1
|
Zhao T, Zhang Z, Chen Z, Xu G, Wang Y, Wang F. Biological functions of 5-methylcytosine RNA-binding proteins and their potential mechanisms in human cancers. Front Oncol 2025; 15:1534948. [PMID: 39990690 PMCID: PMC11842269 DOI: 10.3389/fonc.2025.1534948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 01/21/2025] [Indexed: 02/25/2025] Open
Abstract
The 5-methylcytosine (m5C) modification is a crucial epigenetic RNA modification, which is involved in the post-transcriptional regulation of genes. It plays an important role in various biological processes, including cell metabolism, growth, apoptosis, and tumorigenesis. By affecting the proliferation, migration, invasion, and drug sensitivity of tumor cells, m5C methylation modification plays a vital part in the initiation and progression of tumors and is closely associated with the poor tumor prognosis. m5C-related proteins are categorized into three functional groups: m5C methyltransferases (m5C writers), m5C demethylases (m5C erasers), and m5C methyl-binding proteins (m5C readers). This paper introduces several common methodologies for detecting m5C methylation; and reviews the molecular structure and biological functions of m5C readers, including ALYREF, YBX1, YBX2, RAD52, YTHDF2, FMRP, and SRSF2. It further summarizes their roles and regulatory mechanisms in tumors, offering novel targets and insights for tumor treatment.
Collapse
Affiliation(s)
| | | | | | | | | | - Fang Wang
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
2
|
Venkatesalu S, Dilliyappan S, Satish Kumar A, Palaniyandi T, Baskar G, Ravi M, Sivaji A. Prospectives and retrospectives of microfluidics devices and lab-on-A-chip emphasis on cancer. Clin Chim Acta 2024; 552:117646. [PMID: 38000458 DOI: 10.1016/j.cca.2023.117646] [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: 08/26/2023] [Revised: 11/06/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023]
Abstract
Microfluidics is a science and technology that deals with the concept of "less sample-to-more precision" enabling portable device development via fabrication for in vitro analysis. On evolution, microfluidic system lead to the development of Organ-on-chip where recapitulation of organ's functionality and pathophysiological response can be performed under controlled environment. Further microfluidic-based "Lab-on-chip" device, a versatile innovation credited for its number of parameters that has capability to leverage next-generation companion of medicines. This emulsion science has enormous practise in the field of regenerative medicine, drug screening, medical diagnosis and therapy for accuracy in results. In this era of personalized medicine, getting precise tools for applying these theranostics is crucial. Oncological theranostics create a new gateway to develop precision in personalized medicine for cancer, where microfluidic chips are involved in diagnosis and therapy of various cancers using biomarkers for thyroid, lung cancers, and assay based for breast, circulating tumor cells and colorectal cancers and nanoparticles for ovarian cancer. This review shows more comprehensive approach to the state of art with respect to microfluidic devices in cancer theranostics.
Collapse
Affiliation(s)
- Sneha Venkatesalu
- Department of Biotechnology, Dr. M.G.R. Educational and Research Institute, Chennai, India
| | | | - Avanthika Satish Kumar
- Department of Biotechnology, Dr. M.G.R. Educational and Research Institute, Chennai, India
| | - Thirunavukkarasu Palaniyandi
- Department of Biotechnology, Dr. M.G.R. Educational and Research Institute, Chennai, India; Department of Anatomy, Biomedical Research Unit and Laboratory Animal Centre, Saveetha Dental College and Hospital, SIMATS, Saveetha University, Chennai, India.
| | - Gomathy Baskar
- Department of Biotechnology, Dr. M.G.R. Educational and Research Institute, Chennai, India
| | - Maddaly Ravi
- Department of Human Genetics, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| | - Asha Sivaji
- Department of Biochemistry, DKM College for Women, Vellore, India
| |
Collapse
|
3
|
Zhang T, Cao W, Sun H, Yu D, Zhong C. Diallyl Trisulfide Suppresses the Renal Cancer Stem-like Cell Properties via Nanog. Nutr Cancer 2023; 75:971-979. [PMID: 36562732 DOI: 10.1080/01635581.2022.2156553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cancer stem-like cells (CSCs), which play an important role in tumor initiation and progression, have been identified in many cancers. Diallyl trisulfide (DATS) is an organosulfur compound extracted from garlic with anticancer activities. Nanog is a transcription factor responsible for maintaining the stemness of CSCs, but its role in the DATS-induced attenuation of renal CSC properties is unknown. In this study, renal CSCs were enriched from human renal cancer cell lines 786-O and ACHN cultured in a serum-free medium (SFM). The properties of CSCs were analyzed by evaluating the ability of the cells in sphere formation and measuring the expression of stem cell markers. We found that downregulation of Nanog inhibited renal CSC properties. DATS suppressed renal CSC activities by reducing tumorsphere formation, decreasing stem cell markers including Nanog, CD44, ALDH1A1, and Oct4, inhibiting cell proliferation and promoting apoptosis. We further revealed that overexpression of Nanog reversed the suppressive effects of DATS on renal CSCs. Taken together, our results demonstrated that DATS inhibited renal CSCs by suppressing Nanog. These novel findings suggested that, through Nanog targeting, DATS can potentially be used as an anti-tumor agent for renal cancer.
Collapse
Affiliation(s)
- Taotao Zhang
- Health Management Center , The Second Affiliated Hospital of Anhui Medical University, Hefei, China.,Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wanshuang Cao
- Cancer Research Division, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Hongliang Sun
- Department of Urology, Affiliated Taikang Xianlin Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Dexin Yu
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Caiyun Zhong
- Cancer Research Division, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| |
Collapse
|
4
|
Vasefifar P, Motafakkerazad R, Maleki LA, Najafi S, Ghrobaninezhad F, Najafzadeh B, Alemohammad H, Amini M, Baghbanzadeh A, Baradaran B. Nanog, as a key cancer stem cell marker in tumor progression. Gene X 2022; 827:146448. [PMID: 35337852 DOI: 10.1016/j.gene.2022.146448] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 02/16/2022] [Accepted: 03/18/2022] [Indexed: 12/20/2022] Open
Abstract
Cancer stem cells (CSCs) are a small population of malignant cells that induce tumor onset and development. CSCs share similar features with normal stem cells in the case of self-renewal and differentiation. They also contribute to chemoresistance and metastasis of cancer cells, leading to therapeutic failure. To identify CSCs, multiple cell surface markers have been characterized, including Nanog, which is found at high levels in different cancers. Recent studies have revealed that Nanog upregulation has a substantial association with the advanced stages and poor prognosis of malignancies, playing a pivotal role through tumorigenesis of multiple human cancers, including leukemia, liver, colorectal, prostate, ovarian, lung, head and neck, brain, pancreatic, gastric and breast cancers. Nanog through different signaling pathways, like JAK/STAT and Wnt/β-catenin pathways, induces stemness, self-renewal, metastasis, invasiveness, and chemoresistance of cancer cells. Some of these signaling pathways are common in various types of cancers, but some have been found in one or two cancers. Therefore, this review aimed to focus on the function of Nanog in multiple cancers based on recent studies surveying the suitable approaches to target Nanog and inhibit CSCs residing in tumors to gain favorable results from cancer treatments.
