1
|
Ma Y, Song B, Peng J, Wei F, Hao Y, Wen Y, Lv H, Shi X, Wang Y, Peng T. Cornua cervi degelatinatum inhibits breast cancer stem-like cell properties and metastasis via miR-148a-3p-mediated TGF-β/Smad2 pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 346:119709. [PMID: 40157402 DOI: 10.1016/j.jep.2025.119709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/12/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cornua cervi degelatinatum (CCD) is formed by removing the gelatinous substance from deer antlers according to traditional methods. It was first recorded in the Shennong's Classic of Materia Medica and has been included in the Pharmacopoeia of the People's Republic of China. It is commonly used in clinical practice for the treatment of diseases such as cancer and infertility. AIM OF THE STUDY This study aims to investigate the impact of CCD aqueous extract on the proliferation and stemness of breast cancer (BC) cells, with an emphasis on its regulation of miR-148a-3p expression and associated molecular pathways. MATERIALS AND METHODS Breast cancer cells were treated with various concentrations of CCD to assess its effects on cancer stem cell (CSC) features, epithelial-mesenchymal transition (EMT) markers, and overall plasticity. The UALCAN platform was utilized to analyze the relationship between miR-148a-3p and Smad2 expression. Functional experiments involving miR-148a-3p overexpression were performed to elucidate CCD's modulatory effects on the TGF-β/Smad2 pathway. Furthermore, molecular docking analysis was conducted to predict the binding affinity of CCD's active components to Smad2. RESULTS The CCD aqueous extract significantly reduced BC cell viability in vitro and dose-dependently suppressed the expression of stemness- and EMT-related proteins. Bioinformatics analysis and luciferase reporter assays validated miR-148a-3p as a direct regulator of Smad2, inhibiting the TGF-β/Smad2 signaling pathway. Molecular docking revealed strong binding interactions between CCD's active components and Smad2. CONCLUSIONS CCD exhibits anti-BC effects by working synergistically with miR-148a-3p to inhibit the TGF-β/Smad2 pathway, thereby reducing BC stemness and EMT progression. These findings provide valuable insights into the molecular mechanisms underlying CCD's therapeutic potential in BC treatment.
Collapse
Affiliation(s)
- Yanmiao Ma
- College of Basic Medical Sciences, Shanxi University of Chinese Medicine, Taiyuan, 030619, China; Shanxi Provincial Key Laboratory of Classical Prescription Strengthening Yang, Taiyuan, 030013, China; Shanxi Provincial Key Laboratory of Prescription Compatibility and Functions, Taiyuan, 030619, China.
| | - Bo Song
- College of Third Clinical Medicine, Shanxi University of Chinese Medicine, Taiyuan, 030619, China
| | - Jiehao Peng
- College of Third Clinical Medicine, Shanxi University of Chinese Medicine, Taiyuan, 030619, China
| | - Fuxia Wei
- College of Third Clinical Medicine, Shanxi University of Chinese Medicine, Taiyuan, 030619, China
| | - Yuanhui Hao
- College of Third Clinical Medicine, Shanxi University of Chinese Medicine, Taiyuan, 030619, China
| | - Ya Wen
- College of First Clinical Medicine, Shanxi University of Chinese Medicine, Taiyuan, 030619, China
| | - Huiling Lv
- Experimental Management Center, Shanxi University of Chinese Medicine, Taiyuan, 030619, China
| | - Xiaoli Shi
- Department of Rehabilitation, Shanxi Acupuncture Hospital, Taiyuan, 030006, China
| | - Yonghui Wang
- College of Basic Medical Sciences, Shanxi University of Chinese Medicine, Taiyuan, 030619, China; Shanxi Provincial Key Laboratory of Classical Prescription Strengthening Yang, Taiyuan, 030013, China.
| | - Tao Peng
- Shanxi Hospital of Integrated Traditional Chinese and Western Medicine, Taiyuan, 030013, China; Shanxi Provincial Key Laboratory of Classical Prescription Strengthening Yang, Taiyuan, 030013, China; Shanxi Provincial Key Laboratory of Prescription Compatibility and Functions, Taiyuan, 030619, China.
| |
Collapse
|
2
|
Khan SU, Cervantes-Villagrana RD, Eduardo Del Río-Robles J, Tomás-Morales JA, Torres-Santos Y, Vázquez-Prado J, Reyes-Cruz G. Calcium sensing receptor stimulates breast cancer cell migration and invasion via protein kinase C ζ. Exp Cell Res 2025; 447:114523. [PMID: 40120711 DOI: 10.1016/j.yexcr.2025.114523] [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: 03/15/2025] [Accepted: 03/16/2025] [Indexed: 03/25/2025]
Abstract
Calcium-sensing receptor (CaSR), a G protein-coupled receptor, is overexpressed in certain breast cancer tumors where it drives cell migration and secretion of chemotactic agonists, likely contributing to metastatic dissemination. Since CaSR activates breast cancer cell migration via the Gβγ-PI3K-mTORC2/Rac-1 pathway, we hypothesized that PKCζ and perhaps other protein kinase C (PKC) isoforms, known as mTORC2-regulated effectors, are involved in migratory and invasive signaling elicited by CaSR. We analyzed the effect of PKC inhibitors and siRNAs which pointed to PKCζ as effector of CaSR in cell migration and invasion. In breast cancer phosphoproteomic CPTAC datasets, we identified a group of Luminal A subtype cancer patients having active PKCζ, according to its phosphorylation status at the turn motif. In addition, various phosphorylated RacGEFs, including TRIO, ARHGEF26, DOCK1 and DOCK7, clustered as phosphoproteins with active PKCζ. We therefore introduce atypical PKCζ as an effector component of the CaSR-Gβγ-PI3K-mTORC2 pathway in the activation of the promigratory small GTPase Rac. These results support ongoing initiatives to establish critical elements of the CaSR signaling pathway as potential targets in metastatic breast cancer.
Collapse
Affiliation(s)
- Safir Ullah Khan
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del IPN (Cinvestav-IPN), Mexico City, Mexico
| | | | - Jorge Eduardo Del Río-Robles
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del IPN (Cinvestav-IPN), Mexico City, Mexico
| | - Janik Adriana Tomás-Morales
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del IPN (Cinvestav-IPN), Mexico City, Mexico
| | - Yazmin Torres-Santos
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del IPN (Cinvestav-IPN), Mexico City, Mexico
| | - José Vázquez-Prado
- Department of Pharmacology, Centro de Investigación y de Estudios Avanzados del IPN (Cinvestav-IPN), Mexico City, Mexico
| | - Guadalupe Reyes-Cruz
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del IPN (Cinvestav-IPN), Mexico City, Mexico.
| |
Collapse
|
3
|
Huang J, Luo S, Shen J, Lee M, Chen R, Ma S, Sun LQ, Li JJ. Cellular polarity pilots breast cancer progression and immunosuppression. Oncogene 2025; 44:783-793. [PMID: 40057606 PMCID: PMC11913746 DOI: 10.1038/s41388-025-03324-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 02/03/2025] [Accepted: 02/19/2025] [Indexed: 03/19/2025]
Abstract
Disrupted cellular polarity (DCP) is a hallmark of solid cancer, the malignant disease of epithelial tissues, which occupies ~90% of all human cancers. DCP has been identified to affect not only the cancer cell's aggressive behavior but also the migration and infiltration of immune cells, although the precise mechanism of DCP-affected tumor-immune cell interaction remains unclear. This review discusses immunosuppressive tumor microenvironments (TME) caused by DCP-driven tumor cell proliferation with DCP-impaired immune cell functions. We will revisit the fundamental roles of cell polarity (CP) proteins in sustaining mammary luminal homeostasis, epithelial transformation, and breast cancer progression. Then, the current data on CP involvement in immune cell activation, maturation, migration, and tumor infiltration are evaluated. The CP status on the immune effector cells and their targeted tumor cells are highlighted in tumor immune regulation, including the antigen presentation and the formation of immune synapses (IS). CP-regulated antigen presentation and delivery and the formation of IS between the immune cells, especially between the immune effectors and tumor cells, will be addressed. Alterations of CP on the tumor cells, infiltrated immune effector cells, or both are discussed with these aspects. We conclude that CP-mediated tumor aggressiveness coupled with DCP-impaired immune cell disability may decide the degree of immunosuppressive status and responsiveness to immune checkpoint blockade (ICB). Further elucidating the dynamics of CP- or DCP-mediated immune regulation in TME will provide more critical insights into tumor-immune cell dynamics, which is required to invent more effective approaches for cancer immunotherapy.
