metastasis, and chemoresistance, leading to poor clinical outcomes. Despite progress, the mechanistic basis of these aggressive behaviors remains poorly understood. Using single-cell and spatial transcriptome analysis, here we discovered basal epithelial subpopulations located within the stroma that exhibit chemoresistance characteristics. The subpopulations are defined by distinct signature genes that show a frequent gain in copy number and exhibit an activated epithelial-to-mesenchymal transition program. A subset of these genes can accurately predict chemotherapy response and are associated with poor prognosis. Interestingly, among these genes, elevated ITGB1 participates in enhancing intercellular signaling while ACTN1 confers a survival advantage to foster chemoresistance. Furthermore, by subjecting the transcriptional signatures to drug repurposing analysis, we find that chemoresistant tumors may benefit from distinct inhibitors in treatment-naive versus post-NAC patients. These findings shed light on the mechanistic basis of chemoresistance while providing the best-in-class biomarker to predict chemotherapy response and alternate therapeutic avenues for improved management of TNBC patients resistant to chemotherapy.
Collapse
Affiliation(s)
- Mohammed Inayatullah
- Institute for Molecular Medicine, University of Southern Denmark, Odense M, Denmark
| | - Arun Mahesh
- Institute for Molecular Medicine, University of Southern Denmark, Odense M, Denmark
| | - Arran K Turnbull
- Edinburgh Breast Cancer Now Research Group, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, EH4 2XU, UK
| | - J Michael Dixon
- Edinburgh Breast Cancer Now Research Group, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, EH4 2XU, UK
| | - Rachael Natrajan
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Vijay K Tiwari
- Institute for Molecular Medicine, University of Southern Denmark, Odense M, Denmark.
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queens University Belfast, Belfast, BT9 7BL, UK.
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, BT9 7AE, UK.
- Danish Institute for Advanced Study (DIAS), Odense M, Denmark.
- Department of Clinical Genetics, Odense University Hospital, Odense C, Denmark.
| |
Collapse
|
6
|
Nasr MM, Lynch CC. How circulating tumor cluster biology contributes to the metastatic cascade: from invasion to dissemination and dormancy. Cancer Metastasis Rev 2023; 42:1133-1146. [PMID: 37442876 PMCID: PMC10713810 DOI: 10.1007/s10555-023-10124-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 07/05/2023] [Indexed: 07/15/2023]
Abstract
Circulating tumor cells (CTCs) are known to be prognostic for metastatic relapse and are detected in patients as solitary cells or cell clusters. Circulating tumor cell clusters (CTC clusters) have been observed clinically for decades and are of significantly higher metastatic potential compared to solitary CTCs. Recent studies suggest distinct differences in CTC cluster biology regarding invasion and survival in circulation. However, differences regarding dissemination, dormancy, and reawakening require more investigations compared to solitary CTCs. Here, we review the current state of CTC cluster research and consider their clinical significance. In addition, we discuss the concept of collective invasion by CTC clusters and molecular evidence as to how cluster survival in circulation compares to that of solitary CTCs. Molecular differences between solitary and clustered CTCs during dormancy and reawakening programs will also be discussed. We also highlight future directions to advance our current understanding of CTC cluster biology.
Collapse
Affiliation(s)
- Mostafa M Nasr
- Tumor Biology Department, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
- Cancer Biology Ph.D. Program, University of South Florida, Tampa, FL, USA
| | - Conor C Lynch
- Tumor Biology Department, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA.
| |
Collapse
|
7
|
Sturgess V, Azubuike UF, Tanner K. Vascular regulation of disseminated tumor cells during metastatic spread. BIOPHYSICS REVIEWS 2023; 4:011310. [PMID: 38510161 PMCID: PMC10903479 DOI: 10.1063/5.0106675] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 02/17/2023] [Indexed: 03/22/2024]
Abstract
Cancer cells can travel to other organs via interconnected vascular systems to form new lesions in a process known as metastatic spread. Unfortunately, metastasis remains the leading cause of patient lethality. In recent years, it has been demonstrated that physical cues are just as important as chemical and genetic perturbations in driving changes in gene expression, cell motility, and survival. In this concise review, we focus on the physical cues that cancer cells experience as they migrate through the lymphatic and blood vascular networks. We also present an overview of steps that may facilitate organ specific metastasis.
Collapse
Affiliation(s)
- Victoria Sturgess
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 37, Room 2132, Bethesda MD 20892, USA
| | - Udochi F. Azubuike
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 37, Room 2132, Bethesda MD 20892, USA
| | - Kandice Tanner
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 37, Room 2132, Bethesda MD 20892, USA
| |
Collapse
|
8
|
Biomechanics of cancer stem cells. Essays Biochem 2022; 66:359-369. [PMID: 35942932 DOI: 10.1042/ebc20220014] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/27/2022] [Accepted: 08/02/2022] [Indexed: 12/27/2022]
Abstract
Cancer stem cells (CSCs) have been believed to be one driving force for tumor progression and drug resistance. Despite the significance of biochemical signaling in malignancy, highly malignant tumor cells or CSCs exhibit lower cellular stiffness than weakly malignant cells or non-CSCs, which are softer than their healthy counterparts, suggesting the inverse correlation between cell stiffness and malignancy. Recent years have witnessed the rapid accumulation of evidence illustrating the reciprocity between cell cytoskeleton/mechanics and CSC functions and the potential of cellular stiffness for specific targeting of CSCs. However, a systematic understanding of tumor cell mechanics and their role in CSCs and tumor progression is still lacking. The present review summarizes the recent progress in the alterations of tumor cell cytoskeleton and stiffness at different stages of tumor progression and recapitulates the relationship between cellular stiffness and CSC functions. The altered cell mechanics may mediate the mechanoadaptive responses that possibly empower CSCs to survive and thrive during metastasis. Furthermore, we highlight the possible impact of tumor cell mechanics on CSC malignancy, which may potentiate low cell stiffness as a mechanical marker for CSC targeting.
Collapse
|
9
|
Abstract
Metastasis is responsible for a large majority of death from malignant solid tumors. Bone is one of the most frequently affected organs in cancer metastasis, especially in breast and prostate cancer. Development of bone metastasis requires cancer cells to successfully complete a number of challenging steps, including local invasion and intravasation, survival in circulation, extravasation and initial seeding, and finally, formation of metastatic colonies after a period of dormancy or indolent growth. During this process, cancer cells often undergo a series of cellular and molecular changes to gain cellular plasticity that helps them adapt to various environments they encounter along the journey of metastasis. Understanding the mechanisms behind cellular plasticity and adaptation during the formation of bone metastasis is crucial for the development of novel therapies.
Collapse
Affiliation(s)
- Cao Fang
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Yibin Kang
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA.
| |
Collapse
|
10
|
Greaves D, Calle Y. Epithelial Mesenchymal Transition (EMT) and Associated Invasive Adhesions in Solid and Haematological Tumours. Cells 2022; 11:649. [PMID: 35203300 PMCID: PMC8869945 DOI: 10.3390/cells11040649] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 12/13/2022] Open
Abstract
In solid tumours, cancer cells that undergo epithelial mesenchymal transition (EMT) express characteristic gene expression signatures that promote invasive migration as well as the development of stemness, immunosuppression and drug/radiotherapy resistance, contributing to the formation of currently untreatable metastatic tumours. The cancer traits associated with EMT can be controlled by the signalling nodes at characteristic adhesion sites (focal contacts, invadopodia and microtentacles) where the regulation of cell migration, cell cycle progression and pro-survival signalling converge. In haematological tumours, ample evidence accumulated during the last decade indicates that the development of an EMT-like phenotype is indicative of poor disease prognosis. However, this EMT phenotype has not been directly linked to the assembly of specific forms of adhesions. In the current review we discuss the role of EMT in haematological malignancies and examine its possible link with the progression towards more invasive and aggressive forms of these tumours. We also review the known types of adhesions formed by haematological malignancies and speculate on their possible connection with the EMT phenotype. We postulate that understanding the architecture and regulation of EMT-related adhesions will lead to the discovery of new therapeutic interventions to overcome disease progression and resistance to therapies.
Collapse
Affiliation(s)
| | - Yolanda Calle
- School of Life Sciences and Health, University of Roehampton, London SW15 4JD, UK;
| |
Collapse
|
11
|
Xiang Z, He Q, Huang L, Xiong B, Xiang Q. Breast Cancer Classification Based on Tumor Budding and Stem Cell-Related Signatures Facilitate Prognosis Evaluation. Front Oncol 2022; 11:818869. [PMID: 35083162 PMCID: PMC8784696 DOI: 10.3389/fonc.2021.818869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 12/13/2021] [Indexed: 11/13/2022] Open
Abstract
Background Tumor budding (TB) is emerging as a prognostic factor in multiple cancers. Likewise, the stemness of cancer cells also plays a vital role in cancer progression. However, nearly no research has focused on the interaction of TB and tumor stemness in cancer. Methods Tissue microarrays including 229 cases of invasive breast cancer (BC) were established and subjected to pan-cytokeratin immunohistochemical staining to evaluate molecular expression. Univariate and multivariate analyses were applied to identify prognostic factors of BC, and the Chi-square test was used for comparison of categorical variables. Results High-grade TB was significantly associated with T stage, lymph node metastasis, tumor node metastasis (TNM) stage, epithelial-mesenchymal transition, and poor disease-free survival (DFS) of BC patients. We also found that the prognostic value of TB varied widely among different subtypes and subgroups. Cox regression analysis then showed that TB grade was an independent prognostic factor. Moreover, cancer stem cell (CSC) markers CD44 and ALDH1A1 were significantly higher in high-grade TB tumors. Consequently, patients were classified into high CSC score subgroup and low CSC score subgroups. Further research found that CSC scores correlated with clinicopathological features and DFS of BC patients. Based on TB grade and CSC scores, we classified BC patients into TBlow-CSCslow (type I), TBlow-CSCshigh (type II), TBhigh-CSCslow (type III), and TBhigh-CSCshigh (type IV) subgroups. Survival analysis showed that patients in the type I subgroup had the best DFS, whereas those in the type IV subgroup had the worst DFS. Finally, a TB-CSC-based nomogram for use in BC was established. The nomogram was well calibrated to predict the probability of 5-year DFS, and the C-index was 0.837. Finally, the area under the curve value for the nomogram (0.892) was higher than that of the TNM staging system (0.713). Conclusion The combination of TB grade with CSC score improves the prognostic evaluation of BC patients. A novel nomogram containing TB grade and CSC score provides doctors with a candidate tool to guide the individualized treatment of cancer patients.