Collapse
Affiliation(s)
- Parisa Vasefifar
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | - Souzan Najafi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Basira Najafzadeh
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Hajar Alemohammad
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Mohammad Amini
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
5
|
Jiang D, Qiu T, Peng J, Li S, Tala, Ren W, Yang C, Wen Y, Chen CH, Sun J, Wu Y, Liu R, Zhou J, Wu K, Liu W, Mao X, Zhou Z, Chen C. YB-1 is a positive regulator of KLF5 transcription factor in basal-like breast cancer. Cell Death Differ 2022; 29:1283-1295. [PMID: 35022570 PMCID: PMC9177637 DOI: 10.1038/s41418-021-00920-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 11/30/2021] [Accepted: 12/02/2021] [Indexed: 12/25/2022] Open
Abstract
Y-box binding protein 1 (YB-1) is a well-known oncogene highly expressed in various cancers, including basal-like breast cancer (BLBC). Beyond its role as a transcription factor, YB-1 is newly defined as an epigenetic regulator involving RNA 5-methylcytosine. However, its specific targets and pro-cancer functions are poorly defined. Here, based on clinical database, we demonstrate a positive correlation between Kruppel-like factor 5 (KLF5) and YB-1 expression in breast cancer patients, but a negative correlation with that of Dachshund homolog 1 (DACH1). Mechanistically, YB-1 enhances KLF5 expression not only through transcriptional activation that can be inhibited by DACH1, but also by stabilizing KLF5 mRNA in a RNA 5-methylcytosine modification-dependent manner. Additionally, ribosomal S6 kinase 2 (RSK2) mediated YB-1 phosphorylation at Ser102 promotes YB-1/KLF5 transcriptional complex formation, which co-regulates the expression of BLBC specific genes, Keratin 16 (KRT16) and lymphocyte antigen 6 family member D (Ly6D), to promote cancer cell proliferation. The RSK inhibitor, LJH685, suppressed BLBC cell tumourigenesis in vivo by disturbing YB-1-KLF5 axis. Our data suggest that YB-1 positively regulates KLF5 at multiple levels to promote BLBC progression. The novel RSK2-YB-1-KLF5-KRT16/Ly6D axis provides candidate diagnostic markers and therapeutic targets for BLBC.
Collapse
Affiliation(s)
- Dewei Jiang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- Kunming College of Lifesciences, University of Chinese Academy Sciences, Kunming, China
| | - Ting Qiu
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- Kunming College of Lifesciences, University of Chinese Academy Sciences, Kunming, China
| | - Junjiang Peng
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Siyuan Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Tala
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Wenlong Ren
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- College of Life Sciences, China University of Science and Technology, Hefei, Anhui, China
| | - Chuanyu Yang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Yi Wen
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Chuan-Huizi Chen
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, China
| | - Jian Sun
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- Kunming College of Lifesciences, University of Chinese Academy Sciences, Kunming, China
| | - Yingying Wu
- The First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Rong Liu
- The First Affiliated Hospital, Peking University, Beijing, China
| | - Jun Zhou
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Wen Liu
- School of Pharmaceutical Science, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, China
| | - Xiaoyun Mao
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China.
| | - Zhongmei Zhou
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.
| | - Ceshi Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.
- Kunming College of Lifesciences, University of Chinese Academy Sciences, Kunming, China.
- KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.
| |
Collapse
|
6
|
YB-1 as an Oncoprotein: Functions, Regulation, Post-Translational Modifications, and Targeted Therapy. Cells 2022; 11:cells11071217. [PMID: 35406781 PMCID: PMC8997642 DOI: 10.3390/cells11071217] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/26/2022] [Accepted: 04/02/2022] [Indexed: 02/04/2023] Open
Abstract
Y box binding protein 1 (YB-1) is a protein with a highly conserved cold shock domain (CSD) that also belongs to the family of DNA- and RNA-binding proteins. YB-1 is present in both the nucleus and cytoplasm and plays versatile roles in gene transcription, RNA splicing, DNA damage repair, cell cycle progression, and immunity. Cumulative evidence suggests that YB-1 promotes the progression of multiple tumor types and serves as a potential tumor biomarker and therapeutic target. This review comprehensively summarizes the emerging functions, mechanisms, and regulation of YB-1 in cancers, and further discusses targeted strategies.
Collapse
|
7
|
Najafzadeh B, Motafakkerazad R, Najafi S, Amini M, Alemohammad H, Vasefifar P, Baradaran B. Nanog suppression enhanced the chemosensitivity of Human Non-Small-Cell Lung Cancer cells to Cisplatin and inhibited cell migration. Pathol Res Pract 2022; 233:153869. [DOI: 10.1016/j.prp.2022.153869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 03/21/2022] [Accepted: 03/30/2022] [Indexed: 11/25/2022]
|
8
|
Lettau K, Khozooei S, Kosnopfel C, Zips D, Schittek B, Toulany M. Targeting the Y-box Binding Protein-1 Axis to Overcome Radiochemotherapy Resistance in Solid Tumors. Int J Radiat Oncol Biol Phys 2021; 111:1072-1087. [PMID: 34166770 DOI: 10.1016/j.ijrobp.2021.06.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 06/07/2021] [Accepted: 06/10/2021] [Indexed: 12/18/2022]
Abstract
Multifunctional Y-box binding protein-1 (YB-1) is highly expressed in different human solid tumors and is involved in various cellular processes. DNA damage is the major mechanism by which radiochemotherapy (RCT) induces cell death. On induction of DNA damage, a multicomponent signal transduction network, known as the DNA damage response, is activated to induce cell cycle arrest and initiate DNA repair, which protects cells against damage. YB-1 regulates nearly all cancer hallmarks described to date by participating in DNA damage response, gene transcription, mRNA splicing, translation, and tumor stemness. YB-1 lacks kinase activity, and p90 ribosomal S6 kinase and AKT are the key kinases within the RAS/mitogen-activated protein kinase and phosphoinositide 3-kinase pathways that directly activate YB-1. Thus, the molecular targeting of ribosomal S6 kinase and AKT is thought to be the most effective strategy for blocking the cellular function of YB-1 in human solid tumors. In this review, after describing the prosurvival effect of YB-1 with a focus on DNA damage repair and cancer cell stemness, clinical evidence will be provided indicating an inverse correlation between YB-1 expression and the treatment outcome of solid tumors after RCT. In the interest of being concise, YB-1 signaling cascades will be briefly discussed and the current literature on YB-1 posttranslational modifications will be summarized. Finally, the current status of targeting the YB-1 axis, especially in combination with RCT, will be highlighted.
Collapse
Affiliation(s)
- Konstanze Lettau
- Division of Radiobiology and Molecular Environmental Research, Department of Radiation Oncology, University of Tübingen, Tübingen Germany; German Cancer Consortium (DKTK), partner site Tübingen, and German Cancer Research Center (DKFZ) Heidelberg, Germany
| | - Shayan Khozooei
- Division of Radiobiology and Molecular Environmental Research, Department of Radiation Oncology, University of Tübingen, Tübingen Germany; German Cancer Consortium (DKTK), partner site Tübingen, and German Cancer Research Center (DKFZ) Heidelberg, Germany
| | - Corinna Kosnopfel
- Department of Dermatology, University Hospital Würzburg, Würzburg, Germany
| | - Daniel Zips
- Division of Radiobiology and Molecular Environmental Research, Department of Radiation Oncology, University of Tübingen, Tübingen Germany; German Cancer Consortium (DKTK), partner site Tübingen, and German Cancer Research Center (DKFZ) Heidelberg, Germany
| | - Birgit Schittek
- Department of Dermatology, Division of Dermatooncology, Eberhard-Karls-Universität, Tübingen, Tübingen, Germany
| | - Mahmoud Toulany
- Division of Radiobiology and Molecular Environmental Research, Department of Radiation Oncology, University of Tübingen, Tübingen Germany; German Cancer Consortium (DKTK), partner site Tübingen, and German Cancer Research Center (DKFZ) Heidelberg, Germany.