Collapse
Affiliation(s)
- Jie Huang
- Department of Thoracic Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, China
- Department of Radiation Oncology, University of California Davis, Sacramento, California, USA
| | - Shufeng Luo
- Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Cancer Center, Central South University, China, Hunan, Changsha
| | - Juan Shen
- Department of Thoracic Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, China
| | - Maya Lee
- Department of Radiation Oncology, University of California Davis, Sacramento, California, USA
| | - Rachel Chen
- Department of Radiation Oncology, University of California Davis, Sacramento, California, USA
| | - Shenglin Ma
- Department of Thoracic Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, China
| | - Lun-Quan Sun
- Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Cancer Center, Central South University, China, Hunan, Changsha.
| | - Jian Jian Li
- Department of Radiation Oncology, University of California Davis, Sacramento, California, USA.
- NCI-designated Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, California, USA.
| |
Collapse
|
4
|
Kumari L, Yadav R, Kumar Y, Bhatia A. Role of tight junction proteins in shaping the immune milieu of malignancies. Expert Rev Clin Immunol 2024; 20:1305-1321. [PMID: 39126381 DOI: 10.1080/1744666x.2024.2391915] [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: 05/07/2024] [Revised: 08/05/2024] [Accepted: 08/09/2024] [Indexed: 08/12/2024]
Abstract
INTRODUCTION Tight junctions (TJs) and their constituent proteins play pivotal roles in cellular physiology and anatomy by establishing functional boundaries within and between neighboring cells. While the involvement of TJ proteins, such as claudins, in cancer is extensively studied, studies highlighting their interaction with immune system are still meager. Studies indicate that alterations in cytokines and immune cell populations can affect TJ proteins, compromising TJ barrier function and exacerbating pro-inflammatory conditions, potentially leading to epithelial cell malignancy. Disrupted TJs in established tumors may foster a pro-tumor immune microenvironment, facilitating tumor progression, invasion, epithelial-to-mesenchymal transition and metastasis. Although previous literature contains many studies describing the involvement of TJs in pathogenesis of malignancies their role in modulating the immune microenvironment of tumors is just beginning to be unleashed. AREAS COVERED This article for the first time attempts to discern the importance of interaction between TJs and immune microenvironment in malignancies. To achieve the above aim a thorough search of databases like PubMed and Google Scholar was conducted to identify the recent and relevant articles on the topic. EXPERT OPINION Breaking the vicious cycle of dysbiosis/infections/chemical/carcinogen-induced inflammation-TJ remodeling-malignancy-TJ dysregulation-more inflammation can be used as a strategy to complement the effect of immunotherapies in various malignancies.
Collapse
Affiliation(s)
- Laxmi Kumari
- Department of Experimental Medicine and Biotechnology, Post Graduate Institute of Medical Education & Research, Chandigarh, India
| | - Reena Yadav
- Department of Experimental Medicine and Biotechnology, Post Graduate Institute of Medical Education & Research, Chandigarh, India
| | - Yashwant Kumar
- Department of Immunopathology, Post Graduate Institute of medical Education and Research, Chandigarh, India
| | - Alka Bhatia
- Department of Experimental Medicine and Biotechnology, Post Graduate Institute of Medical Education & Research, Chandigarh, India
| |
Collapse
|
5
|
Haque M, Shyanti RK, Mishra MK. Targeted therapy approaches for epithelial-mesenchymal transition in triple negative breast cancer. Front Oncol 2024; 14:1431418. [PMID: 39450256 PMCID: PMC11499239 DOI: 10.3389/fonc.2024.1431418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 09/24/2024] [Indexed: 10/26/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is distinguished by negative expression of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2), making it an aggressive subtype of breast cancer and contributes to 15-20% of the total incidence. TNBC is a diverse disease with various genetic variations and molecular subtypes. The tumor microenvironment involves multiple cells, including immune cells, fibroblast cells, extracellular matrix (ECM), and blood vessels that constantly interact with tumor cells and influence each other. The ECM undergoes significant structural changes, leading to induced cell proliferation, migration, adhesion, invasion, and epithelial-to-mesenchymal transition (EMT). The involvement of EMT in the occurrence and development of tumors through invasion and metastasis in TNBC has been a matter of concern. Therefore, EMT markers could be prognostic predictors and potential therapeutic targets in TNBC. Chemotherapy has been one of the primary options for treating patients with TNBC, but its efficacy against TNBC is still limited. Targeted therapy is a critical emerging option with enhanced efficacy and less adverse effects on patients. Various targeted therapy approaches have been developed based on the specific molecules and the signaling pathways involved in TNBC. These include inhibitors of signaling pathways such as TGF-β, Wnt/β-catenin, Notch, TNF-α/NF-κB and EGFR, as well as immune checkpoint inhibitors, such as pembrolizumab, 2laparib, and talazoparib have been widely explored. This article reviews recent developments in EMT in TNBC invasion and metastasis and potential targeted therapy strategies.
Collapse
Affiliation(s)
| | | | - Manoj K. Mishra
- Cancer Research Center, Department of Biological Sciences, Alabama State
University, Montgomery, AL, United States
| |
Collapse
|
6
|
Song B, Wei F, Peng J, Wei X, Liu M, Nie Z, Ma Y, Peng T. Icariin Regulates EMT and Stem Cell-Like Character in Breast Cancer through Modulating lncRNA NEAT1/TGFβ/SMAD2 Signaling Pathway. Biol Pharm Bull 2024; 47:399-410. [PMID: 38220208 DOI: 10.1248/bpb.b23-00668] [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: 01/16/2024]
Abstract
Metastases and drug resistance are the major risk factors associated with breast cancer (BC), which is the most common type of tumor affecting females. Icariin (ICA) is a traditional Chinese medicine compound that possesses significant anticancer properties. Long non-coding RNAs (lncRNAs) are involved in a wide variety of biological and pathological processes and have been shown to modulate the effectiveness of certain drugs in cancer. The purpose of this study was to examine the potential effect of ICA on epithelial mesenchymal transition (EMT) and stemness articulation in BC cells, as well as the possible relationship between its inhibitory action on EMT and stemness with the NEAT1/transforming growth factor β (TGFβ)/SMAD2 pathway. The effect of ICA on the proliferation (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and colony assays), EMT (Western blotting, immunofluorescence, and wound healing), and stemness (mammosphere formation assays, Western blotting) of BC cells were examined. According to the findings, ICA suppressed the proliferation, EMT, and stem cell-like in MDA-MB-231 cells, and exerted its inhibitory impact by downregulating the TGFβ/SMAD2 signaling pathway. ICA could significantly downregulate the expression of lncRNA NEAT1, and silencing NEAT1 enhanced the effect of ICA in suppressing EMT and expression of different stem cell markers. In addition, silencing NEAT1 was found to attenuate the TGFβ/SMAD2 signaling pathway, thereby improving the inhibitory impact of ICA on stemness and EMT in BC cells. In conclusion, ICA can potentially inhibit the metastasis of BC via affecting the NEAT1/TGFβ/SMAD2 pathway, which provides a theoretical foundation for understanding the mechanisms involved in potential application of ICA for BC therapy.
Collapse
Affiliation(s)
- Bo Song
- School of Third Clinical Medicine, Shanxi University of Chinese Medicine
| | - Fuxia Wei
- School of Third Clinical Medicine, Shanxi University of Chinese Medicine
| | - Jiehao Peng
- School of Third Clinical Medicine, Shanxi University of Chinese Medicine
| | - Xiuhong Wei
- School of Basic Medical Sciences, Shanxi University of Chinese Medicine
| | - Mingran Liu
- School of Basic Medical Sciences, Shanxi University of Chinese Medicine
| | - Zhongbiao Nie
- Pharmaceutical Department, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University
| | - Yanmiao Ma
- School of Basic Medical Sciences, Shanxi University of Chinese Medicine
| | - Tao Peng
- Famous Chinese Medicine Studio, Shanxi Hospital of Integrated Traditional Chinese and Western Medicine
- Shanxi Provincial Key Laboratory of Classical Prescription Strengthening Yang, Shanxi Hospital of Integrated Traditional Chinese and Western Medicine
| |
Collapse
|
7
|
Wang C, Zhao X, Zhao L, Wang Y, Jia Y, Zhang X, Ma W. PKCζ phosphorylates VASP to mediate chemotaxis in breast cancer cells. Exp Cell Res 2023; 433:113823. [PMID: 37890607 DOI: 10.1016/j.yexcr.2023.113823] [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: 06/02/2023] [Revised: 09/17/2023] [Accepted: 10/15/2023] [Indexed: 10/29/2023]
Abstract
Breast carcinoma (BC) is one of the most common malignant cancers in females, and metastasis remains the leading cause of death in these patients. Chemotaxis plays an important role in cancer cell metastasis and the mechanism of breast cancer chemotaxis has become a central issue in contemporary research. PKCζ, a member of the atypical PKC family, has been reported to be an essential component of the EGF-stimulated chemotactic signaling pathway. However, the molecular mechanism through which PKCζ regulates chemotaxis remains unclear. Here, we used a proteomic approach to identify PKCζ-interacting proteins in breast cancer cells and identified VASP as a potential binding partner. Intriguingly, stimulation with EGF enhanced this interaction and induced the translocalization of PKCζ and VASP to the cell membrane. Further experiments showed that PKCζ catalyzes the phosphorylation of VASP at Ser157, which is critical for the biological function of VASP in regulating chemotaxis and actin polymerization in breast cancer cells. Furthermore, in PKCζ knockdown BC cells, the enrichment of VASP at the leading edge was reduced, and its interaction with profilin1 was attenuated, thereby reducing the chemotaxis and overall motility of breast cancer cells after EGF treatment. In functional assays, PKCζ promoted chemotaxis and motility of BC cells through VASP. Our findings demonstrate that PKCζ, a new kinase of VASP, plays an important role in promoting breast cancer metastasis and provides a theoretical basis for expanding new approaches to tumor biotherapy.
