1
|
Rembiałkowska N, Kucharczyk J, Radzevičiūtė-Valčiukė E, Novickij V, Tonci M, Dündar A, Kulbacka J, Szlasa W. Enhancing lung cancer growth inhibition with calcium ions: Role of mid- and high-frequency electric field pulses. Biomed Pharmacother 2024; 181:117691. [PMID: 39557010 DOI: 10.1016/j.biopha.2024.117691] [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: 08/08/2024] [Revised: 11/09/2024] [Accepted: 11/12/2024] [Indexed: 11/20/2024] Open
Abstract
Calcium electroporation (CaEP) involves the combination of calcium ions with electroporation, which is induced by pulsed electric fields (PEFs). This study explores the application of high-frequency unipolar nanosecond pulsed electric fields (nsPEFs: 8-14 kV/cm, 200 ns, 10 kHz, 100 kHz, 1 MHz repetition frequency pulse bursts, n = 100) and their potential in inhibiting lung cancer cell growth. As a reference, standard microsecond range parametric protocols were used (100 µs x 8 pulses). Methods included cell permeability quantification through Yo-Pro-1 uptake, cell viability assays, immunofluorescence studies for apoptosis and EMT markers, analysis of cell death types depending on repetition frequency pulse bursts. We determined the susceptibility of human lung cancer to electric pulses, characterized the efficacy of CaEP, and investigated cell death types depending on repetition frequency pulse bursts. We have shown that adding calcium ions to the applied nsPEF protocol increases cytotoxicity. Additionally, the use of these electroporation parameters can modulate key cellular processes, such as the epithelial-mesenchymal transition and apoptosis, as indicated by changes in the expression of markers such as E-cadherin, N-cadherin, BCL-2, and p53. Changes in cell morphology over time were observed using holotomographic microscopy. The study provides insights into the modulation of key cellular processes, indicating that nsPEF technology could improve the outcomes of conventional cancer treatments through enhanced efficacy and potentially mitigating drug resistance mechanisms. The promising results advocate for further research to optimize nsPEF protocols for clinical application, highlighting the potential of electrical fields in advancing cancer therapy.
Collapse
Affiliation(s)
- Nina Rembiałkowska
- Wroclaw Medical University, Faculty of Pharmacy, Department of Molecular and Cellular Biology, Wroclaw, Poland.
| | - Julia Kucharczyk
- The Students' Research Group, UMW, SKN No. 148, Wroclaw Medical University, Faculty of Pharmacy, Department of Molecular and Cellular Biology, Wroclaw, Poland.
| | - Eivina Radzevičiūtė-Valčiukė
- Vilnius Gediminas Technical University, Faculty of Electronics, Vilnius, Lithuania; State Research Institute Centre for Innovative Medicine, Department of Immunology and Bioelectrochemistry, Vilnius, Lithuania.
| | - Vitalij Novickij
- Vilnius Gediminas Technical University, Faculty of Electronics, Vilnius, Lithuania; State Research Institute Centre for Innovative Medicine, Department of Immunology and Bioelectrochemistry, Vilnius, Lithuania.
| | - Margherita Tonci
- Faculty of Medicine and Surgery, University of Pisa, Pisa, Italy.
| | - Ata Dündar
- Faculty of Medicine, Istanbul University Cerrahpaşa, Istanbul, Turkey.
| | - Julita Kulbacka
- Wroclaw Medical University, Faculty of Pharmacy, Department of Molecular and Cellular Biology, Wroclaw, Poland; State Research Institute Centre for Innovative Medicine, Department of Immunology and Bioelectrochemistry, Vilnius, Lithuania.
| | - Wojciech Szlasa
- Wroclaw Medical University, Faculty of Pharmacy, Department of Molecular and Cellular Biology, Wroclaw, Poland; Medical University Hospital, Wroclaw, Poland.
| |
Collapse
|
2
|
Zhang Z, Westover D, Tang Z, Liu Y, Sun J, Sun Y, Zhang R, Wang X, Zhou S, Hesilaiti N, Xia Q, Du Z. Wnt/β-catenin signaling in the development and therapeutic resistance of non-small cell lung cancer. J Transl Med 2024; 22:565. [PMID: 38872189 PMCID: PMC11170811 DOI: 10.1186/s12967-024-05380-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 06/06/2024] [Indexed: 06/15/2024] Open
Abstract
Wnt/β-catenin signaling is a critical pathway that influences development and therapeutic response of non-small cell lung cancer (NSCLC). In recent years, many Wnt regulators, including proteins, miRNAs, lncRNAs, and circRNAs, have been found to promote or inhibit signaling by acting on Wnt proteins, receptors, signal transducers and transcriptional effectors. The identification of these regulators and their underlying molecular mechanisms provides important implications for how to target this pathway therapeutically. In this review, we summarize recent studies of Wnt regulators in the development and therapeutic response of NSCLC.