| |
Collapse
|
9
|
Li S, Wang H, Ma R, Wang L. Schisandrin B inhibits epithelial‑mesenchymal transition and stemness of large‑cell lung cancer cells and tumorigenesis in xenografts via inhibiting the NF‑κB and p38 MAPK signaling pathways. Oncol Rep 2021; 45:115. [PMID: 33907830 PMCID: PMC8107653 DOI: 10.3892/or.2021.8066] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 01/29/2021] [Indexed: 01/06/2023] Open
Abstract
Lung cancer is one of the most common types of cancer in the world, resulting in numerous cancer‑associated deaths. The properties of cancer stem cells (CSCs) are important for the initiation and deterioration of lung cancer. Schisandrin B (SchB), an active compound extracted from Schisandra chinensis, exerts anticancer effects in various malignancies, including lung cancer. Nevertheless, the potential of SchB in epithelial‑mesenchymal transition (EMT) and CSC features of large‑cell lung cancer remains unclear. The present study established cancer stem‑like cells derived from large‑cell lung cancer cells, NCI‑H460 and H661, and revealed that SchB inhibited the viability of cancer stem‑like cells at concentrations of ≥40 µmol/l. Moreover, SchB prominently inhibited cell migration, invasion and EMT. Sphere‑forming assays and western blotting demonstrated that the stemness of cancer stem‑like cells was alleviated by SchB treatment. Mechanistically, the current findings revealed that SchB contributed to the suppression of the NF‑κB and p38 MAPK signaling pathways. Notably, further results revealed that the malignant behaviors of NCI‑H460‑CSCs induced by the activation of the NF‑κB and p38 MAPK signaling pathways were suppressed by SchB treatment. Consistently, the inhibitory role of SchB in EMT and CSC activities, as well as in the activation of the NF‑κB and p38 MAPK signaling pathways, was confirmed in vivo. In conclusion, the present study demonstrated that SchB exerted inhibitory effects on large‑cell lung cancer cells via targeting the NF‑κB and p38 MAPK signaling pathways, suggesting that SchB may act as a potential therapeutic drug for large‑cell lung cancer.
Collapse
Affiliation(s)
- Shuping Li
- Department of Thoracic and Cardiac Surgery, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Hong Wang
- Department of Thoracic and Cardiac Surgery, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Ruidong Ma
- Department of Thoracic and Cardiac Surgery, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Li Wang
- Department of Anesthesiology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| |
Collapse
|
10
|
Zhang T, Sun H, Liu R, Cao W, Zhang T, Li E, Sun X, Wu W, Yu D, Zhong C. Nanog mediates tobacco smoke-induced enhancement of renal cancer stem cell properties. ENVIRONMENTAL TOXICOLOGY 2020; 35:1274-1283. [PMID: 32649042 DOI: 10.1002/tox.22992] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 06/05/2020] [Accepted: 06/06/2020] [Indexed: 06/11/2023]
Abstract
Nanog plays an important role in the regulation of cancer stem cells (CSCs) which participate in tumorgenesis and progression. In renal cancer, tobacco smoke (TS) is considered a major risk factor. However, the molecular mechanism by which TS induces the development of renal CSC properties remains largely unknown. In this study, we showed that the level of Nanog was elevated in renal cell carcinoma (RCC) patients with a smoking history, and that Nanog overexpression promoted the traits of CSCs in renal cancer. We further demonstrated that a 8-week exposure of TS enhanced the formation of renal tumorspheres, increased the population of CD133-positive cells, and stimulated the expression of Nanog and CSC markers. In addition, TS was found to play a role in accelerating the cell growth transition from G1 to S phase in renal CSCs. Finally, we demonstrated that the TS-induced effects in renal CSCs could be reversed through the downregulation of Nanog. Our results suggested that Nanog plays a role in mediating TS-induced renal CSC properties. This study may provide new insights into the molecular mechanism of TS-related renal tumorigenesis, which can contribute to the future development of therapeutics for renal cancer.
Collapse
Affiliation(s)
- Taotao Zhang
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hongliang Sun
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Rui Liu
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wanshuang Cao
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Tao Zhang
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Enlai Li
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xianchao Sun
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wangyu Wu
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Dexin Yu
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Caiyun Zhong
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, China
| |
Collapse
|
11
|
Takahashi S, Asada K, Takasawa K, Shimoyama R, Sakai A, Bolatkan A, Shinkai N, Kobayashi K, Komatsu M, Kaneko S, Sese J, Hamamoto R. Predicting Deep Learning Based Multi-Omics Parallel Integration Survival Subtypes in Lung Cancer Using Reverse Phase Protein Array Data. Biomolecules 2020; 10:1460. [PMID: 33086649 PMCID: PMC7603376 DOI: 10.3390/biom10101460] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 10/13/2020] [Accepted: 10/18/2020] [Indexed: 12/13/2022] Open
Abstract
Mortality attributed to lung cancer accounts for a large fraction of cancer deaths worldwide. With increasing mortality figures, the accurate prediction of prognosis has become essential. In recent years, multi-omics analysis has emerged as a useful survival prediction tool. However, the methodology relevant to multi-omics analysis has not yet been fully established and further improvements are required for clinical applications. In this study, we developed a novel method to accurately predict the survival of patients with lung cancer using multi-omics data. With unsupervised learning techniques, survival-associated subtypes in non-small cell lung cancer were first detected using the multi-omics datasets from six categories in The Cancer Genome Atlas (TCGA). The new subtypes, referred to as integration survival subtypes, clearly divided patients into longer and shorter-surviving groups (log-rank test: p = 0.003) and we confirmed that this is independent of histopathological classification (Chi-square test of independence: p = 0.94). Next, an attempt was made to detect the integration survival subtypes using only one categorical dataset. Our machine learning model that was only trained on the reverse phase protein array (RPPA) could accurately predict the integration survival subtypes (AUC = 0.99). The predicted subtypes could also distinguish between high and low risk patients (log-rank test: p = 0.012). Overall, this study explores novel potentials of multi-omics analysis to accurately predict the prognosis of patients with lung cancer.
Collapse
Affiliation(s)
- Satoshi Takahashi
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, 1-4-1 Nihonbashi, Chuo-ku, Tokyo 103-0027, Japan; (S.T.); (K.T.); (N.S.); (K.K.); (M.K.)
- Division of Molecular Modification and Cancer Biology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; (R.S.); (A.S.); (A.B.); (S.K.); (J.S.)
| | - Ken Asada
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, 1-4-1 Nihonbashi, Chuo-ku, Tokyo 103-0027, Japan; (S.T.); (K.T.); (N.S.); (K.K.); (M.K.)
- Division of Molecular Modification and Cancer Biology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; (R.S.); (A.S.); (A.B.); (S.K.); (J.S.)
| | - Ken Takasawa
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, 1-4-1 Nihonbashi, Chuo-ku, Tokyo 103-0027, Japan; (S.T.); (K.T.); (N.S.); (K.K.); (M.K.)