Collapse
Affiliation(s)
- Chunqing Wang
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Laboratory Medicine China
| | - Xiaoqing Zhao
- Department of Clinical Laboratory Medicine, The Second Hospital, Cheeloo College of Medicine, Shandong University, 250012 Jinan, Shandong China
| | - Liqing Zhao
- Department of Pediatrics, Zaozhuang Municipal Hospital, Zaozhuang, Shandong 277000, China
| | - Yunqiu Wang
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Laboratory Medicine China
| | - Yan Jia
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China.
| | - Xiaofang Zhang
- Department of Clinical Laboratory Medicine, Tianjin Medical University General Hospital, Tianjin, China.
| | - Wanshan Ma
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Laboratory Medicine China.
| |
Collapse
|
8
|
Whitford MKM, McCaffrey L. Polarity in breast development and cancer. Curr Top Dev Biol 2023; 154:245-283. [PMID: 37100520 DOI: 10.1016/bs.ctdb.2023.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2023]
Abstract
Mammary gland development and breast cancer progression are associated with extensive remodeling of epithelial tissue architecture. Apical-basal polarity is a key feature of epithelial cells that coordinates key elements of epithelial morphogenesis including cell organization, proliferation, survival, and migration. In this review we discuss advances in our understanding of how apical-basal polarity programs are used in breast development and cancer. We describe cell lines, organoids, and in vivo models commonly used for studying apical-basal polarity in breast development and disease and discuss advantages and limitations of each. We also provide examples of how core polarity proteins regulate branching morphogenesis and lactation during development. We describe alterations to core polarity genes in breast cancer and their associations with patient outcomes. The impact of up- or down-regulation of key polarity proteins in breast cancer initiation, growth, invasion, metastasis, and therapeutic resistance are discussed. We also introduce studies demonstrating that polarity programs are involved in regulating the stroma, either through epithelial-stroma crosstalk, or through signaling of polarity proteins in non-epithelial cell types. Overall, a key concept is that the function of individual polarity proteins is highly contextual, depending on developmental or cancer stage and cancer subtype.
Collapse
Affiliation(s)
- Mara K M Whitford
- Goodman Cancer Institute, McGill University, Montreal, Quebec, Canada; Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | - Luke McCaffrey
- Goodman Cancer Institute, McGill University, Montreal, Quebec, Canada; Department of Biochemistry, McGill University, Montreal, Quebec, Canada; Gerald Bronfman Department of Oncology, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
9
|
Wang HF, Jiang J, Wu JS, Zhang M, Pang X, Dai L, Tang YL, Liang XH. Hypermethylation of PRKCZ Regulated by E6 Inhibits Invasion and EMT via Cdc42 in HPV-Related Head and Neck Squamous Cell Carcinoma. Cancers (Basel) 2022; 14:cancers14174151. [PMID: 36077689 PMCID: PMC9454700 DOI: 10.3390/cancers14174151] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 08/18/2022] [Accepted: 08/24/2022] [Indexed: 12/09/2022] Open
Abstract
Purpose: To study the role of target genes with aberrant DNA methylation in HPV+ HNSCC. Methods: A HumanMethylation450 BeadChip array (Illumina) was used to identify differentially methylated genes. CCK-8, flow cytometry, wound healing, and cell invasion assays were conducted to analyze the biological roles of PRKCZ. Western blot, qRT-PCR, immunohistochemistry, and animal studies were performed to explore the mechanisms underlying the functions of PRKCZ. Results: We selected PRKCZ, which is associated with HPV infection, as our target gene. PRKCZ was hypermethylated in HPV+ HNSCC patients, and PRKCZ methylation status was negatively related to the pathological grading of HNSCC patients. Silencing PRKCZ inhibited the malignant capacity of HPV+ HNSCC cells. Mechanistically, HPV might promote DNMT1 expression via E6 to increase PRKCZ methylation. Cdc42 was required for the PRKCZ-mediated mechanism of action, contributing to the occurrence of epithelial-mesenchymal transition (EMT) in HPV+ HNSCC cells. In addition, blocking PRKCZ delayed tumor growth in HPV16-E6/E7 transgenic mice. Cdc42 expression was decreased, whereas E-cadherin levels increased. Conclusion: We suggest that PRKCZ hypermethylation induces EMT via Cdc42 to act as a potent tumor promoter in HPV+ HNSCC.
Collapse
Affiliation(s)
- Hao-Fan Wang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Jian Jiang
- Department of Head and Neck Surgery, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610041, China
| | - Jia-Shun Wu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Mei Zhang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xin Pang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Li Dai
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Ya-Ling Tang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, Department of Pathology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Correspondence: (Y.-L.T.); (X.-H.L.)
| | - Xin-Hua Liang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Correspondence: (Y.-L.T.); (X.-H.L.)
| |
Collapse
|
10
|
MicroRNA-582-5p regulates cell invasion in bladder cancer through targeting Forkhead Box G1. Pathol Res Pract 2021; 230:153752. [PMID: 34979422 DOI: 10.1016/j.prp.2021.153752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 12/19/2021] [Accepted: 12/21/2021] [Indexed: 11/21/2022]
Abstract
Burgeoning evidence shows that microRNAs (miRNAs) are associated with tumorigenesis and progression. However, the alteration and function of many miRNAs in bladder cancer (BCa) are not clear. Here, we explored the regulatory effect of microRNA-582 (miR-582) on cell invasion in BCa and underlying mechanisms. The expression of miR-582 in BCa tissues and cell lines was examined by quantitative real-time PCR (qRT-PCR). The target gene of miR-582 and their binding site were predicted by bioinformatics analysis. Luciferase reporter assay and western blot analysis were performed to confirm miR-582 directly targeting Forkhead Box G1 (FOXG1). The role of miR-582-FOXG1 axis in regulating BCa invasion was evaluated in cell models. The association of miR-582 with clinicopathologic features and prognosis was analyzed. Experimental results indicated that miR-582 was downregulated in BCa tissues and cell lines. Forced miR-582 decreased cell invasion, regulating expression levels of invasion-related proteins, such as MMP2, MMP9 and ZO-1. MiR-582 directly targeted FOXG1 by binding to its 3'UTR. Overexpression of FOXG1 rescued the regulating function in BCa cells induced by miR-582. Moreover, miR-582-FOXG1 axis has obvious clinical relevance with prognosis in BCa patients. Our results indicate that miR-582-FOXG1 axis may act as a key role on cell invasion and serve as a potential prognostic predicted biomarker.