Collapse
Affiliation(s)
- Zixu Zhang
- Department of Genetic and Developmental Biology, School of Medicine, Southeast University, Nanjing, 210003, China
| | - David Westover
- High-Throughput Analytics, Analytical Research and Development, Merck & Co. Inc., Rahway, NJ, USA
| | - Zhantong Tang
- Department of Genetic and Developmental Biology, School of Medicine, Southeast University, Nanjing, 210003, China
| | - Yue Liu
- Department of Genetic and Developmental Biology, School of Medicine, Southeast University, Nanjing, 210003, China
| | - Jinghan Sun
- School of Life Science and Technology, Southeast University, Nanjing, 210018, China
| | - Yunxi Sun
- Department of Genetic and Developmental Biology, School of Medicine, Southeast University, Nanjing, 210003, China
| | - Runqing Zhang
- Department of Genetic and Developmental Biology, School of Medicine, Southeast University, Nanjing, 210003, China
| | - Xingyue Wang
- Department of Genetic and Developmental Biology, School of Medicine, Southeast University, Nanjing, 210003, China
| | - Shihui Zhou
- Department of Genetic and Developmental Biology, School of Medicine, Southeast University, Nanjing, 210003, China
| | - Nigaerayi Hesilaiti
- Department of Genetic and Developmental Biology, School of Medicine, Southeast University, Nanjing, 210003, China
| | - Qi Xia
- Department of Genetic and Developmental Biology, School of Medicine, Southeast University, Nanjing, 210003, China
| | - Zhenfang Du
- Department of Genetic and Developmental Biology, School of Medicine, Southeast University, Nanjing, 210003, China.
| |
Collapse
|
3
|
Ghosh M, Khanam R, Sengupta A, Chakraborty S. Oxidative-stress induced Bmp2-Smad1/5/8 signaling dependent differentiation of early cardiomyocytes from embryonic and adult epicardial cells. Differentiation 2024; 136:100756. [PMID: 38471281 DOI: 10.1016/j.diff.2024.100756] [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: 06/19/2023] [Revised: 02/15/2024] [Accepted: 02/17/2024] [Indexed: 03/14/2024]
Abstract
Heart failure has become a major life-threatening cause affecting millions globally, characterized by the permanent loss of adult functional cardiomyocytes leading to fibrosis which ultimately deprives the heart of its functional efficacy. Here we investigated the reparative property of embryonic and adult epicardial cells towards cardiomyocyte differentiation under oxidative stress-induced conditions along with the identification of a possible molecular signaling pathway. Isolated epicardial cells from embryonic chick hearts subjected to oxidative stress and hypoxia induction. Initial assessment of successful injury induction reveals hypertrophy of isolated epicardial cells. Detailed marker gene expression analyses and inhibitor studies reveal Bone morphogenic protein (Bmp)2-Smad1/5/8 signaling dependent cardiomyocyte lineage specification via epithelial to mesenchymal transition (EMT) post-injury. EMT is further confirmed by increased proliferation, migration, and differentiation towards cardiomyocyte lineage. We have also established an in-vivo model in adult male rats using Isoproterenol. Successful oxidative stress-mediated injury induction in adult heart was marked by increased activated fibroblasts followed by apoptosis of adult cardiomyocytes. The detailed characterization of adult epicardial cells reveals similar findings to our avian in-vitro data. Both in-vitro and in-vivo results show a significant increase in the expression of cardiomyocyte specific markers indicative of lineage specificity and activation of epicardial cells post oxidative stress mediated injury. Our findings suggest an EMT-induced reactivation of epicardial cells and early cardiomyocyte lineage specification following oxidative stress in a Bmp2- Smad1/5/8 dependent manner. Overall, this regulatory mechanism of cardiomyocyte differentiation induced by oxidative stress may contribute to the field of cardiac repair and regenerative therapeutics.