- Division of Molecular Modification and Cancer Biology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; (R.S.); (A.S.); (A.B.); (S.K.); (J.S.)
| | - Ryo Shimoyama
- Division of Molecular Modification and Cancer Biology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; (R.S.); (A.S.); (A.B.); (S.K.); (J.S.)
| | - Akira Sakai
- Division of Molecular Modification and Cancer Biology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; (R.S.); (A.S.); (A.B.); (S.K.); (J.S.)
| | - Amina Bolatkan
- Division of Molecular Modification and Cancer Biology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; (R.S.); (A.S.); (A.B.); (S.K.); (J.S.)
| | - Norio Shinkai
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, 1-4-1 Nihonbashi, Chuo-ku, Tokyo 103-0027, Japan; (S.T.); (K.T.); (N.S.); (K.K.); (M.K.)
- Division of Molecular Modification and Cancer Biology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; (R.S.); (A.S.); (A.B.); (S.K.); (J.S.)
| | - Kazuma Kobayashi
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, 1-4-1 Nihonbashi, Chuo-ku, Tokyo 103-0027, Japan; (S.T.); (K.T.); (N.S.); (K.K.); (M.K.)
- Division of Molecular Modification and Cancer Biology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; (R.S.); (A.S.); (A.B.); (S.K.); (J.S.)
| | - Masaaki Komatsu
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, 1-4-1 Nihonbashi, Chuo-ku, Tokyo 103-0027, Japan; (S.T.); (K.T.); (N.S.); (K.K.); (M.K.)
- Division of Molecular Modification and Cancer Biology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; (R.S.); (A.S.); (A.B.); (S.K.); (J.S.)
| | - Syuzo Kaneko
- Division of Molecular Modification and Cancer Biology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; (R.S.); (A.S.); (A.B.); (S.K.); (J.S.)
| | - Jun Sese
- Division of Molecular Modification and Cancer Biology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; (R.S.); (A.S.); (A.B.); (S.K.); (J.S.)
- Humanome Lab, 2-4-10 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Ryuji Hamamoto
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, 1-4-1 Nihonbashi, Chuo-ku, Tokyo 103-0027, Japan; (S.T.); (K.T.); (N.S.); (K.K.); (M.K.)
- Division of Molecular Modification and Cancer Biology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; (R.S.); (A.S.); (A.B.); (S.K.); (J.S.)
| |
Collapse
|
12
|
Bates M, Boland A, McDermott N, Marignol L. YB-1: The key to personalised prostate cancer management? Cancer Lett 2020; 490:66-75. [PMID: 32681926 DOI: 10.1016/j.canlet.2020.07.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 06/30/2020] [Accepted: 07/09/2020] [Indexed: 12/14/2022]
Abstract
Y-box-binding protein 1 (YB-1) is a DNA/RNA binding protein increasingly implicated in the regulation of cancer cell biology. Normally located in the cytoplasm, nuclear localisation in prostate cancer is associated with more aggressive, potentially treatment-resistant disease. This is attributed to the ability of YB-1 to act as a transcription factor for various target genes associated with androgen receptor signalling, survival, DNA repair, proliferation, invasion, differentiation, angiogenesis and hypoxia. This review aims to examine the clinical potential of YB-1 in the detection and therapeutic management of prostate cancer.
Collapse
Affiliation(s)
- Mark Bates
- Translational Radiobiology and Molecular Oncology Group, Applied Radiation Therapy Trinity, Discipline of Radiation Therapy, Trinity College Dublin, Dublin 2, Ireland
| | - Anna Boland
- Translational Radiobiology and Molecular Oncology Group, Applied Radiation Therapy Trinity, Discipline of Radiation Therapy, Trinity College Dublin, Dublin 2, Ireland
| | - Niamh McDermott
- Translational Radiobiology and Molecular Oncology Group, Applied Radiation Therapy Trinity, Discipline of Radiation Therapy, Trinity College Dublin, Dublin 2, Ireland
| | - Laure Marignol
- Translational Radiobiology and Molecular Oncology Group, Applied Radiation Therapy Trinity, Discipline of Radiation Therapy, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
13
|
Cui Y, Li F, Xie Q, Zhao S, Guo T, Guo P, Hu S, Hao J, Tian C, Yu W, Li Z, Fang L, Zhao L, Chen M, Wu T, Gu C. YBX1 mediates autophagy by targeting p110β and decreasing the sensitivity to cisplatin in NSCLC. Cell Death Dis 2020; 11:476. [PMID: 32561752 PMCID: PMC7305216 DOI: 10.1038/s41419-020-2555-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 04/21/2020] [Accepted: 04/22/2020] [Indexed: 01/26/2023]
Abstract
Y-box binding protein 1 (YBX1) is involved in the development of multiple types of tumors. However, the relationship between YBX1 and autophagy in non-small cell lung cancer (NSCLC) remains unclear. In this study, we analyzed the expression and clinical significance of YBX1 and markers of autophagy (LC3I/II) in NSCLC and examined their roles in regulating sensitivity to cisplatin in NSCLC. The retrospective analysis of patients with NSCLC indicated that YBX1 was positively correlated with autophagy. Increased levels of YBX1 or autophagy also observed in NSCLC cells compared with those in 16HBE cells. Compared to the controls, the knockdown of YBX1 expression suppressed autophagy, increased drug sensitivity and promoted apoptosis in response to cisplatin in NSCLC cells by targeting the p110β promoter and inhibiting p110β/Vps34/beclin1 signaling pathways. We also demonstrated in an in vivo study that the overexpressed YBX1 effectively increased NSCLC growth and progression and decreased the sensitivity to cisplatin by inducing autophagy in a xenograft tumor model, and these effects were concomitant with the increasing of p110β and beclin1 expression. Collectively, these results show that YBX1 plays an essential role in autophagy in NSCLC.
Collapse
Affiliation(s)
- Yanwei Cui
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, 116011, Dalian, China
- Departments of Respiratory Medicine, Zhongshan Hospital, Dalian Univerdity, 116011, Dalian, China
| | - Fengzhou Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, 116011, Dalian, China
| | - Qiang Xie
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, 116011, Dalian, China
| | - Shilei Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, 116011, Dalian, China
| | - Tao Guo
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, 116011, Dalian, China
| | - Ping Guo
- Institute of Cancer Stem Cell, Dalian Medical University, 116011, Dalian, China
| | - Sheng Hu
- Institute of Cancer Stem Cell, Dalian Medical University, 116011, Dalian, China
| | - Jiaojiao Hao
- Institute of Cancer Stem Cell, Dalian Medical University, 116011, Dalian, China
| | - Chunfang Tian
- Institute of Cancer Stem Cell, Dalian Medical University, 116011, Dalian, China
| | - Wendan Yu
- Institute of Cancer Stem Cell, Dalian Medical University, 116011, Dalian, China
| | - Zhuoshi Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, 116011, Dalian, China
| | - Lei Fang
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, 116011, Dalian, China
| | - Lei Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, 116011, Dalian, China
| | - Manyu Chen
- Institute of Cancer Stem Cell, Dalian Medical University, 116011, Dalian, China
| | - Taihua Wu
- Departments of Respiratory Medicine, The First Affiliated Hospital of Dalian Medical University, 116011, Dalian, China.
| | - Chundong Gu
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, 116011, Dalian, China.