Collapse
Affiliation(s)
- Zhenxian Xiang
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China.,Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China.,Hubei Cancer Clinical Study Center, Wuhan, China
| | - Qiuming He
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China.,Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China.,Hubei Cancer Clinical Study Center, Wuhan, China
| | - Li Huang
- Department of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bin Xiong
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China.,Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China.,Hubei Cancer Clinical Study Center, Wuhan, China
| | - Qingming Xiang
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China.,Hubei Cancer Clinical Study Center, Wuhan, China.,Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Hubei Key Laboratory of Tumor Biological Behaviors & Hubei Cancer Clinical Study Center, Wuhan, China
| |
Collapse
|
12
|
Katsarou SD, Messaritakis I, Voumvouraki A, Kakavogiannis S, Κotsakis A, Alkahtani S, Stournaras C, Martin SS, Georgoulias V, Kallergi G. Detyrosinated α-Tubulin, Vimentin and PD-L1 in Circulating Tumor Cells (CTCs) Isolated from Non-Small Cell Lung Cancer (NSCLC) Patients. J Pers Med 2022; 12:jpm12020154. [PMID: 35207643 PMCID: PMC8875112 DOI: 10.3390/jpm12020154] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 01/07/2022] [Accepted: 01/17/2022] [Indexed: 12/10/2022] Open
Abstract
Upregulation of Vimentin (VIM), alpha-Tubulin (TUB) and Detyrosinated tubulin (GLU) in circulating tumor cells (CTCs) derived from breast cancer patients is related to poor prognosis. In the current study we evaluated for the first time, these cytoskeletal proteins in sixty Non-Small Cell Lung Cancer (NSCLC) patients’ CTCs (33 treatment-naïve and 27 pre-treated). Samples were isolated using the ISET platform and stained with a pancytokeratin (CK)/CD45/TUB, CK/GLU/VIM and CK/programmed death ligand 1 (PD-L1) combination of antibodies. Subsequently, slides were analyzed using confocal laser scanning microscopy. CTCs were detected in 86.7% of the patients. CTCs with TUB expression were identified in 65.4% (34/52) of the CK (+)-patients. GLU, VIM and PD-L1 were also evaluated. The frequency of the observed phenotypes was as follow: (CK+/GLU−/VIM−): 35.2%, (CK+/GLU+/VIM+): 63.0%, (CK+/GLU+/VIM−): 16.7%, (CK+/GLU−/VIM+): 72.2%, (CK+/PD-L1−): 75% and (CK+/PD-L1+): 55%. The OS was significantly decreased in patients with high GLU (3.8 vs. 7.9 months; p = 0.018) and/or high VIM (3.2 vs. 7.1 months; p = 0.029) expression in their CTCs. PD-L1 was also related to OS (3.4 vs. 7.21 months; p = 0.035). Moreover, TUB-high and TUB-low expression in CTCs inversely influenced patients’ OS as independent prognostic factors (p = 0.041 and p = 0.009). The current study revealed that TUB, GLU, VIM and PD-L1 were overexpressed in CTCs from NSCLC patients. Furthermore, the presence of GLU, VIM-positive and PD-L1 in CTCs is potentially related to patients’ outcomes.
Collapse
Affiliation(s)
- Spyridoula D. Katsarou
- Division of Genetics, Cell and Developmental Biology, Department of Biology, University of Patras, 26504 Patras, Greece; (S.D.K.); (S.K.)
- Department of Zoology, Science College, King Saud University, Riyadh 11451, Saudi Arabia;
- Department of Biochemistry, Medical School, University of Crete, 71003 Heraklion, Greece; (A.V.); (C.S.)
| | - Ippokratis Messaritakis
- Laboratory of Translational Oncology, Medical School, University of Crete, 71003 Heraklion, Greece;
| | - Anastasia Voumvouraki
- Department of Biochemistry, Medical School, University of Crete, 71003 Heraklion, Greece; (A.V.); (C.S.)
| | - Stavros Kakavogiannis
- Division of Genetics, Cell and Developmental Biology, Department of Biology, University of Patras, 26504 Patras, Greece; (S.D.K.); (S.K.)
| | - Athanasios Κotsakis
- Department of Medical Oncology, University General Hospital of Larisa, 41334 Larisa, Greece;
- Hellenic Oncology Research Group (HORG), 11526 Athens, Greece;
| | - Saad Alkahtani
- Department of Zoology, Science College, King Saud University, Riyadh 11451, Saudi Arabia;
- Department of Biochemistry, Medical School, University of Crete, 71003 Heraklion, Greece; (A.V.); (C.S.)
| | - Christos Stournaras
- Department of Biochemistry, Medical School, University of Crete, 71003 Heraklion, Greece; (A.V.); (C.S.)
| | - Stuart S. Martin
- Department of Physiology, School of Medicine, University of Maryland, Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD 21201, USA;
| | | | - Galatea Kallergi
- Division of Genetics, Cell and Developmental Biology, Department of Biology, University of Patras, 26504 Patras, Greece; (S.D.K.); (S.K.)
- Correspondence: ; Tel.: +30-26-1096-9248
| |
Collapse
|
13
|
Tymon-Rosario J, Adjei NN, Roque DM, Santin AD. Microtubule-Interfering Drugs: Current and Future Roles in Epithelial Ovarian Cancer Treatment. Cancers (Basel) 2021; 13:6239. [PMID: 34944858 PMCID: PMC8699494 DOI: 10.3390/cancers13246239] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 12/06/2021] [Indexed: 11/17/2022] Open
Abstract
Taxanes and epothilones are chemotherapeutic agents that ultimately lead to cell death through inhibition of normal microtubular function. This review summarizes the literature demonstrating their current use and potential promise as therapeutic agents in the treatment of epithelial ovarian cancer (EOC), as well as putative mechanisms of resistance. Historically, taxanes have become the standard of care in the front-line and recurrent treatment of epithelial ovarian cancer. In the past few years, epothilones (i.e., ixabepilone) have become of interest as they may retain activity in taxane-treated patients since they harbor several features that may overcome mechanisms of taxane resistance. Clinical data now support the use of ixabepilone in the treatment of platinum-resistant or refractory ovarian cancer. Clinical data strongly support the use of microtubule-interfering drugs alone or in combination in the treatment of epithelial ovarian cancer. Ongoing clinical trials will shed further light into the potential of making these drugs part of current standard practice.
Collapse
Affiliation(s)
- Joan Tymon-Rosario
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT 06520, USA; (J.T.-R.); (N.N.A.)
| | - Naomi N. Adjei
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT 06520, USA; (J.T.-R.); (N.N.A.)
| | - Dana M. Roque
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
| | - Alessandro D. Santin
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT 06520, USA; (J.T.-R.); (N.N.A.)
| |
Collapse
|
14
|
Khanipouyani F, Akrami H. Tamoxifen Downregulates the Expression of Notch1 and DLL1 Genes in MKN-45 Gastric Cancer Cells. J Gastrointest Cancer 2021; 52:922-927. [PMID: 32901446 DOI: 10.1007/s12029-020-00511-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE Gastric cancer is one of the most prevalent cancers worldwide and the second most common cause for cancer associated mortality. Anti-tumor effects of tamoxifen in breast cancer are well-established. However, no study has so far investigated the effects of tamoxifen on gene expression of Notch1 and DLL1 in gastric cancer cell line. The present study was conducted to explore the effects of tamoxifen, as a repurposed drug, on gene expression of Notch1 and DLL1 in MKN-45, a gastric cancer cell line. METHODS MKN-45 cells were cultured in DMEM/F12 medium containing 10% FBS. Cytotoxic effects of tamoxifen on these cells at various concentrations were evaluated by trypan blue exclusion assay. For gene expression analysis, the cells were first incubated with 100 μM tamoxifen followed by total RNA extraction from treated and control cells. Then, cDNA was synthesized. Quantitative real-time PCR using specific primers for Notch1 and DLL1 was performed to assess the effect of tamoxifen on the transcript of them. RESULTS Treatment with tamoxifen decreased viability of MKN-45 cells in a dose-dependent manner. CC50 was estimated to be around 200 μM. Also, tamoxifen at the dose of 100 μM could significantly downregulate mRNA levels of both Notch1 and DLL1 genes as compared with untreated cells by 24% and 92%, respectively. CONCLUSION Based on these results, tamoxifen interferes with Notch signaling pathway through downregulating the expression of Notch1 and DLL1 genes and this could be regarded as a mechanism for its anti-cancer effects in this malignant disease.