Collapse
Affiliation(s)
- Madhurima Ghosh
- Department of Life Sciences, Presidency University, Kolkata, 700073, India
| | - Riffat Khanam
- Department of Life Sciences, Presidency University, Kolkata, 700073, India
| | - Arunima Sengupta
- Department of Life Science and Biotechnology, Jadavpur University, Kolkata, 700032, India
| | | |
Collapse
|
4
|
Bartolomé RA, Pintado-Berninches L, Martín-Regalado Á, Robles J, Calvo-López T, Ortega-Zapero M, Llorente-Sáez C, Boukich I, Fernandez-Aceñero MJ, Casal JI. A complex of cadherin 17 with desmocollin 1 and p120-catenin regulates colorectal cancer migration and invasion according to the cell phenotype. J Exp Clin Cancer Res 2024; 43:31. [PMID: 38263178 PMCID: PMC10807196 DOI: 10.1186/s13046-024-02956-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 01/14/2024] [Indexed: 01/25/2024] Open
Abstract
BACKGROUND Cadherin-17 (CDH17), a marker of differentiation in intestinal cells, binds and activates α2β1 integrin to promote cell adhesion and proliferation in colorectal cancer (CRC) metastasis. Furthermore, CDH17 associates with p120- and β-catenin in a manner yet to be fully elucidated. In this report, we explored the molecular mediators involved in this association, their contribution to CRC dissemination and potential therapeutic implications. METHODS Proteomic and confocal analyses were employed to identify and validate CDH17 interactors. Functional characterization involved the study of proliferation, migration, and invasion in cell lines representative of various phenotypes. Immunohistochemistry was conducted on CRC tissue microarrays (TMA). In vivo animal experiments were carried out for metastatic studies. RESULTS We found that desmocollin-1 (DSC1), a desmosomal cadherin, interacts with CDH17 via its extracellular domain. DSC1 depletion led to increased or decreased invasion in CRC cells displaying epithelial or mesenchymal phenotype, respectively, in a process mediated by the association with p120-catenin. Down-regulation of DSC1 resulted in an increased expression of p120-catenin isoform 1 in epithelial cells or a shift in cellular location in mesenchymal cells. Opposite results were observed after forced expression of CDH17. DSC1 is highly expressed in budding cells at the leading edge of the tumor and associates with poor prognosis in the stem-like, mesenchymal CRC subtypes, while correlates with a more favorable prognosis in the less-aggressive subtypes. In vivo experiments demonstrated that DSC1 silencing reduced tumor growth, liver homing, and metastasis in CRC mesenchymal cells. Furthermore, a synthetic peptide derived from CDH17, containing the NLV motif, effectively inhibited invasion and liver homing in vivo, opening up new possibilities for the development of novel therapies focused on desmosomal cadherins. CONCLUSIONS These findings shed light on the multifaceted roles of CDH17, DSC1, and p120-catenin in CRC metastasis, offering insights into potential therapeutic interventions for targeting desmosomal cadherins in poorly-differentiated carcinomas.
Collapse
Affiliation(s)
- Rubén A Bartolomé
- Department of Biomolecular Medicine, Centro de Investigaciones Biológicas Margarita Salas (CSIC), Ramiro de Maeztu 9, Madrid, 28040, Spain.
| | | | - Ángela Martín-Regalado
- Department of Biomolecular Medicine, Centro de Investigaciones Biológicas Margarita Salas (CSIC), Ramiro de Maeztu 9, Madrid, 28040, Spain
| | - Javier Robles
- Department of Biomolecular Medicine, Centro de Investigaciones Biológicas Margarita Salas (CSIC), Ramiro de Maeztu 9, Madrid, 28040, Spain
- Protein Alternatives SL. Tres Cantos, Madrid, Spain
| | - Tania Calvo-López
- Department of Biomolecular Medicine, Centro de Investigaciones Biológicas Margarita Salas (CSIC), Ramiro de Maeztu 9, Madrid, 28040, Spain
| | - Marina Ortega-Zapero
- Department of Biomolecular Medicine, Centro de Investigaciones Biológicas Margarita Salas (CSIC), Ramiro de Maeztu 9, Madrid, 28040, Spain
| | - Celia Llorente-Sáez
- Department of Biomolecular Medicine, Centro de Investigaciones Biológicas Margarita Salas (CSIC), Ramiro de Maeztu 9, Madrid, 28040, Spain
- Present address: Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain
| | - Issam Boukich
- Department of Biomolecular Medicine, Centro de Investigaciones Biológicas Margarita Salas (CSIC), Ramiro de Maeztu 9, Madrid, 28040, Spain
- Protein Alternatives SL. Tres Cantos, Madrid, Spain
| | - María Jesús Fernandez-Aceñero
- Pathology Service. Hospital Clínico San Carlos, Madrid, Spain
- Fundación de Investigación Biomédica del HCSC (FIBHCSC), Madrid, Spain
| | - J Ignacio Casal
- Department of Biomolecular Medicine, Centro de Investigaciones Biológicas Margarita Salas (CSIC), Ramiro de Maeztu 9, Madrid, 28040, Spain.