| |
Collapse
|
14
|
Xu X, Jiang Z, Wang J, Ren Y, Wu A. Microfluidic applications on circulating tumor cell isolation and biomimicking of cancer metastasis. Electrophoresis 2020; 41:933-951. [PMID: 32144938 DOI: 10.1002/elps.201900402] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 02/20/2020] [Accepted: 02/28/2020] [Indexed: 02/02/2023]
Abstract
The prognosis of malignant tumors is challenged by insufficient means to effectively detect tumors at early stage. Liquid biopsy using circulating tumor cells (CTCs) as biomarkers demonstrates a promising solution to tackle the challenge, because CTCs play a critical role in cancer metastatic process via intravasation, circulation, extravasation, and formation of secondary tumor. However, the effectiveness of the solution is compromised by rarity, heterogeneity, and vulnerability associated with CTCs. Among a plethora of novel approaches for CTC isolation and enrichment, microfluidics leads to isolation and detection of CTCs in a cost-effective and operation-friendly way. Development of microfluidics also makes it feasible to model the cancer metastasis in vitro using a microfluidic system to mimick the in vivo microenvironment, thereby enabling analysis and monitor of tumor metastasis. This paper aims to review the latest advances for exploring the dual-roles microfluidics has played in early cancer diagnosis via CTC isolation and investigating the role of CTCs in cancer metastasis; the merits and drawbacks for dominating microfluidics-based CTC isolation methods are discussed; biomimicking cancer metastasis using microfluidics are presented with example applications on modelling of tumor microenvironment, tumor cell dissemination, tumor migration, and tumor angiogenesis. The future perspectives and challenges are discussed.
Collapse
Affiliation(s)
- Xiawei Xu
- Cixi Institute of Biomedical Engineering, CAS Key Laboratory of Magnetic Materials and Devices, & Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, P. R. China.,Research Group for Fluids and Thermal Engineering, University of Nottingham Ningbo China, Ningbo, P. R. China.,Department of Mechanical, Materials and Manufacturing Engineering, University of Nottingham Ningbo China, Ningbo, P. R. China
| | - Zhenqi Jiang
- Cixi Institute of Biomedical Engineering, CAS Key Laboratory of Magnetic Materials and Devices, & Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, P. R. China
| | - Jing Wang
- Department of Electrical and Electronic Engineering, University of Nottingham Ningbo China, Ningbo, P. R. China
| | - Yong Ren
- Research Group for Fluids and Thermal Engineering, University of Nottingham Ningbo China, Ningbo, P. R. China.,Department of Mechanical, Materials and Manufacturing Engineering, University of Nottingham Ningbo China, Ningbo, P. R. China
| | - Aiguo Wu
- Cixi Institute of Biomedical Engineering, CAS Key Laboratory of Magnetic Materials and Devices, & Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, P. R. China
| |
Collapse
|
15
|
Yang F, Chen S, He S, Huo Q, Hu Y, Xie N. YB-1 interplays with ERα to regulate the stemness and differentiation of ER-positive breast cancer stem cells. Theranostics 2020; 10:3816-3832. [PMID: 32206124 PMCID: PMC7069074 DOI: 10.7150/thno.41014] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 01/21/2020] [Indexed: 02/06/2023] Open
Abstract
Background: Some stemness-associated transcription factors consistently play essential roles in the maintenance of pluripotency or induce the differentiation of cancer stem cells (CSCs). However, the regulatory mechanism of CSC stemness mediated by transcription factors has not been extensively explored. Here, we show that two transcription factors (YB-1 and ERα), which are simultaneously highly expressed in estrogen receptor (ER)-positive CSCs, interact with each other to regulate the stemness and differentiation of ER-positive CSCs. Methods: The expression of YB-1 was examined in ER-positive CSCs and patient specimens. Western blot, real-time PCR, cell viability analysis, tumorsphere formation assay and subcutaneous tumorigenesis assays were used to study the stemness functions of YB-1 and ERα in CSCs. The relationship between YB-1 and ERα in cells was studied by promoter activity analysis, the electrophoretic mobility shift assay (EMSA) and the Co-IP assay. The mechanisms and functional significance of YB-1 in the sensitivity of CSCs to tamoxifen were further investigated with both in vitro and in vivo models. Results: YB-1 was aberrantly upregulated in the cancerous tissue of ER-positive breast cancer patients and in CSCs. Knockdown of YB-1 in ER-positive CSCs significantly inhibited cell stemness and induced differentiation, and the expression of YB-1 could be regulated by estrogen signaling and ERα in ER-positive breast CSCs. The Co-IP results showed that YB-1 interacted directly with ERα specifically in ER-positive non-CSCs and that YB-1 induced ERα degradation by ubiquitination via direct interaction in differentiated cells. Cell differentiation induced by FBS could inhibit YB-1 phosphorylation and promote YB-1 protein transfer from the nucleus to the cytoplasm. Moreover, cell differentiation induced by targeting inhibited the expression of YB-1 in ER-positive CSCs, which increased the sensitivity of cells to tamoxifen in vitro and in vivo. Conclusion: The ERα/YB-1 axis has an important role in the regulation of ER-positive breast cancer stemness. The dephosphorylation of YB-1 and the interaction between YB-1 and ERα may be the switch that initiates the differentiation of ER-positive CSCs. Targeting YB-1 to sensitize ER-positive CSCs to antiestrogen therapy might represent a new therapeutic strategy that warrants further exploration.
Collapse
Affiliation(s)
- Fan Yang
- Biobank, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen 518035, People's Republic of China
- Shenzhen institute of advanced technology, Chinese academy of sciences, Shenzhen 518035, People's Republic of China
| | - Siqi Chen
- Biobank, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen 518035, People's Republic of China
| | - Shengnan He
- Biobank, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen 518035, People's Republic of China
| | - Qin Huo
- Biobank, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen 518035, People's Republic of China
| | - Ye Hu
- Biobank, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen 518035, People's Republic of China
| | - Ni Xie
- Biobank, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen 518035, People's Republic of China
| |
Collapse
|
16
|
Johnson TG, Schelch K, Mehta S, Burgess A, Reid G. Why Be One Protein When You Can Affect Many? The Multiple Roles of YB-1 in Lung Cancer and Mesothelioma. Front Cell Dev Biol 2019; 7:221. [PMID: 31632972 PMCID: PMC6781797 DOI: 10.3389/fcell.2019.00221] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 09/18/2019] [Indexed: 12/14/2022] Open
Abstract
Lung cancers and malignant pleural mesothelioma (MPM) have some of the worst 5-year survival rates of all cancer types, primarily due to a lack of effective treatment options for most patients. Targeted therapies have shown some promise in thoracic cancers, although efficacy is limited only to patients harboring specific mutations or target expression. Although a number of actionable mutations have now been identified, a large population of thoracic cancer patients have no therapeutic options outside of first-line chemotherapy. It is therefore crucial to identify alternative targets that might lead to the development of new ways of treating patients diagnosed with these diseases. The multifunctional oncoprotein Y-box binding protein-1 (YB-1) could serve as one such target. Recent studies also link this protein to many inherent behaviors of thoracic cancer cells such as proliferation, invasion, metastasis and involvement in cancer stem-like cells. Here, we review the regulation of YB-1 at the transcriptional, translational, post-translational and sub-cellular levels in thoracic cancer and discuss its potential use as a biomarker and therapeutic target.