Collapse
Affiliation(s)
| | - Hassan Akrami
- Gastroenterohepatology Research Center, Shiraz University of Medical Sciences, Shiraz, 71935-1311, Iran.
| |
Collapse
|
15
|
Xu G, Zhao H, Xu J, Zhang Y, Qi X, Shi A. Hard antler extract inhibits invasion and epithelial-mesenchymal transition of triple-negative and Her-2 + breast cancer cells by attenuating nuclear factor-κB signaling. JOURNAL OF ETHNOPHARMACOLOGY 2021; 269:113705. [PMID: 33346025 DOI: 10.1016/j.jep.2020.113705] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/24/2020] [Accepted: 12/15/2020] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Hard antler extract (HAE) is a traditional Chinese medicine and has potent antitumor, antioxidative, anti-inflammatory, and immunomodulatory activities. Previous studies have demonstrated that HAE can inhibit human prostate cancer metastasis and murine breast cancer proliferation. However, the effect of HAE on human breast cancer cells has not been clarified. AIM OF THE STUDY To investigate the effects and underlying mechanism of HAE on self-renewal of stem-like cells and spontaneous and transforming growth factor (TGF)-β1-enhanced wound healing, invasion and epithelial-mesenchymal transition (EMT) in breast cancer cells. METHODS HAE was prepared from sika deer by sequential enzymatic digestions and the active compounds were determined by HPLC. The effects of HAE on the viability, mammosphere formation, wound healing and invasion of MDA-MB-231 and SK-BR3 cells were determined. The impact of HAE treatment on spontaneous and TGF-β1-promoted EMT and the nuclear factor (NF)-κB signaling in breast cancer cells was examined by quantitative RT-PCR and western blotting. RESULTS Treatment with HAE at varying concentrations did not change the viability of breast cancer cells. However, HAE at 0.25 or 0.5 mg/mL significantly reduced the number and size of formed mammospheres, and inhibited spontaneous and TGF-β1-enhanced wound healing, invasion and EMT in MDA-MB-231 and SK-BR3 cells in a dose-dependent manner. TGF-β1 treatment significantly decreased IκBα expression and increased NF-kBp65 phosphorylation in breast cancer cells, indicating that TGF-β1 enhanced NF-κB signaling. In contrast, HAE treatment attenuated the spontaneous and TGF-β1-enhanced NF-κB signaling in breast cancer cells. CONCLUSION Our data indicated that HAE inhibited the self-renewal of stem-like cells and spontaneous and TGF-β1-enhanced wound healing, invasion and EMT in breast cancer cells by attenuating the NF-κB signaling in vitro.
Collapse
Affiliation(s)
- Gege Xu
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun, 130021, China
| | - Haiping Zhao
- Institute of Special Animal and Plant Sciences of CAAS, Changchun, 130112, China
| | - Jingdong Xu
- Biology Major, the University of Texas at Austin, Austin, TX, 78705, USA
| | - Yu Zhang
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun, 130021, China
| | - Xiaoyan Qi
- Institute of Special Animal and Plant Sciences of CAAS, Changchun, 130112, China
| | - Aiping Shi
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun, 130021, China.
| |
Collapse
|
16
|
Qin Q, Yang B, Liu Z, Xu L, Song E, Song Y. Polychlorinated biphenyl quinone induced the acquisition of cancer stem cells properties and epithelial-mesenchymal transition through Wnt/β-catenin. CHEMOSPHERE 2021; 263:128125. [PMID: 33297114 DOI: 10.1016/j.chemosphere.2020.128125] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/20/2020] [Accepted: 08/24/2020] [Indexed: 06/12/2023]
Abstract
Polychlorinated biphenyls (PCBs) are persistent industrial pollutants that have been linked to breast cancer progression. However, their molecular mechanism(s) are currently unclear. Our previous assessment suggested that the highly reactive PCB metabolite 2,3,5-trichloro-6-phenyl-[1,4]-benzoquinone (PCB29-pQ) induces the metastasis of breast cancer. Here, our data illustrate that PCB29-pQ increases cancer stem cell (CSC) marker expression, resulting in an increase in the epithelial-mesenchymal transition (EMT) in MDA-MB-231 breast cancer cells; further, the Wnt/β-catenin pathway also becomes activated by PCB29-pQ. When the Wnt/β-catenin pathway is inhibited, the promotion of CSC properties and EMT by PCB29-pQ were accordingly reversed. In addition, the overproduction of reactive oxygen species (ROS) mediated by PCB29-pQ plays a key role in Wnt/β-catenin activation. Collectively, our current data designated the regulatory role of Wnt/β-catenin in PCB29-pQ-triggered acquisition of CSC properties and EMT.
Collapse
Affiliation(s)
- Qi Qin
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, People's Republic of China
| | - Bingwei Yang
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, People's Republic of China
| | - Zixuan Liu
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, People's Republic of China
| | - Lei Xu
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, People's Republic of China
| | - Erqun Song
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, People's Republic of China
| | - Yang Song
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, People's Republic of China.
| |
Collapse
|
17
|
Ranganathan S, Kumar S, Mohanty SS, Jolly MK, Rangarajan A. Cellular Plasticity in Matrix-attached and -Detached Cells: Implications in Metastasis. J Indian Inst Sci 2020. [DOI: 10.1007/s41745-020-00179-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
18
|
Kong D, Hughes CJ, Ford HL. Cellular Plasticity in Breast Cancer Progression and Therapy. Front Mol Biosci 2020; 7:72. [PMID: 32391382 PMCID: PMC7194153 DOI: 10.3389/fmolb.2020.00072] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 03/31/2020] [Indexed: 12/24/2022] Open
Abstract
With the exception of non-melanoma skin cancer, breast cancer is the most frequently diagnosed malignant disease among women, with the majority of mortality being attributable to metastatic disease. Thus, even with improved early screening and more targeted treatments which may enable better detection and control of early disease progression, metastatic disease remains a significant problem. While targeted therapies exist for breast cancer patients with particular subtypes of the disease (Her2+ and ER/PR+), even in these subtypes the therapies are often not efficacious once the patient's tumor metastasizes. Increases in stemness or epithelial-to-mesenchymal transition (EMT) in primary breast cancer cells lead to enhanced plasticity, enabling tumor progression, therapeutic resistance, and distant metastatic spread. Numerous signaling pathways, including MAPK, PI3K, STAT3, Wnt, Hedgehog, and Notch, amongst others, play a critical role in maintaining cell plasticity in breast cancer. Understanding the cellular and molecular mechanisms that regulate breast cancer cell plasticity is essential for understanding the biology of breast cancer progression and for developing novel and more effective therapeutic strategies for targeting metastatic disease. In this review we summarize relevant literature on mechanisms associated with breast cancer plasticity, tumor progression, and drug resistance.
Collapse
Affiliation(s)
- Deguang Kong
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of General Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Connor J. Hughes
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Pharmacology Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Medical Scientist Training Program, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Heide L. Ford
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Pharmacology Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Medical Scientist Training Program, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
19
|
Zhou JM, Hu SQ, Jiang H, Chen YL, Feng JH, Chen ZQ, Wen KM. OCT4B1 Promoted EMT and Regulated the Self-Renewal of CSCs in CRC: Effects Associated with the Balance of miR-8064/PLK1. MOLECULAR THERAPY-ONCOLYTICS 2019; 15:7-20. [PMID: 31650021 PMCID: PMC6804455 DOI: 10.1016/j.omto.2019.08.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 08/20/2019] [Indexed: 12/24/2022]
Abstract
Cancer stem cells (CSCs) are the main cause of tumor generation, recurrence, metastasis, and therapy failure in various malignancies including colorectal cancer (CRC). Accumulating evidence suggests that tumor cells can acquire CSC characteristics through the epithelial-mesenchymal transition (EMT) process. However, the molecular mechanism of CSCs remains unclear. OCT4B1 is a transcript of OCT4, which is initially expressed in embryonic stem and carcinoma cells, and is involved in the regulation and maintenance of an undifferentiated state of stem cells. In this study, three-dimensional (3D) microspheres were confirmed as CRC stem cells. Compared with that of parental cells, their self-renewal ability was significantly increased, and OCT4B1 expression was increased and promoted the EMT process. The knockdown of OCT4B1 decreased the self-renewal of CSCs and reversed EMT. Moreover, OCT4B1 induced the expression of Polo-like kinase 1 (PLK1), which is a key regulator of EMT in tumor cells. Further examination showed that OCT4B1 regulated the miR-8064/PLK1 balance to exert its function. Taken together, our data suggest that OCT4B1 may be involved in regulating the self-renewal of colorectal CSCs through EMT, which is at least partially due to the miR-8064/PLK1 balance. This study indicates that OCT4B1 is a potential therapeutic target for CRC by targeting CSCs.