Collapse
|
11
|
Tandon M, Othman AH, Winogradzki M, Pratap J. Bone metastatic breast cancer cells display downregulation of PKC-ζ with enhanced glutamine metabolism. Gene 2021; 775:145419. [PMID: 33444686 DOI: 10.1016/j.gene.2021.145419] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 10/28/2020] [Accepted: 01/05/2021] [Indexed: 11/20/2022]
Abstract
BACKGROUND Breast cancer is the most commonly diagnosed cancer among women and its metastases results in poor survival rates in patients. The ability to alter metabolism is a key attribute cancer cells use to survive within different metastatic microenvironments and cause organ failure. We hypothesized that evaluation of metabolic alterations within tumor cells could provide a better understanding of cancer metastasis. Therefore, to investigate underlying metabolic alterations during metastases, we utilized human MDA-MB-231 and mouse 4T1 models that closely mimic human breast cancer metastasis. METHODS The glycolysis and glutamine pathway-related changes were examined in bone metastatic cells by XF-24 extracellular flux analyzer and western blotting. The expression levels of genes related to metabolism were examined by PCR arrays. RESULTS The MDA-MB-231 cells isolated after bone metastases showed reduced glucose uptake and glycolysis compared to parental cells, suggesting that these cells could alter metabolic requirements for survival. To understand these metabolic changes, we investigated glutamine, a common and naturally occurring non-essential amino acid. Interestingly, in reduced glucose conditions both cell lines showed dependence on glutamine for cell survival, and with glutamine withdrawal significantly increasing apoptotic cell death. Glutamine was also critical for normal cell proliferation even in the presence of high glucose concentrations. To further understand this metabolic switch in metastatic cells, we examined the genes related to metabolism and identified a more than seven-fold downregulation of protein kinase C zeta (PKC-ζ) expression levels in bone-derived MDA-MB-231 cells compared to the parental population. The PKC-ζ levels were also significantly reduced in metastatic 4T1 cells compared to non-metastatic MT1A2 cells. Since PKC-ζ deficiency promotes glutamine utilization via the serine biosynthesis pathway, we examined glutamine metabolism. The ectopic expression of PKC-ζ inhibited glutamine conversion to glutamate, while mutant PKC-ζ reversed this effect. Furthermore, the gene expression levels of enzymes involved in serine biosynthesis, phosphoserine phosphatase (PSPH), phosphoserine aminotransferase (PSAT1), and phosphoglycerate dehydrogenase (PHGDH) showed upregulation following glucose deprivation with PKC-ζ deficiency. The PHGDH upregulation was inhibited by ectopically expressing wild type but not mutated PKC-ζ in glucose-deprived conditions. CONCLUSIONS Our results support the upregulation of serine biosynthesis pathway genes and downregulation of PKC-ζ as potential metabolic alterations for bone metastatic breast cancer cells.
Collapse
Affiliation(s)
- Manish Tandon
- Suite 507, Armour Academic Building, Cell & Molecular Medicine, Rush University Medical Center, Chicago, IL 60612, United States
| | - Ahmad H Othman
- Suite 507, Armour Academic Building, Cell & Molecular Medicine, Rush University Medical Center, Chicago, IL 60612, United States
| | - Marcus Winogradzki
- Suite 507, Armour Academic Building, Cell & Molecular Medicine, Rush University Medical Center, Chicago, IL 60612, United States
| | - Jitesh Pratap
- Suite 507, Armour Academic Building, Cell & Molecular Medicine, Rush University Medical Center, Chicago, IL 60612, United States.
| |
Collapse
|
12
|
Pratt SJP, Hernández-Ochoa E, Martin SS. Calcium signaling: breast cancer's approach to manipulation of cellular circuitry. Biophys Rev 2020; 12:1343-1359. [PMID: 33569087 PMCID: PMC7755621 DOI: 10.1007/s12551-020-00771-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 10/29/2020] [Indexed: 12/11/2022] Open
Abstract
Calcium is a versatile element that participates in cell signaling for a wide range of cell processes such as death, cell cycle, division, migration, invasion, metabolism, differentiation, autophagy, transcription, and others. Specificity of calcium in each of these processes is achieved through modulation of intracellular calcium concentrations by changing the characteristics (amplitude/frequency modulation) or location (spatial modulation) of the signal. Breast cancer utilizes calcium signaling as an advantage for survival and progression. This review integrates evidence showing that increases in expression of calcium channels, GPCRs, pumps, effectors, and enzymes, as well as resulting intracellular calcium signals, lead to high calcium and/or an elevated calcium- mobilizing capacity necessary for malignant functions such as migratory, invasive, proliferative, tumorigenic, or metastatic capacities.
Collapse
Affiliation(s)
- Stephen J P Pratt
- Program in Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD USA.,Department of Physiology, University of Maryland School of Medicine, Baltimore, MD USA.,Marlene and Stewart Greenebaum NCI Comprehensive Cancer Center, University of Maryland School of Medicine, 655 W. Baltimore Street, Bressler Research Building, Rm 10-020 D, Baltimore, MD 21201 USA
| | - Erick Hernández-Ochoa
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD USA
| | - Stuart S Martin
- Program in Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD USA.,Department of Physiology, University of Maryland School of Medicine, Baltimore, MD USA.,Marlene and Stewart Greenebaum NCI Comprehensive Cancer Center, University of Maryland School of Medicine, 655 W. Baltimore Street, Bressler Research Building, Rm 10-020 D, Baltimore, MD 21201 USA
| |
Collapse
|
13
|
He J, Xue Y, Wang Q, Zhou X, Liu L, Zhang T, Shang C, Ma J, Ma T. Long non-coding RNA MIAT regulates blood tumor barrier permeability by functioning as a competing endogenous RNA. Cell Death Dis 2020; 11:936. [PMID: 33127881 PMCID: PMC7603350 DOI: 10.1038/s41419-020-03134-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 10/12/2020] [Accepted: 10/14/2020] [Indexed: 12/12/2022]
Abstract
Blood-tumor barrier (BTB) presents a major obstacle to brain drug delivery. Therefore, it is urgent to enhance BTB permeability for the treatment of glioma. In this study, we demonstrated that MIAT, ZAK, and phosphorylated NFκB-p65 (p-NFκB-p65) were upregulated, while miR-140-3p was downregulated in glioma-exposed endothelial cells (GECs) of BTB compared with those in endothelial cells cocultured with astrocytes (ECs) of blood-brain barrier (BBB). MIAT inhibited miR-140-3p expression, increased the expression of ZAK, enhanced the ratio of p-NFκB-p65:NFκB-p65, and promoted the endothelial leakage of BTB. Our current study revealed that miR-140-3p was complementary to the ZAK 3'untranslated regions (3'-UTR), and luciferase activity of ZAK was inhibited by miR-140-3p in 293T cells. MiR-140-3p silencing resulted in an increase in BTB permeability by targeting ZAK, while overexpression of miR-140-3p had the opposite results in GECs of BTB. Overexpression of ZAK induced an increase in BTB permeability, and this effect was related to ZAK's ability to mediate phosphorylation of NFκB-p65. Conversely, ZAK silencing get opposite results in GECs of BTB. As a molecular sponge of miR-140-3p, MIAT attenuated its negative regulation of the target gene ZAK by adsorbing miR-140-3p. P-NFκB-p65 as a transcription factor negatively regulated the expression of TJ-associated proteins by means of chip assay and luciferase assay. Single or combined application of MIAT and miR-140-3p effectively promoted antitumor drug doxorubicin (Dox) across BTB to induce apoptosis of glioma cells. In summary, MIAT functioned as a miR-140-3p sponge to regulate the expression of its target gene ZAK, which contribution to phosphorylation of NFκB-p65 was associated with an increase in BTB permeability by down-regulating the expression of TJ associated proteins, thereby promoting Dox delivery across BTB. These results might provide a novel strategy and target for chemotherapy of glioma.
Collapse
Affiliation(s)
- Jiayuan He
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, and Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, 110122, China
| | - Yixue Xue
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, and Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, 110122, China
| | - Qingyuan Wang
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, and Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, 110122, China
| | - Xinxin Zhou
- Liaoning University of Traditional Chinese Medicine, Shenyang, 110034, China
| | - Libo Liu
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, and Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, 110122, China
| | - Tianyuan Zhang
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, and Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, 110122, China
| | - Chao Shang
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, and Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, 110122, China
| | - Jun Ma
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, and Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, 110122, China
| | - Teng Ma
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China. .,Key Laboratory of Cell Biology, Ministry of Public Health of China, and Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, 110122, China.
| |
Collapse
|
14
|
Activity to Breast Cancer Cell Lines of Different Malignancy and Predicted Interaction with Protein Kinase C Isoforms of Royleanones. Int J Mol Sci 2020; 21:ijms21103671. [PMID: 32456148 PMCID: PMC7279380 DOI: 10.3390/ijms21103671] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/21/2020] [Accepted: 05/21/2020] [Indexed: 12/17/2022] Open
Abstract
Plants have been used for centuries to treat several illnesses. The Plectranthus genus has a vast variety of species that has allowed the isolation of cytotoxic compounds with notable activities. The abietane diterpenes 6,7-dehydroroyleanone (DeRoy, 1), 7α-acetoxy-6β-hydroxyroyleanone (Roy, 2), and Parvifloron D (ParvD, 3) were obtained from Plectranthus spp. and showed promising biological activities, such as cytotoxicity. The inhibitory effects of the different natural abietanes (1-3) were compared in MFC7, SkBr3, and SUM159 cell lines, as well as SUM159 grown in cancer stem cell-inducing conditions. Based on the royleanones’ bioactivity, the derivatives RoyBz (4), RoyBzCl (5), RoyPr2 (6), and DihydroxyRoy (7), previously obtained from 2, were selected for further studies. Protein kinases C (PKCs) are involved in several carcinogenic processes. Thus, PKCs are potential targets for cancer therapy. To date, the portfolio of available PKC modulators remains very limited due to the difficulty of designing isozyme-selective PKC modulators. As such, molecular docking was used to evaluate royleanones 1-6 as predicted isozyme-selective PKC binders. Subtle changes in the binding site of each PKC isoform change the predicted interaction profiles of the ligands. Subtle changes in royleanone substitution patterns, such as a double substitution only with non-substituted phenyls, or hydroxybenzoate at position four that flips the binding mode of ParvD (3), can increase the predicted interactions in certain PKC subtypes.