Collapse
Affiliation(s)
- Thomas G Johnson
- Asbestos Diseases Research Institute, Sydney, NSW, Australia.,Cell Division Laboratory, The ANZAC Research Institute, Sydney, NSW, Australia.,School of Medicine, The University of Sydney, Sydney, NSW, Australia.,Sydney Catalyst Translational Cancer Research Centre, The University of Sydney, Sydney, NSW, Australia
| | - Karin Schelch
- Institute of Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Sunali Mehta
- Department of Pathology, University of Otago, Dunedin, New Zealand.,Maurice Wilkins Centre, University of Otago, Dunedin, New Zealand
| | - Andrew Burgess
- Cell Division Laboratory, The ANZAC Research Institute, Sydney, NSW, Australia.,School of Medicine, The University of Sydney, Sydney, NSW, Australia
| | - Glen Reid
- Department of Pathology, University of Otago, Dunedin, New Zealand.,Maurice Wilkins Centre, University of Otago, Dunedin, New Zealand
| |
Collapse
|
17
|
Whole Genome Analysis and Prognostic Model Construction Based on Alternative Splicing Events in Endometrial Cancer. BIOMED RESEARCH INTERNATIONAL 2019; 2019:2686875. [PMID: 31355251 PMCID: PMC6634061 DOI: 10.1155/2019/2686875] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 06/04/2019] [Indexed: 12/31/2022]
Abstract
Objectives A growing body of evidence has shown that aberrant alternative splicing (AS) is closely related to the occurrence and development of cancer. However, prior studies mainly have concentrated on a few genes that exhibit aberrant AS. This study aimed to determine AS events through whole genome analysis and construct a prognostic model of endometrial cancer (EC). Methods We downloaded gene expression RNAseq data from UCSC Xena, and seven types of AS events from TCGA SpliceSeq. Univariate Cox regression was employed to analyze the prognostic-related alternative splicing events (PASEs) and splicing factors; multivariate Cox regression was conducted to analyze the effect of risk score (All) and clinicopathological parameters on EC prognosis. An underlying interaction network of PASEs of EC was constructed by Cytoscape Reactome FI, GO, and KEGG pathway enrichment was performed by DAVID. ROC curves and Kaplan-Meir analysis were used to assess the diagnostic value of prognostic model. The correlation between PASEs and splicing factors was analyzed by GraphPad Prism; then a network was constructed using Cytoscape. Results In total, 28,281 AS events in EC were identified, which consisted of 1166 PASEs. RNPS1, NEK2, and CTNNB1 were the hub genes in the network of the top 600 PASEs. The area under the curve (AUC) of risk score (All) reached 0.819. Risk score (All) together with FIGO stage, cancer status, and primary therapy outcome success was risk factors of the prognosis of EC patients. Splicing factors YBX1, HNRNPDL, and HNRNPA1 were significantly related to the overall survival (OS). The splicing network indicated that the expression of splicing factors was significantly correlated with percent-splice-in (PSI) value of PASEs. Conclusion We constructed a model for predicting the prognosis of EC patients based on PASEs using whole genome analysis of AS events and thereby provided a reliable theoretical basis for EC clinical prognosis evaluation.
Collapse
|
18
|
Ruzycka M, Cimpan MR, Rios-Mondragon I, Grudzinski IP. Microfluidics for studying metastatic patterns of lung cancer. J Nanobiotechnology 2019; 17:71. [PMID: 31133019 PMCID: PMC6537392 DOI: 10.1186/s12951-019-0492-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 05/04/2019] [Indexed: 01/09/2023] Open
Abstract
The incidence of lung cancer continues to rise worldwide. Because the aggressive metastasis of lung cancer cells is the major drawback of successful therapies, the crucial challenge of modern nanomedicine is to develop diagnostic tools to map the molecular mechanisms of metastasis in lung cancer patients. In recent years, microfluidic platforms have been given much attention as tools for novel point-of-care diagnostic, an important aspect being the reconstruction of the body organs and tissues mimicking the in vivo conditions in one simple microdevice. Herein, we present the first comprehensive overview of the microfluidic systems used as innovative tools in the studies of lung cancer metastasis including single cancer cell analysis, endothelial transmigration, distant niches migration and finally neoangiogenesis. The application of the microfluidic systems to study the intercellular crosstalk between lung cancer cells and surrounding tumor microenvironment and the connection with multiple molecular signals coming from the external cellular matrix are discussed. We also focus on recent breakthrough technologies regarding lab-on-chip devices that serve as tools for detecting circulating lung cancer cells. The superiority of microfluidic systems over traditional in vitro cell-based assays with regard to modern nanosafety studies and new cancer drug design and discovery is also addressed. Finally, the current progress and future challenges regarding printable and paper-based microfluidic devices for personalized nanomedicine are summarized.
Collapse
Affiliation(s)
- Monika Ruzycka
- Department of Applied Toxicology, Faculty of Pharmacy, Medical University of Warsaw, 1 Banacha Street, 02-097, Warsaw, Poland
| | - Mihaela R Cimpan
- Biomaterials - Department for Clinical Dentistry, University of Bergen, Årstadveien 19, 5009, Bergen, Norway
| | - Ivan Rios-Mondragon
- Biomaterials - Department for Clinical Dentistry, University of Bergen, Årstadveien 19, 5009, Bergen, Norway
| | - Ireneusz P Grudzinski
- Department of Applied Toxicology, Faculty of Pharmacy, Medical University of Warsaw, 1 Banacha Street, 02-097, Warsaw, Poland.
| |
Collapse
|
19
|
Li Z, Guo T, Fang L, Li N, Wang X, Wang P, Zhao S, Li F, Cui Y, Shu X, Zhao L, Li J, Gu C. MACC1 overexpression in carcinoma‑associated fibroblasts induces the invasion of lung adenocarcinoma cells via paracrine signaling. Int J Oncol 2019; 54:1367-1375. [PMID: 30720137 DOI: 10.3892/ijo.2019.4702] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 11/26/2018] [Indexed: 11/05/2022] Open
Abstract
Carcinoma‑associated fibroblasts (CAFs) are essential for initiating lung cancer cell invasion and metastasis. An elevated MACC1 expression has been implicated in the progression of lung adenocarcinoma. Hitherto, the role of MACC1 in lung adenocarcinoma‑derived CAFs remains unclear. In this study, CAFs isolated from the tissues of patients with lung adenocarcinoma expressed typical CAF markers (shown by immunohistochemical and immunofluorescence analysis) and exhibited enhanced migration and invasion abilities when co‑cultured with A549 cells in a microfluidic model. MACC1‑overexpressing CAFs not only demonstrated an increased invasion, but also exerted a promoting effect on the invasion of tumor cells. The reduced expression of MACC1 impaired the invasive ability of the CAFs. Western blot analysis and RT‑qPCR analysis demonstrated that multiple paracrine pathways were activated in the MACC1‑overexpressing CAFs. Overall, this study presents a novel role of MACC1 in CAF‑induced lung adenocarcinoma cell invasion, which possibly occurs via paracrine signaling. Furthermore, MACC1 was indicated to be a potential therapeutic target for lung adenocarcinoma.