Collapse
Affiliation(s)
- Jun-Min Zhou
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Shui-Qing Hu
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Hang Jiang
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Yi-Lin Chen
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Ji-Hong Feng
- Department of Oncology, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Zheng-Quan Chen
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Kun-Ming Wen
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, China
| |
Collapse
|
20
|
Tan W, Liang G, Xie X, Jiang W, Tan L, Sanders AJ, Liu Z, Ling Y, Zhong W, Tian Z, Lin W, Gong C. Incorporating MicroRNA into Molecular Phenotypes of Circulating Tumor Cells Enhances the Prognostic Accuracy for Patients with Metastatic Breast Cancer. Oncologist 2019; 24:e1044-e1054. [PMID: 31300482 PMCID: PMC6853100 DOI: 10.1634/theoncologist.2018-0697] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 06/06/2019] [Indexed: 01/26/2023] Open
Abstract
The molecular phenotype of circulating tumor cells is associated with clinical outcome of patients with breast cancer. The aim of this study was to enhance the prognostic accuracy of the circulating tumor cell phenotype in metastatic breast cancer by incorporating miRNA into a combined prediction model. Background. The molecular phenotype of circulating tumor cells (CTCs) was associated with clinical outcome of patients with breast cancer. CTCs isolated from patients with metastatic breast cancer (MBC) display a unique microRNA (miRNA) expression profile. The aim of this study was to enhance the prognostic accuracy of the CTC phenotype in patients with MBC, by incorporating miRNA into a combined prediction model. Subjects, Materials, and Methods. CTCs were detected by CellSearch and enriched by magnetic cell sorting. miRNA deep sequencing and quantitative polymerase chain reaction were used to screen and verify potentially CTC‐specific miRNA candidates. Patients with MBC were enrolled from two independent cohorts, and overall survival (OS) and chemotherapy response were analyzed. Results. We screened and identified that miR‐106b was an upregulated molecule in patients with MBC with CTC ≥5/7.5 mL (n = 16) compared with patients with CTC = 0/7.5 mL (n = 16) and healthy donors (n = 8). The expression of CTC‐specific miR‐106b correlated with vimentin and E‐cadherin in CTC and acted as an independent factor for predicting OS (hazard ratio 2.157, 95% confidence interval [CI] 1.098–4.239, p = .026). Although CTC‐specific miR‐106b, E‐cadherin, and vimentin showed a prognostic potential independently, the prognostic performance for OS based on the combination of three markers was significantly enhanced in Cohort 1 (area under the curve [AUC] 0.752, 95% CI 0.658–0.847, n = 128) and further validated in Cohort 2 (AUC 0.726, 95% CI 0.595–0.856, n = 91). Besides, a combined model incorporating miR‐106b was associated with therapy response. Conclusion. The phenotypic assemblies of CTC incorporating miR‐106b show enhanced prognostic accuracy of overall survival in patients with MBC. Implications for Practice. In order to enhance the prognostic accuracy of the circulating tumor cell (CTC) phenotype in patients with metastatic breast cancer (MBC), this study screened and identified a CTC‐specific microRNA (miRNA), miR‐106b, as an upregulated molecule based on the comparison of miRNA profile between CTCs, primary tumors, and healthy blood donors. By incorporating miR‐106b into a combined prediction model, the prognostic accuracy of the CTC phenotype for patients with MBC was greatly improved in both the training and validation cohorts. This work provides clinical evidence supporting the prognostic potential of CTC‐specific miRNA for patients with MBC. These results indicate that developing CTC‐specific miRNAs as new biomarkers will help to further optimize personalized therapy.
Collapse
Affiliation(s)
- Weige Tan
- Breast Surgery Department, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Gehao Liang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation and Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Xinhua Xie
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Wenguo Jiang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation and Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Cardiff University, Heath Park, Cardiff, United Kingdom
| | - Luyuan Tan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation and Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Andrew J Sanders
- Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Cardiff University, Heath Park, Cardiff, United Kingdom
| | - Zihao Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation and Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yun Ling
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation and Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Wenjing Zhong
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation and Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Zhenluan Tian
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation and Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Wanyi Lin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation and Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Chang Gong
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation and Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Cardiff University, Heath Park, Cardiff, United Kingdom
| |
Collapse
|
21
|
Modulating the antitumoral activity by the design of new platinum(II) compounds: Synthesis, characterization, DFT, ultrastructure and mechanistic studies. J Inorg Biochem 2019; 194:200-213. [DOI: 10.1016/j.jinorgbio.2018.12.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Revised: 12/22/2018] [Accepted: 12/24/2018] [Indexed: 02/04/2023]
|
22
|
Vijay GV, Zhao N, Den Hollander P, Toneff MJ, Joseph R, Pietila M, Taube JH, Sarkar TR, Ramirez-Pena E, Werden SJ, Shariati M, Gao R, Sobieski M, Stephan CC, Sphyris N, Miura N, Davies P, Chang JT, Soundararajan R, Rosen JM, Mani SA. GSK3β regulates epithelial-mesenchymal transition and cancer stem cell properties in triple-negative breast cancer. Breast Cancer Res 2019; 21:37. [PMID: 30845991 PMCID: PMC6407242 DOI: 10.1186/s13058-019-1125-0] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 02/22/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Triple-negative breast cancers (TNBCs), which lack receptors for estrogen, progesterone, and amplification of epidermal growth factor receptor 2, are highly aggressive. Consequently, patients diagnosed with TNBCs have reduced overall and disease-free survival rates compared to patients with other subtypes of breast cancer. TNBCs are characterized by the presence of cancer cells with mesenchymal properties, indicating that the epithelial to mesenchymal transition (EMT) plays a major role in the progression of this disease. The EMT program has also been implicated in chemoresistance, tumor recurrence, and induction of cancer stem cell (CSC) properties. Currently, there are no targeted therapies for TNBC, and hence, it is critical to identify the novel targets to treat TNBC. METHODS A library of compounds was screened for their ability to inhibit EMT in cells with mesenchymal phenotype as assessed using the previously described Z-cad reporters. Of the several drugs tested, GSK3β inhibitors were identified as EMT inhibitors. The effects of GSK3β inhibitors on the properties of TNBC cells with a mesenchymal phenotype were assessed using qRT-PCR, flow cytometry, western blot, mammosphere, and migration and cell viability assays. Publicly available datasets also were analyzed to examine if the expression of GSK3β correlates with the overall survival of breast cancer patients. RESULTS We identified a GSK3β inhibitor, BIO, in a drug screen as one of the most potent inhibitors of EMT. BIO and two other GSK3β inhibitors, TWS119 and LiCl, also decreased the expression of mesenchymal markers in several different cell lines with a mesenchymal phenotype. Further, inhibition of GSK3β reduced EMT-related migratory properties of cells with mesenchymal properties. To determine if GSK3β inhibitors target mesenchymal-like cells by affecting the CSC population, we employed mammosphere assays and profiled the stem cell-related cell surface marker CD44+/24- in cells after exposure to GSK3β inhibitors. We found that GSK3β inhibitors indeed decreased the CSC properties of cell types with mesenchymal properties. We treated cells with epithelial and mesenchymal properties with GSK3β inhibitors and found that GSK3β inhibitors selectively kill cells with mesenchymal attributes while sparing cells with epithelial properties. We analyzed patient data to identify genes predictive of poor clinical outcome that could serve as novel therapeutic targets for TNBC. The Wnt signaling pathway is critical to EMT, but among the various factors known to be involved in Wnt signaling, only the higher expression of GSK3β correlated with poorer overall patient survival. CONCLUSIONS Taken together, our data demonstrate that GSK3β is a potential target for TNBCs and suggest that GSK3β inhibitors could serve as selective inhibitors of EMT and CSC properties for the treatment of a subset of aggressive TNBC. GSK3β inhibitors should be tested for use in combination with standard-of-care drugs in preclinical TNBC models.
Collapse
Affiliation(s)
- Geraldine Vidhya Vijay
- Department of Translational Molecular Pathology, UT MD Anderson Cancer Center, Houston, TX USA
| | - Na Zhao
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX USA
| | - Petra Den Hollander
- Department of Translational Molecular Pathology, UT MD Anderson Cancer Center, Houston, TX USA
| | - Mike J. Toneff
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX USA
| | - Robiya Joseph
- Department of Translational Molecular Pathology, UT MD Anderson Cancer Center, Houston, TX USA
| | - Mika Pietila
- Turku Centre for Biotechnology, University of Turku, Tykistökatu 6, 20520 Turku, Finland
| | | | - Tapasree R. Sarkar
- Center for Statistical Bioinformatics, Texas A&M University, College Station, TX USA
| | - Esmeralda Ramirez-Pena
- Department of Translational Molecular Pathology, UT MD Anderson Cancer Center, Houston, TX USA
| | - Steven J. Werden
- Department of Translational Molecular Pathology, UT MD Anderson Cancer Center, Houston, TX USA
| | - Maryam Shariati
- Department of Translational Molecular Pathology, UT MD Anderson Cancer Center, Houston, TX USA
| | - Ruli Gao
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Mary Sobieski
- Center for Translational Cancer Research, Texas A&M Health Science Center, Institute of Biosciences and Technology, Houston, TX USA
| | - Clifford C. Stephan
- Center for Translational Cancer Research, Texas A&M Health Science Center, Institute of Biosciences and Technology, Houston, TX USA
| | - Nathalie Sphyris
- Department of Translational Molecular Pathology, UT MD Anderson Cancer Center, Houston, TX USA
| | - Noayuki Miura
- Department of Biochemistry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Peter Davies
- Center for Translational Cancer Research, Texas A&M Health Science Center, Institute of Biosciences and Technology, Houston, TX USA
| | - Jeffrey T. Chang
- Department of Integrative Biology and Pharmacology, School of Medicine, School of Biomedical Informatics, UT Health Sciences Center at Houston, Houston, TX USA
- Center for Clinical and Translational Sciences, The University of Texas Health Science Center at Houston, Houston, TX USA
| | - Rama Soundararajan
- Department of Translational Molecular Pathology, UT MD Anderson Cancer Center, Houston, TX USA
| | - Jeffrey M. Rosen
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX USA
| | - Sendurai A. Mani
- Department of Translational Molecular Pathology, UT MD Anderson Cancer Center, Houston, TX USA
- Metastasis Research Center, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| |
Collapse
|
23
|
Sengodan SK, Rajan A, Hemalatha SK, Nadhan R, Jaleel A, Srinivas P. Proteomic Profiling of β-hCG-Induced Spheres in BRCA1 Defective Triple Negative Breast Cancer Cells. J Proteome Res 2017; 17:276-289. [DOI: 10.1021/acs.jproteome.7b00562] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Satheesh Kumar Sengodan
- Cancer Research Program and ‡Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala 695 014, India
| | - Arathi Rajan
- Cancer Research Program and ‡Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala 695 014, India
| | - Sreelatha Krishnakumar Hemalatha
- Cancer Research Program and ‡Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala 695 014, India
| | - Revathy Nadhan
- Cancer Research Program and ‡Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala 695 014, India
| | - Abdul Jaleel
- Cancer Research Program and ‡Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala 695 014, India
| | - Priya Srinivas
- Cancer Research Program and ‡Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala 695 014, India
| |
Collapse
|
24
|
Notch signaling pathway networks in cancer metastasis: a new target for cancer therapy. Med Oncol 2017; 34:180. [DOI: 10.1007/s12032-017-1039-6] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 09/13/2017] [Indexed: 12/19/2022]
|
25
|
Song W, Mazzieri R, Yang T, Gobe GC. Translational Significance for Tumor Metastasis of Tumor-Associated Macrophages and Epithelial-Mesenchymal Transition. Front Immunol 2017; 8:1106. [PMID: 28955335 PMCID: PMC5601389 DOI: 10.3389/fimmu.2017.01106] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 08/23/2017] [Indexed: 12/18/2022] Open
Abstract
The tumor microenvironment determines development and progression of many cancers. Epithelial–mesenchymal transition (EMT) is fundamental to tumor progression and metastasis not only by increasing invasiveness but also by increasing resistance to cell death, senescence, and various cancer therapies; determining inflammation and immune surveillance; and conferring stem cell properties. It does this by enabling polarized epithelial cells to transform into cells with a mesenchymal, and therefore motile, phenotype. Tumor-associated macrophages (TAMs) are key cells of the tumor microenvironment that orchestrate the connection between inflammation and cancer. Activation of EMT often requires crosstalk between cancer cells and components of the local tumor microenvironment, including TAMs. In this review, clinical and experimental evidence is presented for control of TAMs in promoting cancer cell invasion and migration and their interaction with the EMT process in the metastatic cascade. The translational significance of these findings is that the signaling pathways that interconnect TAMs and EMT-modified cancer cells may represent promising therapeutic targets for the treatment of tumor metastasis.