| |
Collapse
|
5
|
Gisina A, Kim Y, Yarygin K, Lupatov A. Can CD133 Be Regarded as a Prognostic Biomarker in Oncology: Pros and Cons. Int J Mol Sci 2023; 24:17398. [PMID: 38139228 PMCID: PMC10744290 DOI: 10.3390/ijms242417398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/07/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023] Open
Abstract
The CD133 cell membrane glycoprotein, also termed prominin-1, is expressed on some of the tumor cells of both solid and blood malignancies. The CD133-positive tumor cells were shown to exhibit higher proliferative activity, greater chemo- and radioresistance, and enhanced tumorigenicity compared to their CD133-negative counterparts. For this reason, CD133 is regarded as a potential prognostic biomarker in oncology. The CD133-positive cells are related to the cancer stem cell subpopulation in many types of cancer. Recent studies demonstrated the involvement of CD133 in the regulation of proliferation, autophagy, and apoptosis in cancer cells. There is also evidence of its participation in the epithelial-mesenchymal transition associated with tumor progression. For a number of malignant tumor types, high CD133 expression is associated with poor prognosis, and the prognostic significance of CD133 has been confirmed in a number of meta-analyses. However, some published papers suggest that CD133 has no prognostic significance or even demonstrate a certain correlation between high CD133 levels and a positive prognosis. This review summarizes and discusses the existing evidence for and against the prognostic significance of CD133 in cancer. We also consider possible reasons for conflicting findings from the studies of the clinical significance of CD133.
Collapse
Affiliation(s)
- Alisa Gisina
- Laboratory of Cell Biology, V. N. Orekhovich Institute of Biomedical Chemistry, 119121 Moscow, Russia
| | | | | | | |
Collapse
|
6
|
Liu Y, Lei P, Samuel RZ, Kashyap AM, Groth T, Bshara W, Neelamegham S, Andreadis ST. Cadherin-11 increases tumor cell proliferation and metastatic potential via Wnt pathway activation. Mol Oncol 2023; 17:2056-2073. [PMID: 37558205 PMCID: PMC10552893 DOI: 10.1002/1878-0261.13507] [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: 09/16/2022] [Revised: 06/23/2023] [Accepted: 07/08/2023] [Indexed: 08/11/2023] Open
Abstract
During epithelial-mesenchymal transition (EMT) in cancer progression, tumor cells switch cadherin profile from E-cadherin to cadherin-11 (CDH11), which is accompanied by increased invasiveness and metastatic activity. However, the mechanism through which CDH11 may affect tumor growth and metastasis remains elusive. Here, we report that CDH11 was highly expressed in multiple human tumors and was localized on the membrane, in the cytoplasm and, surprisingly, also in the nucleus. Interestingly, β-catenin remained bound to carboxy-terminal fragments (CTFs) of CDH11, the products of CDH11 cleavage, and co-localized with CTFs in the nucleus in the majority of breast cancer samples. Binding of β-catenin to CTFs preserved β-catenin activity, whereas inhibiting CDH11 cleavage led to β-catenin phosphorylation and diminished Wnt signaling, similar to CDH11 knockout. Our data elucidate a previously unknown role of CDH11, which serves to stabilize β-catenin in the cytoplasm and facilitates its translocation to the nucleus, resulting in activation of Wnt signaling, with subsequent increased proliferation, migration and invasion potential.