Collapse
|
15
|
Clayton NS, Ridley AJ. Targeting Rho GTPase Signaling Networks in Cancer. Front Cell Dev Biol 2020; 8:222. [PMID: 32309283 PMCID: PMC7145979 DOI: 10.3389/fcell.2020.00222] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 03/16/2020] [Indexed: 12/16/2022] Open
Abstract
As key regulators of cytoskeletal dynamics, Rho GTPases coordinate a wide range of cellular processes, including cell polarity, cell migration, and cell cycle progression. The adoption of a pro-migratory phenotype enables cancer cells to invade the stroma surrounding the primary tumor and move toward and enter blood or lymphatic vessels. Targeting these early events could reduce the progression to metastatic disease, the leading cause of cancer-related deaths. Rho GTPases play a key role in the formation of dynamic actin-rich membrane protrusions and the turnover of cell-cell and cell-extracellular matrix adhesions required for efficient cancer cell invasion. Here, we discuss the roles of Rho GTPases in cancer, their validation as therapeutic targets and the challenges of developing clinically viable Rho GTPase inhibitors. We review other therapeutic targets in the wider Rho GTPase signaling network and focus on the four best characterized effector families: p21-activated kinases (PAKs), Rho-associated protein kinases (ROCKs), atypical protein kinase Cs (aPKCs), and myotonic dystrophy kinase-related Cdc42-binding kinases (MRCKs).
Collapse
Affiliation(s)
- Natasha S Clayton
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Anne J Ridley
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
16
|
González-Mariscal L, Miranda J, Gallego-Gutiérrez H, Cano-Cortina M, Amaya E. Relationship between apical junction proteins, gene expression and cancer. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183278. [PMID: 32240623 DOI: 10.1016/j.bbamem.2020.183278] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 01/09/2020] [Accepted: 03/06/2020] [Indexed: 12/11/2022]
Abstract
The apical junctional complex (AJC) is a cell-cell adhesion system present at the upper portion of the lateral membrane of epithelial cells integrated by the tight junction (TJ) and the adherens junction (AJ). This complex is crucial to initiate and stabilize cell-cell adhesion, to regulate the paracellular transit of ions and molecules and to maintain cell polarity. Moreover, we now consider the AJC as a hub of signal transduction that regulates cell-cell adhesion, gene transcription and cell proliferation and differentiation. The molecular components of the AJC are multiple and diverse and depending on the cellular context some of the proteins in this complex act as tumor suppressors or as promoters of cell transformation, migration and metastasis outgrowth. Here, we describe these new roles played by TJ and AJ proteins and their potential use in cancer diagnostics and as targets for therapeutic intervention.
Collapse
Affiliation(s)
- Lorenza González-Mariscal
- Department of Physiology, Biophysics and Neuroscience, Center of Research and Advanced Studies (Cinvestav), Mexico City, Mexico.
| | - Jael Miranda
- Department of Physiology, Biophysics and Neuroscience, Center of Research and Advanced Studies (Cinvestav), Mexico City, Mexico
| | - Helios Gallego-Gutiérrez
- Department of Physiology, Biophysics and Neuroscience, Center of Research and Advanced Studies (Cinvestav), Mexico City, Mexico
| | - Misael Cano-Cortina
- Department of Physiology, Biophysics and Neuroscience, Center of Research and Advanced Studies (Cinvestav), Mexico City, Mexico
| | - Elida Amaya
- Department of Physiology, Biophysics and Neuroscience, Center of Research and Advanced Studies (Cinvestav), Mexico City, Mexico
| |
Collapse
|
17
|
PAICS, a Purine Nucleotide Metabolic Enzyme, is Involved in Tumor Growth and the Metastasis of Colorectal Cancer. Cancers (Basel) 2020; 12:cancers12040772. [PMID: 32218208 PMCID: PMC7226071 DOI: 10.3390/cancers12040772] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 03/18/2020] [Accepted: 03/20/2020] [Indexed: 12/15/2022] Open
Abstract
The identification of colorectal cancer (CRC) molecular targets is needed for the development of drugs that improve patient survival. We investigated the functional role of phosphoribosylaminoimidazole carboxylase, phosphoribosylaminoimidazole succinocarboxamide synthetase (PAICS), a de novo purine biosynthetic enzyme involved in DNA synthesis, in CRC progression and metastasis by using cell and animal models. Its clinical utility was assessed in human CRC samples. The expression of PAICS was regulated by miR-128 and transcriptionally activated by Myc in CRC cells. Increased expression of PAICS was involved in proliferation, migration, growth, and invasion of CRC cells irrespective of the p53 and microsatellite status. In mice, the depletion of PAICS in CRC cells led to reduced tumor growth and metastatic cell dissemination to the liver, lungs, and bone. Positron emission tomography imaging showed significantly reduced metastatic lesions in stable PAICS knockdown CRC cells. In cells with PAICS knockdown, there was upregulation of the epithelial mesenchymal transition marker, E-cadherin, and bromodomain inhibitor, JQ1, can target its increased expression by blocking Myc. PAICS was overexpressed in 70% of CRCs, and was associated with poor 5-year survival independent of the pathologic stage, patient’s race, gender, and age. Overall, the findings point to the usefulness of PAICS targeting in the treatment of aggressive colorectal cancer.
Collapse
|
18
|
Fomicheva M, Tross EM, Macara IG. Polarity proteins in oncogenesis. Curr Opin Cell Biol 2019; 62:26-30. [PMID: 31509786 DOI: 10.1016/j.ceb.2019.07.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 07/19/2019] [Accepted: 07/24/2019] [Indexed: 02/02/2023]
Abstract
Most human cancers arise from epithelial tissues, which are apical-basally polarized and possess intercellular adhesive junctions. Epithelial cells grow to characteristic densities, often from proliferative progenitors, which arrest as they mature. Homeostatic mechanisms can maintain this characteristic density if it is exceeded (crowding) or is too low (e.g. in response to wounding). During tumor initiation and progression this homeostatic mechanism is lost. Some aspects of cell polarity are also lost, although many carcinomas retain intercellular junctions and even apical domains. In other cases, and particularly in recurrent tumors, however, the cells become predominantly mesenchymal. A major question, still only incompletely answered, is whether the proteins that determine cell polarity function as tumor suppressors or tumor promoters. Here we discuss recent advances in understanding the role of polarity proteins and homeostasis in cancer.
Collapse
Affiliation(s)
- Maria Fomicheva
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37424, USA
| | - Erica M Tross
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37424, USA
| | - Ian G Macara
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37424, USA.
| |
Collapse
|
19
|
Kim KH, Chung C, Kim JM, Lee D, Cho SY, Lee TH, Cho HJ, Yeo MK. Clinical significance of atypical protein kinase C (PKCι and PKCζ) and its relationship with yes-associated protein in lung adenocarcinoma. BMC Cancer 2019; 19:804. [PMID: 31412817 PMCID: PMC6693135 DOI: 10.1186/s12885-019-5992-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Accepted: 07/30/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Protein kinase C iota (PKCι) and protein kinase C zeta (PKCζ) are two atypical protein kinase (aPKC) enzymes that contribute to cell proliferation and cancer development. The Hippo/YAP pathway is commonly disrupted and upregulated in cancers. Herein, the expression patterns and clinical relevance of PKCι and PKCζ are evaluated in relation to YAP, a downstream effector of Hippo, in lung adenocarcinoma (LAC). The protein and mRNA expression levels of PKCι, PKCζ, YAP, and their phosphorylated forms, namely p-PKCι, p-PKCζ and p-YAP, are evaluated in relation to clinicopathological factors, including patient survival. METHODS A total of 200 primary LAC tissue samples were examined by immunohistochemistry for PKCι, p-PKCι, PKCζ, p-PKCζ, YAP, and p-YAP protein expression. Sixty pairs of LAC and non-neoplastic lung tissue samples were assessed for PRKCI, PRKCZ, and YAP mRNA levels. PKCι, p-PKCι, PKCζ, and p-PKCζ protein expression were evaluated by Western blot analysis in the PC9 and PC9/GR LAC cell lines with YAP modulation. RESULTS LAC demonstrated cytoplasmic PKCι, p-PKCι, PKCζ, and p-PKCζ immunostaining patterns. Positive aPKC protein expressions were related with poor patient survival. Especially, increased p-PKCι protein expression was significantly correlated with higher pathological stage and shortened overall survival. YAP overexpression contributes phosphorylation of PKCι and PKCζ protein expression in the LAC cell line. CONCLUSIONS PKCι and PKCζ are related to YAP in LAC. PKCι and PKCζ play distinct roles in LAC; specifically, p-PKCι overexpression is suggested to underlie factors that indicate a poor prognosis.