Collapse
Affiliation(s)
- Wenzhe Song
- Faculty of Medicine, Translational Research Institute, The University of Queensland Diamantina Institute, Brisbane, QLD, Australia.,Department of General Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Roberta Mazzieri
- Faculty of Medicine, Translational Research Institute, The University of Queensland Diamantina Institute, Brisbane, QLD, Australia
| | - Tao Yang
- Discipline of Pathology, The Western Sydney University, Sydney, NSW, Australia.,SydPath, St Vincent's Hospital, Sydney, NSW, Australia
| | - Glenda C Gobe
- Faculty of Medicine, Translational Research Institute, The University of Queensland Diamantina Institute, Brisbane, QLD, Australia
| |
Collapse
|
26
|
Septin remodeling is essential for the formation of cell membrane protrusions (microtentacles) in detached tumor cells. Oncotarget 2017; 8:76686-76698. [PMID: 29100341 PMCID: PMC5652735 DOI: 10.18632/oncotarget.20805] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 08/17/2017] [Indexed: 02/06/2023] Open
Abstract
Microtentacles are mostly microtubule-based cell protrusions that are formed by detached tumor cells. Here, we report that the formation of tumor cell microtentacles depends on the presence and dynamics of guanine nucleotide-binding proteins of the septin family, which are part of the cytoskeleton. In matrix-attached breast, lung, prostate and pancreas cancer cells, septins are associated with the cytosolic actin cytoskeleton. Detachment of cells causes redistribution of septins to the membrane, where microtentacle formation occurs. Forchlorfenuron, which inhibits septin functions, blocks microtentacle formation. The small GTPase Cdc42 and its effector proteins Borgs regulate septins and are essential for microtentacle formation. Dominant active and inactive Cdc42 inhibit microtentacle formation indicating that the free cycling of Cdc42 between its active and inactive state is essential for septin regulation and microtentacle formation. Cell attachment and aggregation models suggest that septins play an essential role in the metastatic behavior of tumor cells.
Collapse
|
27
|
Wang J, Liu Z, Zhang D, Liu R, Lin Q, Liu J, Yang Z, Ma Q, Sun D, Zhou X, Jiang G. FL118, a novel survivin inhibitor, wins the battle against drug-resistant and metastatic lung cancers through inhibition of cancer stem cell-like properties. Am J Transl Res 2017; 9:3676-3686. [PMID: 28861158 PMCID: PMC5575181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 07/22/2017] [Indexed: 06/07/2023]
Abstract
Failure of cancer treatment caused by drug resistance and metastasis is mainly due to existence of cancer stem cells (CSCs). Therefore, targeting CSCs to overcome cancers is a challenging issue in clinic. In this report, in view of the important role of survivin in tumor growth and CSCs maintaining, we aimed to confirm that FL118, as a novel survivin inhibitor, may effectively inhibit lung cancer stem cells. We showed that lung cancer stem cells have the obviously higher expression of survivin than their parental cells. After treated with FL118, the survivin level in CSCs was suppressed. Consistently, lung cancer stem cells displayed significantly growth inhibition over time. Here, we compared the antitumor efficacy between FL118 and cisplatin. The data revealed that CSCs are more sensitive to FL118 than cisplatin. To further demonstrate the inhibitory effect of FL118 on CSCs, we found that FL118 down-regulated the expression of CSCs markers (ABCG2, ALDH1A1, Oct4) and drug resistant proteins (P-gp, ERCC1), suggesting that FL118 may change CSCs phenotype and improve drug-sensitivity of tumor cells. Moreover, FL118 effectively decreased the invasive ability of CSCs. These findings expand the uniqueness of FL118 as an attractive therapeutic option for cancers with drug-resistant or metastatic potential.
Collapse
Affiliation(s)
- Jin Wang
- School of Basic Medicine, Qingdao UniversityQingdao 266021, China
| | - Zhantao Liu
- School of Pharmacy, Qingdao UniversityQingdao 266021, China
| | - Dandan Zhang
- School of Pharmacy, Qingdao UniversityQingdao 266021, China
| | - Ranran Liu
- School of Pharmacy, Qingdao UniversityQingdao 266021, China
| | - Qian Lin
- School of Pharmacy, Qingdao UniversityQingdao 266021, China
| | - Jia Liu
- School of Pharmacy, Qingdao UniversityQingdao 266021, China
| | - Zhihong Yang
- School of Pharmacy, Qingdao UniversityQingdao 266021, China
| | - Qingxia Ma
- School of Pharmacy, Qingdao UniversityQingdao 266021, China
| | - Dantong Sun
- School of Basic Medicine, Qingdao UniversityQingdao 266021, China
| | - Xin Zhou
- School of Basic Medicine, Qingdao UniversityQingdao 266021, China
| | - Guohui Jiang
- School of Pharmacy, Qingdao UniversityQingdao 266021, China
| |
Collapse
|
28
|
Cytoskeletal organization in microtentacles. Exp Cell Res 2017; 357:291-298. [PMID: 28551375 DOI: 10.1016/j.yexcr.2017.05.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 04/22/2017] [Accepted: 05/24/2017] [Indexed: 11/23/2022]
Abstract
Microtentacles are thin, flexible cell protrusions that have recently been described and whose presence enhances efficient attachment of circulating cells. They are found on circulating tumor cells and can be induced on a wide range of breast cancer cell lines, where they are promoted by factors that either stabilize microtubules or destabilize the actin cytoskeleton. Evidence suggests that they are relevant to the metastatic spread of cancer, so understanding their structure and formation may lead to useful therapies. Microtentacles are formed by microtubules and contain vimentin intermediate filaments, but beyond this, there is little information about their ultrastructure. We have used electron microscopy of high pressure frozen sections and tomography of cryo-prepared intact cells, along with super resolution fluorescence microscopy, to provide the first ultrastructural insights into microtubule and intermediate filament arrangement within microtentacles. By scanning electron microscopy it was seen that microtentacles form within minutes of addition of drugs that stabilize microtubules and destabilize actin filaments. Mature microtentacles were found to be well below one micrometer in diameter, tapering gradually to below 100nm at the distal ends. They also contained frequent branches and bulges suggestive of heterogeneous internal structure. Super resolution fluorescence microscopy and examination of sectioned samples showed that the microtubules and intermediate filaments can occupy different areas within the microtentacles, rather than interacting intimately as had been expected. Cryo-electron tomography of thin regions of microtentacles revealed densely packed microtubules and absence of intermediate filaments. The number of microtubules ranged from several dozen in some areas to just a few in the thinnest regions, with none of the regular arrangement found in axonemes. Improved understanding of the mechanism of microtentacle formation, as well as the resultant structure, will be valuable in developing therapies against metastasis, if the hypothesized role of microtentacles in metastasis is confirmed. This work provides a significant step in this direction.
Collapse
|
29
|
Holmboe S, Hansen PL, Thisgaard H, Block I, Müller C, Langkjær N, Høilund-Carlsen PF, Olsen BB, Mollenhauer J. Evaluation of somatostatin and nucleolin receptors for therapeutic delivery in non-small cell lung cancer stem cells applying the somatostatin-analog DOTATATE and the nucleolin-targeting aptamer AS1411. PLoS One 2017; 12:e0178286. [PMID: 28542563 PMCID: PMC5440050 DOI: 10.1371/journal.pone.0178286] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 05/10/2017] [Indexed: 01/31/2023] Open
Abstract
Cancer stem cells represent the putative tumor-driving subpopulation thought to account for drug resistance, relapse, and metastatic spread of epithelial and other cancer types. Accordingly, cell surface markers for therapeutic delivery to cancer stem cells are subject of intense research. Somatostatin receptor 2 and nucleolin are known to be overexpressed by various cancer types, which have elicited comprehensive efforts to explore their therapeutic utilization. Here, we evaluated somatostatin receptor 2 targeting and nucleolin targeting for therapeutic delivery to cancer stem cells from lung cancer. Nucleolin is expressed highly but not selectively, while somatostatin receptor 2 is expressed selectively but not highly by cancer cells. The non-small cell lung cancer cell lines A549 and H1299, displayed average levels of both surface molecules as judged based on analysis of a larger cell line panel. H1299 compared to A549 cells showed significantly elevated sphere-forming capacity, indicating higher cancer stem cell content, thus qualifying as suitable test system. Nucleolin-targeting 57Co-DOTA-AS1411 aptamer showed efficient internalization by cancer cells and, remarkably, at even higher efficiency by cancer stem cells. In contrast, somatostatin receptor 2 expression levels were not sufficiently high in H1299 cells to confer efficient uptake by either non-cancer stem cells or cancer stem cells. The data provides indication that the nucleolin-targeting AS1411 aptamer might be used for therapeutic delivery to non-small cell lung cancer stem cells.