Collapse
Affiliation(s)
- Zhuoshi Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Tao Guo
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Lei Fang
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Nan Li
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Xiaochao Wang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Peng Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Shilei Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Fengzhou Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Yanwei Cui
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Xin Shu
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Lei Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Jinxiu Li
- Lung Cancer Diagnosis and Treatment Center of Dalian, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Chundong Gu
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| |
Collapse
|
20
|
Cytokeratin 19 (KRT19) has a Role in the Reprogramming of Cancer Stem Cell-Like Cells to Less Aggressive and More Drug-Sensitive Cells. Int J Mol Sci 2018; 19:ijms19051423. [PMID: 29747452 PMCID: PMC5983664 DOI: 10.3390/ijms19051423] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 05/04/2018] [Accepted: 05/07/2018] [Indexed: 02/08/2023] Open
Abstract
Cytokeratin 19 (KRT19) is a cytoplasmic intermediate filament protein, which is responsible for structural rigidity and multipurpose scaffolds. In several cancers, KRT19 is overexpressed and may play a crucial role in tumorigenic transformation. In our previous study, we revealed the role of KRT19 as signaling component which mediated Wnt/NOTCH crosstalk through NUMB transcription in breast cancer. Here, we investigated the function of KRT19 in cancer reprogramming and drug resistance in breast cancer cells. We found that expression of KRT19 was attenuated in several patients-derived breast cancer tissues and patients with a low expression of KRT19 were significantly correlated with poor prognosis in breast cancer patients. Consistently, highly aggressive and drug-resistant breast cancer patient-derived cancer stem cell-like cells (konkuk university-cancer stem cell-like cell (KU-CSLCs)) displayed higher expression of cancer stem cell (CSC) markers, including ALDH1, CXCR4, and CD133, but a much lower expression of KRT19 than that is seen in highly aggressive triple negative breast cancer MDA-MB231 cells. Moreover, we revealed that the knockdown of KRT19 in MDA-MB231 cells led to an enhancement of cancer properties, such as cell proliferation, sphere formation, migration, and drug resistance, while the overexpression of KRT19 in KU-CSLCs resulted in the significant attenuation of cancer properties. KRT19 regulated cancer stem cell reprogramming by modulating the expression of cancer stem cell markers (ALDH1, CXCR4, and CD133), as well as the phosphorylation of Src and GSK3β (Tyr216). Therefore, our data may imply that the modulation of KRT19 expression could be involved in cancer stem cell reprogramming and drug sensitivity, which might have clinical implications for cancer or cancer stem cell treatment.
Collapse
|
21
|
Guo T, Zhao S, Wang P, Xue X, Zhang Y, Yang M, Li N, Li Z, Xu L, Jiang L, Zhao L, Ma PC, Rosell R, Li J, Gu C. YB-1 regulates tumor growth by promoting MACC1/c-Met pathway in human lung adenocarcinoma. Oncotarget 2018. [PMID: 28624808 PMCID: PMC5564630 DOI: 10.18632/oncotarget.18262] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Aberrant overexpression of the transcription/translation factor Y-box-binding protein (YB-1) is associated with poor prognosis of lung adenocarcinoma, however the underlying mechanism by which YB-1 acts has not been fully elucidated. Here, we reported that inhibition of YB-1 diminished proliferation, migration and invasion of lung adenocarcinoma cells. Interestingly, we identified metastasis associated in colon cancer-1 (MACC1) as a target of YB-1. Depletion of YB-1 markedly decreased MACC1 promoter activity and suppressed the MACC1/c-Met signaling pathway in lung adenocarcinoma cells. Additionally, chromatin immunoprecipitation (ChIP) assay demonstrated that YB-1 bound to the MACC1 promoter. Moreover, YB-1 was positively correlated with MACC1, and both proteins were over-expressed in lung adenocarcinoma tissues. The Cox-regression analysis indicated that high YB-1 expression was an independent risk factor for prognosis in enrolled patients. Furthermore, depletion of YB-1 attenuated tumorigenesis in a xenograft mouse model and reduced MACC1 expression in tumor tissues. Collectively, our data suggested that targeting YB-1 suppressed lung adenocarcinoma progression through the MACC1/c-Met pathway and that the high expression of YB-1/MACC1 is a potential prognostic marker in lung adenocarcinoma.
Collapse
Affiliation(s)
- Tao Guo
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.,Lung Cancer Diagnosis and Treatment Center of Dalian, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Shilei Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.,Lung Cancer Diagnosis and Treatment Center of Dalian, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Peng Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.,Lung Cancer Diagnosis and Treatment Center of Dalian, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xiaoyuan Xue
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116044, China
| | - Yan Zhang
- Department of Radiation Oncology, Qianfoshan Hospital Affiliated to Shandong University, Jinan 250000, China
| | - Mengying Yang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116044, China
| | - Nan Li
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116044, China
| | - Zhuoshi Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.,Lung Cancer Diagnosis and Treatment Center of Dalian, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Lingzhi Xu
- The Second Affiliated Hospital, Dalian Medical University, Dalian 116011, China
| | - Lei Jiang
- The Fourth Affiliated Hospital, Anhui Medical University, Hefei 230000, China
| | - Lei Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.,Lung Cancer Diagnosis and Treatment Center of Dalian, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Patrick C Ma
- Aerodigestive Oncology Translational Research THOR, Department of Solid Tumor Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Rafael Rosell
- Breakthrough Cancer Research Unit, Pangaea Biotech, Dexeus University Institute, Catalan Institute of Oncology, Badalona 08916, Spain
| | - Jinxiu Li
- Lung Cancer Diagnosis and Treatment Center of Dalian, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Chundong Gu
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.,Lung Cancer Diagnosis and Treatment Center of Dalian, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| |
Collapse
|
22
|
Pérez-Boza J, Lion M, Struman I. Exploring the RNA landscape of endothelial exosomes. RNA (NEW YORK, N.Y.) 2018; 24:423-435. [PMID: 29282313 PMCID: PMC5824360 DOI: 10.1261/rna.064352.117] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 12/21/2017] [Indexed: 05/05/2023]
Abstract
Exosomes are small extracellular vesicles of around 100 nm of diameter produced by most cell types. These vesicles carry nucleic acids, proteins, lipids, and other biomolecules and function as carriers of biological information in processes of extracellular communication. The content of exosomes is regulated by the external and internal microenvironment of the parent cell, but the intrinsic mechanisms of loading of molecules into exosomes are still not completely elucidated. In this study, by the use of next-generation sequencing we have characterized in depth the RNA composition of healthy endothelial cells and exosomes and provided an accurate profile of the different coding and noncoding RNA species found per compartment. We have also discovered a set of unique genes preferentially included (or excluded) into vesicles. Moreover, after studying the enrichment of RNA motifs in the genes unequally distributed between cells and exosomes, we have detected a set of enriched sequences for several classes of RNA. In conclusion, our results provide the basis for studying the involvement of RNA-binding proteins capable of recognizing RNA sequences and their role in the export of RNAs into exosomes.
Collapse
Affiliation(s)
- Jennifer Pérez-Boza
- Laboratory of Molecular Angiogenesis, GIGA-R, University of Liège, 4000 Liège, Belgium
| | - Michelle Lion
- Laboratory of Molecular Angiogenesis, GIGA-R, University of Liège, 4000 Liège, Belgium
| | - Ingrid Struman
- Laboratory of Molecular Angiogenesis, GIGA-R, University of Liège, 4000 Liège, Belgium
| |
Collapse
|
23
|
Wang Y, Su J, Fu D, Wang Y, Chen Y, Chen R, Qin G, Zuo J, Yue D. The Role of YB1 in Renal Cell Carcinoma Cell Adhesion. Int J Med Sci 2018; 15:1304-1311. [PMID: 30275756 PMCID: PMC6158664 DOI: 10.7150/ijms.25580] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 06/28/2018] [Indexed: 11/05/2022] Open
Abstract
Background: Y-box binding protein 1 (YB1) is a multifunctional protein involved in many processes related to cancer progression and metastasis. Methods: In this study, we constructed YB1 knockdown stable renal cell carcinoma (RCC) cell line 786-0. The gene expression profile of 786-0 was performed by DNA microarray analysis to identify genes that were regulated by YB1. Real-time PCR and western blotting were used to test the genes and proteins expression. Transforming growth factor-β (TGF-β) activity was detected by dual-luciferase reporter assay. Cell adhesion assay was used to determine RCC cell adhesion ability. Results: Pathway analysis revealed that YB1 knockdown influenced cell adhesion molecules (CAMs). We further verified four genes (CLDN4, NRXN3, ITGB8, and VCAN) related to CAMs by real-time PCR, and confirmed that YB1 regulated the expression of ITGB8 in RCC. Functional assays demonstrated that knockdown of YB1 significantly inhibited the cell adhesion of 786-0 cells in vitro. In addition, YB1 affected TGF-β activation. Conclusion: Our study demonstrated that YB1 modulated the adhesion ability of renal cell carcinoma cells by regulating ITGB8 and TGF-β.