Collapse
Affiliation(s)
- Yayu Liu
- Department of Chemical and Biological Engineering, University at BuffaloThe State University of New YorkAmherstNYUSA
| | - Pedro Lei
- Department of Chemical and Biological Engineering, University at BuffaloThe State University of New YorkAmherstNYUSA
| | - Ronel Z. Samuel
- Department of Chemical and Biological Engineering, University at BuffaloThe State University of New YorkAmherstNYUSA
| | - Anagha M. Kashyap
- Department of Chemical and Biological Engineering, University at BuffaloThe State University of New YorkAmherstNYUSA
| | - Theodore Groth
- Department of Chemical and Biological Engineering, University at BuffaloThe State University of New YorkAmherstNYUSA
| | - Wiam Bshara
- Roswell Park Comprehensive Cancer Center Pathology Resource NetworkBuffaloNYUSA
| | - Sriram Neelamegham
- Department of Chemical and Biological Engineering, University at BuffaloThe State University of New YorkAmherstNYUSA
- Department of Biomedical Engineering, University at BuffaloThe State University of New YorkAmherstNYUSA
- New York State Center of Excellence in Bioinformatics and Life SciencesBuffaloNYUSA
- Center for Cell, Gene and Tissue Engineering (CGTE), University at BuffaloThe State University of New YorkAmherstNYUSA
| | - Stelios T. Andreadis
- Department of Chemical and Biological Engineering, University at BuffaloThe State University of New YorkAmherstNYUSA
- Department of Biomedical Engineering, University at BuffaloThe State University of New YorkAmherstNYUSA
- New York State Center of Excellence in Bioinformatics and Life SciencesBuffaloNYUSA
- Center for Cell, Gene and Tissue Engineering (CGTE), University at BuffaloThe State University of New YorkAmherstNYUSA
| |
Collapse
|
7
|
Yehya A, Youssef J, Hachem S, Ismael J, Abou-Kheir W. Tissue-specific cancer stem/progenitor cells: Therapeutic implications. World J Stem Cells 2023; 15:323-341. [PMID: 37342220 PMCID: PMC10277968 DOI: 10.4252/wjsc.v15.i5.323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/14/2023] [Accepted: 04/12/2023] [Indexed: 05/26/2023] Open
Abstract
Surgical resection, chemotherapy, and radiation are the standard therapeutic modalities for treating cancer. These approaches are intended to target the more mature and rapidly dividing cancer cells. However, they spare the relatively quiescent and intrinsically resistant cancer stem cells (CSCs) subpopulation residing within the tumor tissue. Thus, a temporary eradication is achieved and the tumor bulk tends to revert supported by CSCs' resistant features. Based on their unique expression profile, the identification, isolation, and selective targeting of CSCs hold great promise for challenging treatment failure and reducing the risk of cancer recurrence. Yet, targeting CSCs is limited mainly by the irrelevance of the utilized cancer models. A new era of targeted and personalized anti-cancer therapies has been developed with cancer patient-derived organoids (PDOs) as a tool for establishing pre-clinical tumor models. Herein, we discuss the updated and presently available tissue-specific CSC markers in five highly occurring solid tumors. Additionally, we highlight the advantage and relevance of the three-dimensional PDOs culture model as a platform for modeling cancer, evaluating the efficacy of CSC-based therapeutics, and predicting drug response in cancer patients.
Collapse
Affiliation(s)
- Amani Yehya
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Joe Youssef
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Sana Hachem
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Jana Ismael
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Wassim Abou-Kheir
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut 1107-2020, Lebanon.
| |
Collapse
|
8
|
Haage A, Dhasarathy A. Working a second job: Cell adhesion proteins that moonlight in the nucleus. Front Cell Dev Biol 2023; 11:1163553. [PMID: 37169022 PMCID: PMC10164977 DOI: 10.3389/fcell.2023.1163553] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 04/12/2023] [Indexed: 05/13/2023] Open
Abstract
Cells are adept at sensing changes in their environment, transmitting signals internally to coordinate responses to external stimuli, and thereby influencing adaptive changes in cell states and behavior. Often, this response involves modulation of gene expression in the nucleus, which is seen largely as a physically separated process from the rest of the cell. Mechanosensing, whereby a cell senses physical stimuli, and integrates and converts these inputs into downstream responses including signaling cascades and gene regulatory changes, involves the participation of several macromolecular structures. Of note, the extracellular matrix (ECM) and its constituent macromolecules comprise an essential part of the cellular microenvironment, allowing cells to interact with each other, and providing both structural and biochemical stimuli sensed by adhesion transmembrane receptors. This highway of information between the ECM, cell adhesion proteins, and the cytoskeleton regulates cellular behavior, the disruption of which results in disease. Emerging evidence suggests a more direct role for some of these adhesion proteins in chromatin structure and gene regulation, RNA maturation and other non-canonical functions. While many of these discoveries were previously limited to observations of cytoplasmic-nuclear transport, recent advances in microscopy, and biochemical, proteomic and genomic technologies have begun to significantly enhance our understanding of the impact of nuclear localization of these proteins. This review will briefly cover known cell adhesion proteins that migrate to the nucleus, and their downstream functions. We will outline recent advances in this very exciting yet still emerging field, with impact ranging from basic biology to disease states like cancer.