Collapse
Affiliation(s)
- Sif Holmboe
- Lundbeckfonden Center of Excellence NanoCAN, University of Southern Denmark, Odense, Denmark
- Molecular Oncology, University of Southern Denmark, Odense, Denmark
| | - Pernille Lund Hansen
- Lundbeckfonden Center of Excellence NanoCAN, University of Southern Denmark, Odense, Denmark
- Molecular Oncology, University of Southern Denmark, Odense, Denmark
| | - Helge Thisgaard
- Department of Nuclear Medicine, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Ines Block
- Lundbeckfonden Center of Excellence NanoCAN, University of Southern Denmark, Odense, Denmark
- Molecular Oncology, University of Southern Denmark, Odense, Denmark
| | - Carolin Müller
- Lundbeckfonden Center of Excellence NanoCAN, University of Southern Denmark, Odense, Denmark
- Molecular Oncology, University of Southern Denmark, Odense, Denmark
| | - Niels Langkjær
- Department of Nuclear Medicine, Odense University Hospital, Odense, Denmark
| | - Poul Flemming Høilund-Carlsen
- Department of Nuclear Medicine, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | | | - Jan Mollenhauer
- Molecular Oncology, University of Southern Denmark, Odense, Denmark
- Department of Nuclear Medicine, Odense University Hospital, Odense, Denmark
| |
Collapse
|
30
|
Ji M, Jin A, Sun J, Cui X, Yang Y, Chen L, Lin Z. Clinicopathological implications of NQO1 overexpression in the prognosis of pancreatic adenocarcinoma. Oncol Lett 2017; 13:2996-3002. [PMID: 28521407 PMCID: PMC5431416 DOI: 10.3892/ol.2017.5821] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 10/21/2016] [Indexed: 01/18/2023] Open
Abstract
Nicotinamide adenine dinucleotide phosphate: quinone oxidoreductase 1 (NQO1) protects cells from oxidative damage. NQO1 has been reported to be upregulated in numerous solid tumors, suggesting a role in carcinogenesis and tumor progression. The present study attempted to investigate the clinical prognostic significance of NQO1 overexpression in pancreatic ductal adenocarcinoma (PDAC). A total of 181 tissue specimens were studied, including 126 PDAC and 55 normal pancreas specimens, which were selected for immunohistochemical staining of NQO1 protein. Immunofluorescence staining was additionally performed to identify the subcellular localization of NQO1 protein in pancreatic cancer BxPC-3 cells. The association between NQO1 overexpression and the clinical features of PDAC were evaluated by χ2 and Fisher's exact test. Overall survival of PDAC patients was calculated using Kaplan-Meier analysis. Univariate and multivariate analyses were performed using the Cox proportional hazards regression model. The NQO1 protein was mainly located in the cytoplasm and nucleus of BxPC-3 cells. The strongly positive rate of NQO1 expression in PDAC (65.9%, 83/126) was increased compared with that in normal pancreatic tissues (10.9%, 6/55). The positive rate of NQO1 protein was associated with grading, lymph node stage and tumor-node-metastasis (TNM) stage. Additionally, multivariate analysis suggested that NQO1 was a significant independent prognostic factor along with TNM stage in PDAC. In conclusion, high expression of NQO1 appears to be associated with PDAC progression, and may be an independent prognostic biomarker in PDAC.
Collapse
Affiliation(s)
- Meiying Ji
- Cancer Research Center and Department of Pathology, Yanbian University Medical College, Yanji, Jilin 133002, P.R. China
| | - Aihua Jin
- Department of Internal Medicine, Yanbian University Hospital, Yanji, Jilin 133000, P.R. China
| | - Jie Sun
- Cancer Research Center and Department of Pathology, Yanbian University Medical College, Yanji, Jilin 133002, P.R. China
| | - Xuelian Cui
- Cancer Research Center and Department of Pathology, Yanbian University Medical College, Yanji, Jilin 133002, P.R. China
| | - Yang Yang
- Cancer Research Center and Department of Pathology, Yanbian University Medical College, Yanji, Jilin 133002, P.R. China
| | - Liyan Chen
- Cancer Research Center and Department of Pathology, Yanbian University Medical College, Yanji, Jilin 133002, P.R. China.,Department of Biological Chemistry and Molecular Biology, Yanbian University Medical College, Yanji, Jilin 133002, P.R. China
| | - Zhenhua Lin
- Cancer Research Center and Department of Pathology, Yanbian University Medical College, Yanji, Jilin 133002, P.R. China
| |
Collapse
|
31
|
Chakrabarti KR, Andorko JI, Whipple RA, Zhang P, Sooklal EL, Martin SS, Jewell CM. Lipid tethering of breast tumor cells enables real-time imaging of free-floating cell dynamics and drug response. Oncotarget 2016; 7:10486-97. [PMID: 26871289 PMCID: PMC4891134 DOI: 10.18632/oncotarget.7251] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Accepted: 01/26/2016] [Indexed: 02/07/2023] Open
Abstract
Free-floating tumor cells located in the blood of cancer patients, known as circulating tumor cells (CTCs), have become key targets for studying metastasis. However, effective strategies to study the free-floating behavior of tumor cells in vitro have been a major barrier limiting the understanding of the functional properties of CTCs. Upon extracellular-matrix (ECM) detachment, breast tumor cells form tubulin-based protrusions known as microtentacles (McTNs) that play a role in the aggregation and re-attachment of tumor cells to increase their metastatic efficiency. In this study, we have designed a strategy to spatially immobilize ECM-detached tumor cells while maintaining their free-floating character. We use polyelectrolyte multilayers deposited on microfluidic substrates to prevent tumor cell adhesion and the addition of lipid moieties to tether tumor cells to these surfaces through interactions with the cell membranes. This coating remains optically clear, allowing capture of high-resolution images and videos of McTNs on viable free-floating cells. In addition, we show that tethering allows for the real-time analysis of McTN dynamics on individual tumor cells and in response to tubulin-targeting drugs. The ability to image detached tumor cells can vastly enhance our understanding of CTCs under conditions that better recapitulate the microenvironments they encounter during metastasis.
Collapse
Affiliation(s)
- Kristi R Chakrabarti
- Medical Scientist Training Program, University of Maryland School of Medicine, Baltimore, MD 21201, USA.,Graduate Program in Molecular Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA.,Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - James I Andorko
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Rebecca A Whipple
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA.,Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Peipei Zhang
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Elisabeth L Sooklal
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Stuart S Martin
- Graduate Program in Molecular Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA.,Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA.,Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Christopher M Jewell
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA.,Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA.,Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
32
|
Multi-Phenotypic subtyping of circulating tumor cells using sequential fluorescent quenching and restaining. Sci Rep 2016; 6:33488. [PMID: 27647345 PMCID: PMC5028835 DOI: 10.1038/srep33488] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 08/23/2016] [Indexed: 01/06/2023] Open
Abstract
In tissue biopsies formalin fixed paraffin embedded cancer blocks are micro-sectioned producing multiple semi-identical specimens which are analyzed and subtyped proteomically, and genomically, with numerous biomarkers. In blood based biopsies (BBBs), blood is purified for circulating tumor cells (CTCs) and clinical utility is typically limited to cell enumeration, as only 2–3 positive fluorescent markers and 1 negative marker can be used. As such, increasing the number of subtyping biomarkers on each individual CTC could dramatically enhance the clinical utility of BBBs, allowing in depth interrogation of clinically relevant CTCs. We describe a simple and inexpensive method for quenching the specific fluors of fluorescently stained CTCs followed by sequential restaining with additional biomarkers. As proof of principle a CTC panel, immunosuppression panel and stem cell panel were used to sequentially subtype individual fluorescently stained patient CTCs, suggesting a simple and universal technique to analyze multiple clinically applicable immunomarkers from BBBs.
Collapse
|
33
|
Kölbl AC, Jeschke U, Andergassen U. The Significance of Epithelial-to-Mesenchymal Transition for Circulating Tumor Cells. Int J Mol Sci 2016; 17:E1308. [PMID: 27529216 PMCID: PMC5000705 DOI: 10.3390/ijms17081308] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 08/02/2016] [Accepted: 08/04/2016] [Indexed: 12/12/2022] Open
Abstract
Epithelial to mesenchymal transition (EMT) is a process involved in embryonic development, but it also plays a role in remote metastasis formation in tumor diseases. During this process cells lose their epithelial features and adopt characteristics of mesenchymal cells. Thereby single tumor cells, which dissolve from the primary tumor, are enabled to invade the blood vessels and travel throughout the body as so called "circulating tumor cells" (CTCs). After leaving the blood stream the reverse process of EMT, the mesenchymal to epithelial transition (MET) helps the cells to seed in different tissues, thereby generating the bud of metastasis formation. As metastasis is the main reason for tumor-associated death, CTCs and the EMT process are in the focus of research in recent years. This review summarizes what was already found out about the molecular mechanisms driving EMT, the consequences of EMT for tumor cell detection, and suitable markers for the detection of CTCs which underwent EMT. The research work done in this field could open new roads towards combating cancer.