Collapse
Affiliation(s)
- Kyung-Hee Kim
- Department of Pathology, Chungnam National University School of Medicine, Munwha-ro 266, Jung-gu, Daejeon, 35015, Republic of Korea
| | - Chaeuk Chung
- Division of Pulmonology, Department of Internal Medicine, College of Medicine, Chungnam National University, Daejeon, 35015, Republic of Korea
| | - Jin-Man Kim
- Department of Pathology, Chungnam National University School of Medicine, Munwha-ro 266, Jung-gu, Daejeon, 35015, Republic of Korea
| | - Dahye Lee
- Division of Pulmonology, Department of Internal Medicine, College of Medicine, Chungnam National University, Daejeon, 35015, Republic of Korea
| | - Sang Yeon Cho
- School of Medicine, Chungnam National University, Munwha-ro 266, Jung-gu, Daejeon, Republic of Korea
| | - Tae Hee Lee
- The Biobank of Chungnam National University Hospital, Munwha-ro 282, Jung-gu, Daejeon, Republic of Korea
| | - Hyun Jin Cho
- Department of Thoracic Surgery, College of Medicine, Chungnam National University, Daejeon, 35015, Republic of Korea
| | - Min-Kyung Yeo
- Department of Pathology, Chungnam National University School of Medicine, Munwha-ro 266, Jung-gu, Daejeon, 35015, Republic of Korea.
| |
Collapse
|
20
|
Wong WY, Allie S, Limesand KH. PKCζ and JNK signaling regulate radiation-induced compensatory proliferation in parotid salivary glands. PLoS One 2019; 14:e0219572. [PMID: 31287841 PMCID: PMC6615637 DOI: 10.1371/journal.pone.0219572] [Citation(s) in RCA: 15] [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: 03/01/2019] [Accepted: 06/26/2019] [Indexed: 01/05/2023] Open
Abstract
Radiotherapy is a common treatment option for head and neck cancer patients; however, the surrounding healthy salivary glands are often incidentally irradiated during the process. As a result, patients often experience persistent xerostomia and hyposalivation, which deceases their quality of life. Clinically, there is currently no standard of care available to restore salivary function. Repair of epithelial wounds involves cellular proliferation and establishment of polarity in order to regenerate the tissue. This process is partially mediated by protein kinase C zeta (PKCζ), an apical polarity regulator; however, its role following radiation damage is not completely understood. Using an in vivo radiation model, we show a significant decrease in active PKCζ in irradiated murine parotid glands, which correlates with increased proliferation that is sustained through 30 days post-irradiation. Additionally, salivary glands in PKCζ null mice show increased basal proliferation which radiation treatment did not further potentiate. Radiation damage also activates Jun N-terminal kinase (JNK), a proliferation-inducing mitogen-activated protein kinase normally inhibited by PKCζ. In both a PKCζ null mouse model and in primary salivary gland cell cultures treated with a PKCζ inhibitor, there was increased JNK activity and production of downstream proliferative transcripts. Collectively, these findings provide a potential molecular link by which PKCζ suppression following radiation damage promotes JNK activation and radiation-induced compensatory proliferation in the salivary gland.
Collapse
Affiliation(s)
- Wen Yu Wong
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, Arizona, United States of America
| | - Sydney Allie
- Department of Nutritional Sciences, University of Arizona, Tucson, Arizona, United States of America
| | - Kirsten H. Limesand
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, Arizona, United States of America
- Department of Nutritional Sciences, University of Arizona, Tucson, Arizona, United States of America
| |
Collapse
|
21
|
KIBRA Team Up with Partners to Promote Breast Cancer Metastasis. Pathol Oncol Res 2019; 26:627-634. [DOI: 10.1007/s12253-019-00660-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 04/01/2019] [Indexed: 02/06/2023]
|
22
|
Smalley T, Islam SMA, Apostolatos C, Apostolatos A, Acevedo-Duncan M. Analysis of PKC-ζ protein levels in normal and malignant breast tissue subtypes. Oncol Lett 2018; 17:1537-1546. [PMID: 30675210 PMCID: PMC6341665 DOI: 10.3892/ol.2018.9792] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 11/09/2018] [Indexed: 12/18/2022] Open
Abstract
It is estimated that breast cancer will be the second leading cause of cancer-associated mortality in women in 2018. Previous research has demonstrated that the atypical protein kinase C-ζ (PKC-ζ) is a component of numerous dysregulated pathways in breast cancer, including cellular proliferation, survival, and cell cycle upregulation. The present study investigated the PKC-ζ protein in breast tissue to evaluate its potential as a biomarker for breast cancer invasion, and demonstrated that an overexpression of PKC-ζ protein can be indicative of carcinogenesis. The present study analyzed the expression of PKC-ζ in individuals with no tumor complications and malignant female human breast tissue samples (lobular carcinoma in situ, invasive lobular carcinoma, ductal carcinoma in situ and invasive ductal carcinoma) with the use of western blot analysis, immunohistochemistry and statistical analysis (83 samples). The present study also evaluated the invasive behavior of MDA-MB-231 breast cancer cells following the knockdown of PKC-ζ with a Transwell invasion assay and an immunofluorescent probe for filamentous actin (F-actin) organization. The data demonstrated that PKC-ζ expression was identified to be higher in invading tissues when compared with non-invading tissues. The results also suggest that PKC-ζ is more abundant in ductal tissues when compared with lobular tissues. In addition, the protein studies also suggest that PKC-ζ is a component for invasive behavior through the Ras-related C3 botulinum toxin substrate 1 (Rac1) and Ras homolog gene family member A (RhoA) pathway, and PKC-ζ is required for the F-actin reorganization in invasive cells. Therefore, PKC-ζ should be considered to be a biomarker in the development of breast cancer as well as an indicator of invading tumor cells.
Collapse
Affiliation(s)
- Tracess Smalley
- Department of Chemistry, The University of South Florida, Tampa, FL 33620, USA
| | - S M Anisul Islam
- Department of Chemistry, The University of South Florida, Tampa, FL 33620, USA
| | | | - André Apostolatos
- Department of Chemistry, The University of South Florida, Tampa, FL 33620, USA
| | | |
Collapse
|
23
|
Nikitin PV, Potapov AA, Ryzhova MV, Shurkhay VA, Kulikov EE, Zhvanskiy ES, Popov IA, Nikolaev EN. [The role of lipid metabolism disorders, atypical isoforms of protein kinase C, and mutational status of cytosolic and mitochondrial forms of isocitrate dehydrogenase in carcinogenesis of glial tumors]. ZHURNAL VOPROSY NEĬROKHIRURGII IMENI N. N. BURDENKO 2018; 82:112-120. [PMID: 29927433 DOI: 10.17116/neiro2018823112] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The relationship between molecular genetic and metabolic disorders is one of the challenges of modern oncology. In this review, we consider lipid metabolism and its changes as one of the factors of oncogenesis of glial tumors. Also, we demonstrate that the genome and the metabolome are interconnected by a large number of links, and the metabolic pathways, during their reorganization, are able to drastically affect the genetic structure of the cell and, in particular, cause its tumor transformation. Our own observations and analysis of the literature data allow us to conclude that mass spectrometry is a highly accurate current method for assessing metabolic disorders at the cellular level. The use of mass spectrometry during surgery allows the neurosurgeon to obtain real-time data on the level of specific molecular markers in the resected tissue, thereby bringing intraoperative navigation techniques to the molecular level. The generation of molecular fingerprints for each tumor significantly complements the available neuroimaging, molecular genetic, and immunohistochemical data.