Collapse
Affiliation(s)
- Yong Wang
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology and Department of Microbiology, School of Medical Laboratory, Tianjin Medical University, Tianjin 300070, China
| | - Jing Su
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology and Department of Microbiology, School of Medical Laboratory, Tianjin Medical University, Tianjin 300070, China
| | - Donghe Fu
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology and Department of Microbiology, School of Medical Laboratory, Tianjin Medical University, Tianjin 300070, China.,Department of Clinical Laboratory, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin 300052, China
| | - Yiting Wang
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology and Department of Microbiology, School of Medical Laboratory, Tianjin Medical University, Tianjin 300070, China
| | - Yajing Chen
- Research Center of Molecular Biology, Inner Mongolia Medical University, Hohhot 010059, China
| | - Ruibing Chen
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Guoxuan Qin
- School of Microelectronics, Tianjin University, Tianjin 300072, China
| | - Jing Zuo
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology and Department of Microbiology, School of Medical Laboratory, Tianjin Medical University, Tianjin 300070, China
| | - Dan Yue
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology and Department of Microbiology, School of Medical Laboratory, Tianjin Medical University, Tianjin 300070, China
| |
Collapse
|
24
|
Kim KY, Zhang X, Cha IH. Identifying a combined biomarker for bisphosphonate-related osteonecrosis of the jaw. Clin Implant Dent Relat Res 2017; 20:191-198. [PMID: 29266738 DOI: 10.1111/cid.12569] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 10/23/2017] [Accepted: 11/13/2017] [Indexed: 11/28/2022]
Abstract
BACKGROUND For this study, the aim was to identify combined biomarkers associated with bisphosphonate-related osteonecrosis of the jaw (BRONJ). MATERIALS AND METHODS Microarray data for GSE7116 were downloaded from the Gene Expression Omnibus database, which contains 26 samples, including without ONJ, and 5 healthy volunteers. The combined biomarkers were identified using principal component analysis, and the pathway enrichment analyses were performed using the DAVID online tool. RESULTS Two hundred differently expressed genes between groups were detected according to the significances. From functional annotation, Y-box binding protein 1 and heterogeneous nuclear ribonucleoprotein C were found to be included in the most significant 10 pathways. Ten combined gene sets were identified that were effective in classifying multiple myeloma (MM) with ONJ and MM without ONJ. CONCLUSION Identifying combined gene expression profiles is expected to contribute to more personalized management of BRONJ and to improve existing therapies, and it will be helpful in finding new therapies by identifying more predictive biomarkers.
Collapse
Affiliation(s)
- Ki-Yeol Kim
- Dental Education Research Center, BK21 PLUS Project, Yonsei University College of Dentistry, Seoul, Republic of Korea
| | - Xianglan Zhang
- Department of Pathology, Yanbian University Medical College, Yanji City, Jilin Province, China
| | - In-Ho Cha
- Department of Oral and Maxillofacial Surgery, College of Dentistry, Yonsei University, Seoul, Republic of Korea
| |
Collapse
|
25
|
Bledzka K, Schiemann B, Schiemann WP, Fox P, Plow EF, Sossey-Alaoui K. The WAVE3-YB1 interaction regulates cancer stem cells activity in breast cancer. Oncotarget 2017; 8:104072-104089. [PMID: 29262622 PMCID: PMC5732788 DOI: 10.18632/oncotarget.22009] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Accepted: 09/29/2017] [Indexed: 12/12/2022] Open
Abstract
Resistance to therapy is the main cause of tumor recurrence and metastasis and cancer stem cells (CSCs) play a crucial role in this process, especially in triple-negative breast cancers (TNBCs). Unfortunately, no FDA-approved treatment is currently available for this subtype of BC, which explains the high rate of mortality in patients with TNBC tumors. WAVE3, a member of the WASP/WAVE actin-cytoskeleton remodeling family of protein, has been established as a major driver of tumor progression and metastasis of several solid tumors, including those originating in the breast. Our recently published studies found WAVE3 to mediate the process of chemoresistance in TNBCs. The molecular mechanisms whereby WAVE3 regulates chemoresistance in TNBC tumors remains largely unknown, as does the role of WAVE3 in CSC maintenance. Here we show that WAVE3 promotes CSC self-renewal and regulates transcription of CSC-specific genes, which, in part, provides a mechanistic explanation for the function of WAVE3 in chemoresistance in TNBCs. Our data show that WAVE3 is enriched in the CSC-subpopulation of TNBC cell lines. Knockout of WAVE3 via CRISPR/Cas9 significantly attenuates the CSC-subpopulation and inhibits transcription of CSC transcription factors. Mechanistically, we established a link between WAVE3 and the Y-box-binding protein-1 (YB1), a transcription factor and CSC-maintenance gene. Indeed, the interaction of WAVE3 with YB1 is required for YB1 translocation to the nucleus of cancer cells, and activation of transcription of CSC-specific genes. Our findings identify a new WAVE3/YB1 signaling axis that regulates the CSC-mediated resistance to therapy and opens a new therapeutic window for TNBCs treatment.
Collapse
Affiliation(s)
- Kamila Bledzka
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | | | | | - Paul Fox
- Department of Cellular and Molecular Medicine, Cleveland, Ohio, USA
| | - Edward F Plow
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Khalid Sossey-Alaoui
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
26
|
Abstract
Advances in cancer research in the past have led to an evolving understanding of cancer pathogenesis and the development of novel drugs that significantly improve patient outcomes. However, many patients still encounter treatment resistance, recurrence, or metastasis and eventually die from progressing disease. Experimental evidence indicates that a subpopulation of cancer cells, called cancer stem cells (CSCs), possess "stemness" properties similar to normal stem cells, including self-renewal, differentiation, and proliferative potential. These stemness properties are lost during differentiation and are governed by pathways such as STAT3, NANOG, NOTCH, WNT, and HEDGEHOG, which are highly dysregulated in CSCs due to genetic and epigenetic changes. Promising results have been observed in preclinical models targeting these CSCs through the disruption of stemness pathways in combination with current treatment modalities. This has led to anti-CSC-based clinical trials in multiple stages of development. In this review, we discuss the role of CSCs and stemness pathways in cancer treatment and how they relate to clinical observations. Because CSCs and the stemness pathways governing them may explain the negative clinical outcomes observed during treatment, it is important for oncologists to understand how they contribute to cancer progression and how they may be targeted to improve patient outcomes.
Collapse
Affiliation(s)
- Justin D Lathia
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave., NC10, Cleveland, OH, 44195, USA.
| | - Huiping Liu
- Departments of Pharmacology and Medicine (Hematology/Oncology), Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|