Collapse
Affiliation(s)
- Alexandra C Kölbl
- Department of Gynecology and Obstetrics, LMU Munich, Maistrasse 11, 80337 Munich, Germany.
| | - Udo Jeschke
- Department of Gynecology and Obstetrics, LMU Munich, Maistrasse 11, 80337 Munich, Germany.
| | - Ulrich Andergassen
- Department of Gynecology and Obstetrics, LMU Munich, Maistrasse 11, 80337 Munich, Germany.
| |
Collapse
|
34
|
Li Y, Deng X, Zeng X, Peng X. The Role of Mir-148a in Cancer. J Cancer 2016; 7:1233-41. [PMID: 27390598 PMCID: PMC4934031 DOI: 10.7150/jca.14616] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 05/07/2016] [Indexed: 12/16/2022] Open
Abstract
MicroRNAs (miRNAs) are highly conserved noncoding RNAs of about 19-25 nucleotides. Through specifically pairing with complementary sites in 3' untranslated regions (UTRs) of target mRNAs, they mediate post-transcriptional silencing. MicroRNAs have been implicated in many physiological processes including proliferation, differentiation, development, apoptosis, and metabolism. In recent years many studies have revealed that the aberrant expression of miRNA is closely related to oncogenesis and is now an intense field of study. Mir-148a is aberrantly expressed in various cancers and has been identified as an oncogenic or tumor suppressor with crucial roles in the molecular mechanisms of oncogenesis. In this review, we have summarized the role of mir-148a in the oncogenic pathways of gastric, liver, breast and urogenital cancers, and in neurogliocytoma oncogenesis. Studying the functional role of mir-148a is crucial in discovering novel tumor molecular markers and identifying potential therapeutic targets.
Collapse
Affiliation(s)
- Yue Li
- 1. Department of Pathology and Pathophysiology, Hunan Normal University Medical School, Changsha 410013, Hunan, China
| | - Xiyun Deng
- 1. Department of Pathology and Pathophysiology, Hunan Normal University Medical School, Changsha 410013, Hunan, China
| | - Xiaomin Zeng
- 2. Department of Statistics and Epidemiology, Public Health School, Central South University, Changsha 410078, Hunan, China
| | - Xiaoning Peng
- 1. Department of Pathology and Pathophysiology, Hunan Normal University Medical School, Changsha 410013, Hunan, China
| |
Collapse
|
35
|
Northup JRN, Deutscher SL. Cytotoxic Tumor-Targeting Peptides From In Vivo Phage Display. Comb Chem High Throughput Screen 2016; 19:370-7. [PMID: 27055748 DOI: 10.2174/1386207319666160408151423] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 12/07/2015] [Accepted: 03/04/2016] [Indexed: 11/22/2022]
Abstract
We previously utilized an in vivo peptide phage display selection technique, which included the use of detergent elution of phage from excised tumor, to obtain tumor-targeting phage with the ability to extravasate the vasculature and bind directly to prostate tumor tissue. It is hypothesized that this same in vivo phage selection technique can be used to functionally select for molecules that not only bind to cancer cells but also kill them. Here we analyzed two different in vivo phage display selected phage clones, G1 and H5, retrieved from PC-3 human prostate carcinoma xenografted tumors. First, cell de-attachment as an endpoint criterion for apoptosis and cell cycle was examined. After 2.5 hours incubation with G1 phage, PC-3 cell attachment was reduced by 23.8% and the percent of cell population in M phase reduced by 32.1%. In comparison, PC-3 cells incubated with H5 phage had a reduction of 25.0% cell attachment and 33.6% of cell population in M phase. These changes in combination with elevated caspase activation within cells in M phase, and no significant changes to G1/G0 or S phase cell populations suggest that the cytotoxic phages are targeting actively dividing PC-3 cells. Microscopic studies were also performed to further analyze the nature of cytotoxicity of these two phage clones. It was found that G1 phage induced and co- localized with tubulin based projections within apoptotic cells, while H5 phage did not. These phage may form the foundation for a new class of targeted prostate cancer therapeutic agents.
Collapse
Affiliation(s)
| | - Susan L Deutscher
- Department of Biochemistry, University of Missouri, and Harry S. Truman Veterans Memorial Hospital, 117 Schweitzer Hall, Columbia, MO 65211, USA.
| |
Collapse
|
36
|
Yang KR, Mooney SM, Zarif JC, Coffey DS, Taichman RS, Pienta KJ. Niche inheritance: a cooperative pathway to enhance cancer cell fitness through ecosystem engineering. J Cell Biochem 2015; 115:1478-85. [PMID: 24700698 PMCID: PMC4143896 DOI: 10.1002/jcb.24813] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 04/01/2014] [Indexed: 02/03/2023]
Abstract
Cancer cells can be described as an invasive species that is able to establish itself in a new environment. The concept of niche construction can be utilized to describe the process by which cancer cells terraform their environment, thereby engineering an ecosystem that promotes the genetic fitness of the species. Ecological dispersion theory can then be utilized to describe and model the steps and barriers involved in a successful diaspora as the cancer cells leave the original host organ and migrate to new host organs to successfully establish a new metastatic community. These ecological concepts can be further utilized to define new diagnostic and therapeutic areas for lethal cancers. 115: 1478–1485, 2014. © 2014 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Kimberline R Yang
- Cellular and Molecular Medicine Program, Johns Hopkins School of Medicine, Baltimore, Maryland
| | | | | | | | | | | |
Collapse
|
37
|
Sun BO, Fang Y, Li Z, Chen Z, Xiang J. Role of cellular cytoskeleton in epithelial-mesenchymal transition process during cancer progression. Biomed Rep 2015; 3:603-610. [PMID: 26405532 DOI: 10.3892/br.2015.494] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 02/20/2015] [Indexed: 02/06/2023] Open
Abstract
Currently, cancer metastases remain a major clinical problem that highlights the importance of recognition of the metastatic process in cancer diagnosis and treatment. A critical process associated with the metastasis process is the transformation of epithelial cells toward the motile mesenchymal state, a process called epithelial-mesenchymal transition (EMT). Increasing evidence suggests the crucial role of the cytoskeleton in the EMT process. The cytoskeleton is composed of the actin cytoskeleton, the microtubule network and the intermediate filaments that provide structural design and mechanical strength that is necessary for the EMT. The dynamic reorganization of the actin cytoskeleton is a prerequisite for the morphology, migration and invasion of cancer cells. The microtubule network is the cytoskeleton that provides the driving force during cell migration. Intermediate filaments are significantly rearranged, typically switching from cytokeratin-rich to vimentin-rich networks during the EMT process, accompanied by a greatly enhanced cell motility capacity. In the present review, the recent novel insights into the different cytoskeleton underlying EMT are summarized. There are numerous advances in our understanding of the fundamental role of the cytoskeleton in cancer cell invasion and migration.
Collapse
Affiliation(s)
- B O Sun
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Yantian Fang
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Zhenyang Li
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Zongyou Chen
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Jianbin Xiang
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| |
Collapse
|
38
|
Miranda P, Vimalraj S, Selvamurugan N. A feedback expression of microRNA-590 and activating transcription factor-3 in human breast cancer cells. Int J Biol Macromol 2015; 72:145-50. [DOI: 10.1016/j.ijbiomac.2014.07.051] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 07/30/2014] [Accepted: 07/31/2014] [Indexed: 12/28/2022]
|
39
|
Adams DL, Stefansson S, Haudenschild C, Martin SS, Charpentier M, Chumsri S, Cristofanilli M, Tang CM, Alpaugh RK. Cytometric characterization of circulating tumor cells captured by microfiltration and their correlation to the CellSearch(®) CTC test. Cytometry A 2014; 87:137-44. [PMID: 25515318 DOI: 10.1002/cyto.a.22613] [Citation(s) in RCA: 121] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 11/04/2014] [Accepted: 12/02/2014] [Indexed: 12/13/2022]
Abstract
Recent studies reporting hundreds, to thousands, of circulating tumor cells (CTCs) in the blood of cancer patients have raised questions regarding the prevalence of CTCs, as enumerated by the CellSearch(®) CTC Test. Although CellSearch has been shown to consistently detect clinically relevant CTCs; the ability to only capture EpCAM positive cells has led to speculation that it captures limited subsets of CTCs. In contrast, alternative approaches to CTC isolation are often cited as capturing large numbers of CTCs from patient blood. Not surprisingly the number of cells isolated by alternative approaches show poor correlations when compared to CellSearch, even when accounting for EpCAM presence or absence. In an effort to address this discrepancy, we ran an exploratory method comparison study to characterize and compare the CTC subgroups captured from duplicate blood samples from 30 breast and prostate cancer patients using a microfiltration system (CellSieve™) and CellSearch. We then categorized the CellSieve Cytokeratin(CK)+/CD45-/DAPI+ cells into five morphologically distinct subpopulations for correlative analysis. Like other filtration techniques, CellSieve isolated greater numbers of CK+/CD45- cells than CellSearch. Furthermore, analysis showed low correlation between the total CK+/CD45- cells captured by these two assays, regardless of EpCAM presence. However, subgrouping of CK+/CD45-/DAPI+ cells based on distinct cytokeratin staining patterns and nuclear morphologies elucidated a subpopulation correlative to CellSearch. Using method comparison analyses, we identified a specific CTC morphology which is highly correlative between two distinct capture methods. These data suggests that although various morphologic CTCs with similar phenotypic expressions are present in the blood of cancer patients, the clinically relevant cells isolated by CellSearch can potentially be identified using non-EpCAM dependent isolation. © 2014 The Authors. Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Daniel L Adams
- Creatv MicroTech, Inc., 11 Deer Park Dr. Monmouth Junction, New Jersey, 08852
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Overexpression of SMARCA5 correlates with cell proliferation and migration in breast cancer. Tumour Biol 2014; 36:1895-902. [PMID: 25377162 DOI: 10.1007/s13277-014-2791-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Accepted: 10/29/2014] [Indexed: 12/26/2022] Open
Abstract
SMARCA5 partners with RSF-1 to compose the RSF complex, which belongs to the ISWI family of chromatin remodelers. Recent studies referred that SMARCA5 was overexpressed in some malignant tumors. However, expression pattern and biological roles of SMARCA5 in breast cancer have not been examined. In the present study, we found that SMARCA5 was overexpressed in breast cancer specimens by immunohistochemistry. Significant association was observed between SMARCA5 overexpression and TNM stage (p = 0.0199), tumor size (p = 0.0066), high proliferation index (p = 0.0366), and poor overall survival (p = 0.0141). SMARCA5 overexpression also correlated with Rsf-1 expression levels (p = 0.0120). Furthermore, colony formation assay and Matrigel invasion assay showed that knockdown of SMARCA5 expression in MDA-MB-231 and MDA-MB-435s cell lines with high endogenous expression decreased cell proliferation and cell invasion. Flow cytometry showed knockdown of SMARCA5-arrested cell cycle. Further analysis of cell cycle and invasion-related molecules showed that SMARCA5 downregulated cyclin A, MMP2 expression and upregulated p21 expression. In conclusion, our study demonstrated that SMARCA5 was overexpressed in human breast cancers and correlated with poor prognosis. SMARCA5 contributes to breast cancer cell proliferation and invasion.