Collapse
Affiliation(s)
- P V Nikitin
- Burdenko Neurosurgery Institute, 4-ya Tverskaya-Yamskaya Str., 16, Moscow, Russia, 125047
| | - A A Potapov
- Burdenko Neurosurgery Institute, 4-ya Tverskaya-Yamskaya Str., 16, Moscow, Russia, 125047
| | - M V Ryzhova
- Burdenko Neurosurgery Institute, 4-ya Tverskaya-Yamskaya Str., 16, Moscow, Russia, 125047
| | - V A Shurkhay
- Burdenko Neurosurgery Institute, 4-ya Tverskaya-Yamskaya Str., 16, Moscow, Russia, 125047; Moscow Institute of Physics and Technology, Institutskiy Pereulok, 9, Dolgoprudny, Moscow Region, Russia, 141701
| | - E E Kulikov
- Moscow Institute of Physics and Technology, Institutskiy Pereulok, 9, Dolgoprudny, Moscow Region, Russia, 141701; Federal Research Center 'Fundamentals of Biotechnology', Leninskiy Prospect, 33/2, Moscow, Russia, 119071
| | - E S Zhvanskiy
- Moscow Institute of Physics and Technology, Institutskiy Pereulok, 9, Dolgoprudny, Moscow Region, Russia, 141701
| | - I A Popov
- Moscow Institute of Physics and Technology, Institutskiy Pereulok, 9, Dolgoprudny, Moscow Region, Russia, 141701
| | - E N Nikolaev
- Moscow Institute of Physics and Technology, Institutskiy Pereulok, 9, Dolgoprudny, Moscow Region, Russia, 141701; Skolkovo Institute of Science and Technology, Nobelya Str., 3, Moscow, Russia, 143026; Institute of Energy Problems of Chemical Physics, Leninskiy Prospect, 38/2, Moscow, Russia, 119334
| |
Collapse
|
24
|
Guala D, Bernhem K, Blal HA, Jans D, Lundberg E, Brismar H, Sonnhammer ELL. Experimental validation of predicted cancer genes using FRET. Methods Appl Fluoresc 2018; 6:035007. [PMID: 29570091 DOI: 10.1088/2050-6120/aab932] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Huge amounts of data are generated in genome wide experiments, designed to investigate diseases with complex genetic causes. Follow up of all potential leads produced by such experiments is currently cost prohibitive and time consuming. Gene prioritization tools alleviate these constraints by directing further experimental efforts towards the most promising candidate targets. Recently a gene prioritization tool called MaxLink was shown to outperform other widely used state-of-the-art prioritization tools in a large scale in silico benchmark. An experimental validation of predictions made by MaxLink has however been lacking. In this study we used Fluorescence Resonance Energy Transfer, an established experimental technique for detection of protein-protein interactions, to validate potential cancer genes predicted by MaxLink. Our results provide confidence in the use of MaxLink for selection of new targets in the battle with polygenic diseases.
Collapse
Affiliation(s)
- Dimitri Guala
- Science for Life Laboratory, Stockholm Bioinformatics Center, Department of Biochemistry and Biophysics, Stockholm University, Box 1031, 17121 Solna, Sweden
| | | | | | | | | | | | | |
Collapse
|
25
|
Wang Q, Xu H, Zhao X. Baicalin Inhibits Human Cervical Cancer Cells by Suppressing Protein Kinase C/Signal Transducer and Activator of Transcription (PKC/STAT3) Signaling Pathway. Med Sci Monit 2018; 24:1955-1961. [PMID: 29610452 PMCID: PMC5896362 DOI: 10.12659/msm.909640] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Like other human cancers, the malignancy of cervical cancer is also characterized by abilities of proliferation, migration, and invasion. Protein kinase C-zeta (PKCζ) has been highly correlated with several human cancers. Baicalin was proven to regulate PKC. This study aimed to investigate the anti-cancer effect and involved molecular mechanisms of baicalin on human cervical cancer. MATERIAL AND METHODS Baicalin at various concentrations was used to treat 2 human cervical cancer cell lines HeLa and SiHa. The proliferation was assessed by 3-(4,5-dimethylthiazol-2-yl)-2,5 diphenylterazolium bromide (MTT) assay. The apoptosis was detected by terminal transferase UTP nick end labeling (TUNEL) assay. Wound healing assay and Transwell assay were used to evaluate the migration and invasion respectively. Western blotting was performed to assess the protein expression levels. RESULTS Baicalin administration significantly reduced the viability by facilitating the apoptosis in HeLa and SiHa cells. Baicalin treatment also significantly reduced the wound closure and cell amount invaded as measured by Transwell assay. The expression levels of PKCζ, survivin, matrix metalloproteinase (MMP)2, MMP9 as well as the phosphorylation of signal transducer and activator of transcription (STAT) 3 were reduced in baicalin administrated cervical cancer cells. CONCLUSIONS Baicalin exerted anti-cancer effects on human cervical cancer cells by targeting STAT3 regulated signaling pathways.
Collapse
Affiliation(s)
- Qianqian Wang
- Department of Gynecology, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, China (mainland).,People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China (mainland)
| | - Haiou Xu
- Department of Gynecology, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, China (mainland).,People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China (mainland)
| | - Xiaofeng Zhao
- Department of Gynecology, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, China (mainland).,People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China (mainland)
| |
Collapse
|
26
|
Kim KS, Kim J, Oh N, Kim MY, Park KS. ELK3-GATA3 axis modulates MDA-MB-231 metastasis by regulating cell-cell adhesion-related genes. Biochem Biophys Res Commun 2018; 498:509-515. [PMID: 29510139 DOI: 10.1016/j.bbrc.2018.03.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 03/02/2018] [Indexed: 10/25/2022]
Abstract
GATA3 is a master regulator that drives mammary epithelial cell differentiation, and the suppression of GATA3 expression is associated with the development of aggressive breast cancer. However, the mechanism through which GATA3 loss drives cancer development is poorly understood. Previously, we reported that ELK3 suppression in MDA-MB-231 (ELK3 KD) resulted in the reprogramming of these cells from a basal to luminal subtype, which was associated with the induction of GATA3 expression, and that the ELK3-GATA3 axis orchestrated the metastatic characteristics of MDA-MB-231. Here, we show that GATA3 suppression in ELK3 knockdown MDA-MB-231 cells (ELK3/GATA3 DKD) restores the metastatic ability comparably to that of control MDA-MB-231 cells, even though the epithelial cell morphology and TGF-β signaling of ELK3 KD are not recovered in ELK3/GATA3 DKD. The expression of E-cadherin and tight junctional proteins, including occludin, claudin and ZO-1, which is activated in ELK3 KD, is suppressed in ELK3/GATA3 DKD. These results reveal the possibility that the ELK3-GATA3 axis determines the metastatic characteristics of MDA-MB-231 by regulating the expression of cell-cell adhesion factors.
Collapse
Affiliation(s)
- Kwang-Soo Kim
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, South Korea
| | - Jiewan Kim
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, South Korea
| | - Nuri Oh
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, South Korea
| | - Mi-Young Kim
- Digestive Disease Center, CHA Bundang Medical Center, CHA University, Seongnam, South Korea.
| | - Kyung-Soon Park
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, South Korea.
| |
Collapse
|
27
|
Lan T, Hung SH, Su X, Wong SWK, Tseng Y. Integrating transient cellular and nuclear motions to comprehensively describe cell migration patterns. Sci Rep 2018; 8:1488. [PMID: 29367613 PMCID: PMC5784082 DOI: 10.1038/s41598-018-19885-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 01/09/2018] [Indexed: 02/07/2023] Open
Abstract
Various subcellular activities, such as protrusion and detachment, compose a cell migration process. The molecular mechanisms of these subcellular activities have been elucidated. However, there is no method that can assess the contributions of these subcellular activities to the global cell migration pattern of a given cell type. Hence, we develop a powerful approach based on CN correlations that quantitatively profiles the cell migration pattern of a given cell type in terms of assembled subcellular activities. In this way, we bridge migration data at the cellular level with underlying molecular mechanisms. The CN correlation profile is found to uniquely and consistently represent the cell migration pattern of each cell type probed. It can clearly reveal the effects of molecular perturbations, such as Y27632 and Cdc42 knockdown on each subcellular migratory activity. As a result, the CN correlation approach serves as a cell dynamic descriptor that can extract comprehensive quantitative data from cell migration movies for integrative biological analyses.