Collapse
|
41
|
Moyret-Lalle C, Ruiz E, Puisieux A. Epithelial-mesenchymal transition transcription factors and miRNAs: “Plastic surgeons” of breast cancer. World J Clin Oncol 2014; 5:311-322. [PMID: 25114847 PMCID: PMC4127603 DOI: 10.5306/wjco.v5.i3.311] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 03/18/2014] [Accepted: 06/27/2014] [Indexed: 02/06/2023] Open
Abstract
Growing evidence suggests that breast cancer cell plasticity arises due to a partial reactivation of epithelial-mesenchymal transition (EMT) programs in order to give cells pluripotency, leading to a stemness-like phenotype. A complete EMT would be a dead end program that would render cells unable to fully metastasize to distant organs. Evoking the EMT-mesenchymal-to-epithelial transition (MET) cascade promotes successful colonization of distal target tissues. It is unlikely that direct reprogramming or trans-differentiation without passing through a pluripotent stage would be the preferred mechanism during tumor progression. This review focuses on key EMT transcriptional regulators, EMT-transcription factors involved in EMT (TFs) and the miRNA pathway, which are deregulated in breast cancer, and discusses their implications in cancer cell plasticity. Cross-regulation between EMT-TFs and miRNAs, where miRNAs act as co-repressors or co-activators, appears to be a pivotal mechanism for breast cancer cells to acquire a stem cell-like state, which is implicated both in breast metastases and tumor recurrence. As a master regulator of miRNA biogenesis, the ribonuclease type III endonuclease Dicer plays a central role in EMT-TFs/miRNAs regulating networks. All these EMT-MET key regulators represent valuable new prognostic and predictive markers for breast cancer as well as promising new targets for drug-resistant breast cancers.
Collapse
|
42
|
Ginnebaugh KR, Ahmad A, Sarkar FH. The therapeutic potential of targeting the epithelial-mesenchymal transition in cancer. Expert Opin Ther Targets 2014; 18:731-45. [PMID: 24758643 DOI: 10.1517/14728222.2014.909807] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION The process of epithelial-to-mesenchymal transition (EMT) has long been advocated as a process during tumor progression and the acquisition of metastatic potential of human cancers. EMT has also been linked with resistance to cancer therapies. AREAS COVERED Basic research has provided evidence connecting EMT to increased invasion, angiogenesis and metastasis of cancer cells. A number of signaling pathways such as notch, wnt, hedgehog and PI3K-AKT, and various other individual factors therein, have been intricately connected to the onset of EMT. Here, we provide latest updates on the evidences that further highlight an association between various signaling pathways and EMT, with a focus on therapeutic targets that may have the potential to reverse EMT. EXPERT OPINION Our understanding of EMT and its underlying causes is rapidly evolving and a number of putative targets have been identified. It is crucial, now than ever before, to design novel translational and clinical studies for the benefit of advanced stage cancer patients with metastatic disease.
Collapse
Affiliation(s)
- Kevin R Ginnebaugh
- Karmanos Cancer Institute, Wayne State University School of Medicine, Department of Pathology , Detroit, MI 48201 , USA
| | | | | |
Collapse
|
43
|
Zhang J, Yin L, Wu J, Zhang Y, Xu T, Ma R, Cao H, Tang J. Detection of serum VEGF and MMP-9 levels by Luminex multiplexed assays in patients with breast infiltrative ductal carcinoma. Exp Ther Med 2014; 8:175-180. [PMID: 24944618 PMCID: PMC4061234 DOI: 10.3892/etm.2014.1685] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 03/27/2014] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to assess the effect of the combined detection of serum vascular endothelial growth factor (VEGF) and matrix metalloproteinase-9 (MMP-9) by Luminex multiplexed assays for the diagnosis, treatment and prognosis of breast cancer. Preoperative levels of serum VEGF and MMP-9 were detected via a lipid chip-based method in 301 breast cancer cases, 83 breast fibroadenoma cases and 40 healthy adults. Postoperative levels of VEGF and MMP-9 were also detected in 118 breast cancer cases. The levels of serum VEGF and MMP-9 in patients with breast infiltrative ductal carcinoma (IDC) were higher than those in the breast fibroadenoma and healthy control groups (P<0.05); there was no statistically significant difference between the breast fibroadenoma and healthy groups (P>0.05). The levels of VEGF and MMP-9 were shown to correlate with the clinical stage, tumor size and the lymph node metastasis status. However, the levels were not associated with age or gender (P>0.05). In addition, the serum level of MMP-9 exhibited a significantly correlation with the VEGF level (r=0.601, P<0.001). Subgroup analysis revealed that in patients with IDC, serum levels of VEGF and MMP-9 prior to surgery were significantly higher than those following surgery (P<0.05). Therefore, the serum levels of VEGF and MMP-9 can be used as markers for the diagnosis of breast IDC and may also be valuable for the prediction of lymph nodes metastasis.
Collapse
Affiliation(s)
- Junying Zhang
- Department of Oncology, Xuzhou Medical College, Xuzhou, Jiangsu 221004, P.R. China
| | - Li Yin
- Department of Radiation Oncology, Nanjing Medical University Affiliated Cancer Hospital, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210009, P.R. China
| | - Jianzhong Wu
- Research Center for Clinical Oncology, Nanjing Medical University Affiliated Cancer Hospital, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210009, P.R. China
| | - Ye Zhang
- Research Center for Clinical Oncology, Nanjing Medical University Affiliated Cancer Hospital, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210009, P.R. China
| | - Tao Xu
- Department of General Surgery, Nanjing Medical University Affiliated Cancer Hospital, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210009, P.R. China
| | - Rong Ma
- Research Center for Clinical Oncology, Nanjing Medical University Affiliated Cancer Hospital, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210009, P.R. China
| | - Haixia Cao
- Research Center for Clinical Oncology, Nanjing Medical University Affiliated Cancer Hospital, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210009, P.R. China
| | - Jinhai Tang
- Department of General Surgery, Nanjing Medical University Affiliated Cancer Hospital, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210009, P.R. China
| |
Collapse
|
44
|
Yang Y, Zhang Y, Wu Q, Cui X, Lin Z, Liu S, Chen L. Clinical implications of high NQO1 expression in breast cancers. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2014; 33:14. [PMID: 24499631 PMCID: PMC3944477 DOI: 10.1186/1756-9966-33-14] [Citation(s) in RCA: 135] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2014] [Accepted: 01/27/2014] [Indexed: 01/01/2023]
Abstract
Background NAD (P) H: quinone oxidoreductase 1 (NQO1) is a xenobiotic metabolizing enzyme that detoxifies chemical stressors and antioxidants, providing cytoprotection in normal tissues. However, high-level expression of NQO1 has been correlated with numerous human malignancies, suggesting a role in carcinogenesis and tumor progression. This study aimed to explore the clinicopathological significance of NQO1 and as a prognostic determinant in breast cancer. Methods A total of 176 breast cancer patients with strict follow-up, 45 ductal carcinoma in situ (DCIS), 22 hyperplasia and 52 adjacent non-tumor breast tissues were selected for immunohistochemical staining of NQO1 protein. Immunofluorescence staining was also performed to detect the subcellular localization of NQO1 protein in MCF-7 breast cancer cells. Eight fresh breast cancers paired with adjacent non-tumor tissues were quantified using real time RT-PCR (qRT-PCR) and western blot. The correlations between NQO1 overexpression and the clinical features of breast cancer were evaluated using chi-square test and Fisher’s exact tests. The survival rate was calculated using the Kaplan–Meier method, and the relationship between prognostic factors and patient survival was also analyzed by the Cox proportional hazards models. Results NQO1 protein showed a mainly cytoplasmic staining pattern in breast cancer. The strongly positive rate of NQO1 protein was 61.9% (109/176) in breast cancer, and was significantly higher than in DCIS (31.1%, 14/45), hyperplasia tissues (13.6%, 3/22) and adjacent non-tumor tissues (13.5%, 7/52). High-level expression of NQO1 protein was correlated with late clinical stage, poor differentiation, lymph node metastasis, Her2 expression and disease-free and 10-year overall survival rates in breast cancer. Moreover, multivariate analysis suggested that NQO1 emerged as a significant independent prognostic factor along with clinical stage and Her2 expression status in patients with breast cancer. Conclusions High-level expression of NQO1 appears to be associated with breast cancer progression, and may be a potential biomarker for poor prognostic evaluation of breast cancers.
Collapse
Affiliation(s)
| | | | | | | | | | - Shuangping Liu
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji 133002, China.
| | | |
Collapse
|