Collapse
Affiliation(s)
- Tian Lan
- Department of Chemical Engineering, University of Florida, Gainesville, FL, 32611, USA
| | - Shen-Hsiu Hung
- Department of Chemical Engineering, University of Florida, Gainesville, FL, 32611, USA
| | - Xudong Su
- Department of Chemical Engineering, University of Florida, Gainesville, FL, 32611, USA
| | - Samuel W K Wong
- Department of Statistics, University of Florida, Gainesville, FL, 32611, USA
| | - Yiider Tseng
- Department of Chemical Engineering, University of Florida, Gainesville, FL, 32611, USA. .,J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, 32611, USA. .,Institute for Cell & Tissue Science and Engineering, University of Florida, Gainesville, FL, 32611, USA. .,National Cancer Institute-Physical Science Oncology Center, Gainesville, FL, 32611, USA.
| |
Collapse
|
28
|
Cai X, Zhu H, Li Y. PKCζ, MMP‑2 and MMP‑9 expression in lung adenocarcinoma and association with a metastatic phenotype. Mol Med Rep 2017; 16:8301-8306. [PMID: 28983601 DOI: 10.3892/mmr.2017.7634] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 08/01/2017] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to investigate protein kinase C ζ type (PKCζ), matrix metalloproteinase (MMP)‑2 and MMP‑9 expression in lung adenocarcinoma and to define their association with in vitro invasion and metastatic capacity. PKCζ, MMP‑2 and MMP‑9 expression was assessed by immunohistochemistry in 110 cases of lung adenocarcinoma. PKCζ small interfering (si)RNA was transfected into A549 cells, and western blotting was used to confirm PKCζ‑knockdown in transfected cells and to measure MMP‑2 and MMP‑9 levels. A Transwell invasion assay was used to detect in vitro invasive capacity. The rates of positive PKCζ, MMP‑2 and MMP‑9 staining in lung adenocarcinoma tissues were 52.73, 55.45 and 61.82%, respectively. PKCζ expression was increased in malignant tissues compared with adjacent normal lung tissues and was associated with lymph node metastasis (P<0.05), although it was not associated with any other clinicopathological parameters, including sex, age, tumor size, smoking status or distant metastases (all P>0.05). PKCζ, MMP‑2 and MMP‑9 expression was markedly decreased in siPKCζ‑treated A549 cells, which exhibited a significantly decreased invasive capacity in the Transwell invasion assay (P<0.05). In conclusion, PKCζ promoted lung adenocarcinoma invasion and metastasis, and its expression was associated with MMP‑2 and MMP‑9 expression. PKCζ may be a potential target for gene therapy in lung adenocarcinoma.
Collapse
Affiliation(s)
- Xiaoshan Cai
- Department of Pathology, Second People's Hospital of Weifang, Weifang, Shandong 261041, P.R. China
| | - Hongguang Zhu
- Department of Dentistry, Weifang People's Hospital, Weifang, Shandong 261000, P.R. China
| | - Ying Li
- Department of Pathology, Second People's Hospital of Weifang, Weifang, Shandong 261041, P.R. China
| |
Collapse
|
29
|
Retinoic acid directs breast cancer cell state changes through regulation of TET2-PKCζ pathway. Oncogene 2017; 36:3193-3206. [PMID: 28218902 PMCID: PMC5541263 DOI: 10.1038/onc.2016.467] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 10/24/2016] [Accepted: 11/08/2016] [Indexed: 01/05/2023]
Abstract
The key molecular mechanism governing the cancer cell state (stem cell-like state vs differentiation state) to control the cancer stem cell (CSC) pool remains elusive. This study provides the first evidence showing that all-trans retinoic acid (ATRA) induces the interaction and chromatin recruitment of a novel RARβ-TET2 complex to epigenetically activate a specific cohort of gene targets, including MiR-200c. TET2-activated miR-200c further targets and suppresses PKCζ, a cell polarity protein that has a pivotal role in directing asymmetric division of mammalian stem cells to sustain the stem cell pool. Our data reveal that pharmacological concentration of ATRA effectively downregulates PKCζ through activation of miR-200c, leading to a decrease of the stem cell-like populations from non-tumorigenic mammary epithelial cells and non-aggressive breast cancer cells. However, aggressive breast cancer cells that manifest TET2-miR-200c dysregulation sustain a CSC pool highly resistant to ATRA, where inhibition of PKCζ directs the resistant CSCs to the luminal cell-like state and sensitization to tamoxifen, resulting in abrogation of mammary tumor growth and progression. Together, these findings elucidate a novel RARβ-TET2-miR-200c-PKCζ signaling pathway that directs cancer cell state changes and also provide previously unidentified therapeutic implications for PKCζ inhibitors in diminishment of breast CSCs to eradicate breast cancer.
Collapse
|
30
|
Zhang H, Chen X, Li D, Cui L, Li X, Ye X, Wan X. DcR3 promotes hepatoma cell migration by downregulating E-cadherin expression. Oncol Rep 2017; 38:377-383. [PMID: 28560426 DOI: 10.3892/or.2017.5685] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 12/27/2016] [Indexed: 11/06/2022] Open
Abstract
Decoy receptor 3 (DcR3), a decoy molecule belonging to the tumor necrosis factor receptor superfamily (TNFRSF), is a soluble receptor that can neutralize the biological effects of three other TNFSF members, namely, Fas ligand (FasL/TNFSF6/CD95L), LIGHT (TNFSF14) and TNF-like molecule 1A (TL1A/TNFSF15). DcR3 expression is increased in tumor cells. As such, DcR3 has been considered a potential biomarker to predict cancer invasion and progression of inflammation. However, the molecular mechanisms of DcR3 in tumor progression and metastasis remain poorly described. In the present study, DcR3 induced cytoskeleton remodeling, inhibited E-cadherin expression, and promoted cancer cell migration. Immunofluorescence and flow cytometry demonstrated that DcR3 expression was increased in hepatoma cells, whereas E-cadherin expression was significantly downregulated. Immunohistochemistry revealed that DcR3 and E-cadherin exhibited an opposite expression pattern between normal and cancerous liver tissues. Moreover, DcR3 treatment promoted IκBα degradation and p65 nuclear translocation. Therefore, the present study uncovered the mechanism underlying the function of DcR3 in cancer cell migration and provides evidence that DcR3 may be a potential target for cancer therapy.
Collapse
Affiliation(s)
- Hongling Zhang
- Shenzhen Laboratory of Fully Human Antibody Engineering, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, P.R. China
| | - Xuhong Chen
- Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen 518035, P.R. China
| | - Dongming Li
- Basic Medicine College, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Lulu Cui
- Shenzhen Laboratory of Fully Human Antibody Engineering, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, P.R. China
| | - Xin Li
- Shenzhen Laboratory of Fully Human Antibody Engineering, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, P.R. China
| | - Xiufeng Ye
- Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen 518035, P.R. China
| | - Xiaochun Wan
- Shenzhen Laboratory of Fully Human Antibody Engineering, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, P.R. China
| |
Collapse
|
31
|
González-Tarragó V, Elosegui-Artola A, Bazellières E, Oria R, Pérez-González C, Roca-Cusachs P. Binding of ZO-1 to α5β1 integrins regulates the mechanical properties of α5β1-fibronectin links. Mol Biol Cell 2017; 28:1847-1852. [PMID: 28251923 PMCID: PMC5541835 DOI: 10.1091/mbc.e17-01-0006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 02/23/2017] [Accepted: 02/24/2017] [Indexed: 01/06/2023] Open
Abstract
Fundamental processes in cell adhesion, motility, and rigidity adaptation are regulated by integrin-mediated adhesion to the extracellular matrix (ECM). The link between the ECM component fibronectin (fn) and integrin α5β1 forms a complex with ZO-1 in cells at the edge of migrating monolayers, regulating cell migration. However, how this complex affects the α5β1-fn link is unknown. Here we show that the α5β1/ZO-1 complex decreases the resistance to force of α5β1-fn adhesions located at the edge of migrating cell monolayers while also increasing α5β1 recruitment. Consistently with a molecular clutch model of adhesion, this effect of ZO-1 leads to a decrease in the density and intensity of adhesions in cells at the edge of migrating monolayers. Taken together, our results unveil a new mode of integrin regulation through modification of the mechanical properties of integrin-ECM links, which may be harnessed by cells to control adhesion and migration.
Collapse
Affiliation(s)
- Víctor González-Tarragó
- Institute for Bioengineering of Catalonia, 08028 Barcelona, Spain.,University of Barcelona, 08028 Barcelona, Spain
| | | | - Elsa Bazellières
- IBDM, Institut de Biologie du Développement de Marseille, UMR 7288, 13009 Marseille, France
| | - Roger Oria
- Institute for Bioengineering of Catalonia, 08028 Barcelona, Spain.,University of Barcelona, 08028 Barcelona, Spain
| | - Carlos Pérez-González
- Institute for Bioengineering of Catalonia, 08028 Barcelona, Spain.,University of Barcelona, 08028 Barcelona, Spain
| | - Pere Roca-Cusachs
- Institute for Bioengineering of Catalonia, 08028 Barcelona, Spain .,University of Barcelona, 08028 Barcelona, Spain
| |
Collapse
|
32
|
Kobayashi K, Kuki C, Oyama S, Kumura H. Pro-inflammatory cytokine TNF-α is a key inhibitory factor for lactose synthesis pathway in lactating mammary epithelial cells. Exp Cell Res 2016; 340:295-304. [DOI: 10.1016/j.yexcr.2015.10.030] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 10/23/2015] [Accepted: 10/24/2015] [Indexed: 10/24/2022]
|