Collapse
Affiliation(s)
- Amanda Haage
- *Correspondence: Amanda Haage, ; Archana Dhasarathy,
| | | |
Collapse
|
9
|
Eguchi T, Csizmadia E, Kawai H, Sheta M, Yoshida K, Prince TL, Wegiel B, Calderwood SK. SCAND1 Reverses Epithelial-to-Mesenchymal Transition (EMT) and Suppresses Prostate Cancer Growth and Migration. Cells 2022; 11:cells11243993. [PMID: 36552758 PMCID: PMC9777339 DOI: 10.3390/cells11243993] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 12/08/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a reversible cellular program that transiently places epithelial (E) cells into pseudo-mesenchymal (M) cell states. The malignant progression and resistance of many carcinomas depend on EMT activation, partial EMT, or hybrid E/M status in neoplastic cells. EMT is activated by tumor microenvironmental TGFβ signal and EMT-inducing transcription factors, such as ZEB1/2, in tumor cells. However, reverse EMT factors are less studied. We demonstrate that prostate epithelial transcription factor SCAND1 can reverse the cancer cell mesenchymal and hybrid E/M phenotypes to a more epithelial, less invasive status and inhibit their proliferation and migration in DU-145 prostate cancer cells. SCAND1 is a SCAN domain-containing protein and hetero-oligomerizes with SCAN-zinc finger transcription factors, such as MZF1, for accessing DNA and the transcriptional co-repression of target genes. We found that SCAND1 expression correlated with maintaining epithelial features, whereas the loss of SCAND1 was associated with mesenchymal phenotypes of tumor cells. SCAND1 and MZF1 were mutually inducible and coordinately included in chromatin with hetero-chromatin protein HP1γ. The overexpression of SCAND1 reversed hybrid E/M status into an epithelial phenotype with E-cadherin and β-catenin relocation. Consistently, the co-expression analysis in TCGA PanCancer Atlas revealed that SCAND1 and MZF1 expression was negatively correlated with EMT driver genes, including CTNNB1, ZEB1, ZEB2 and TGFBRs, in prostate adenocarcinoma specimens. In addition, SCAND1 overexpression suppressed tumor cell proliferation by reducing the MAP3K-MEK-ERK signaling pathway. Of note, in a mouse tumor xenograft model, SCAND1 overexpression significantly reduced Ki-67(+) and Vimentin(+) tumor cells and inhibited migration and lymph node metastasis of prostate cancer. Kaplan-Meier analysis showed high expression of SCAND1 and MZF1 to correlate with better prognoses in pancreatic cancer and head and neck cancers, although with poorer prognosis in kidney cancer. Overall, these data suggest that SCAND1 induces expression and coordinated heterochromatin-binding of MZF1 to reverse the hybrid E/M status into an epithelial phenotype and, inhibits tumor cell proliferation, migration, and metastasis, potentially by repressing the gene expression of EMT drivers and the MAP3K-MEK-ERK signaling pathway.
Collapse
Affiliation(s)
- Takanori Eguchi
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
- Correspondence: (T.E.); (S.K.C.); Tel.: +81-86-235-6661 (T.E.); +1-617-667-4240 (S.K.C.); Fax: +81-86-235-6664 (T.E.); +1-617-667-4245 (S.K.C.)
| | - Eva Csizmadia
- Division of Surgical Sciences, Department of Surgery, Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Hotaka Kawai
- Department of Oral Pathology and Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
| | - Mona Sheta
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
- Department of Cancer Biology, National Cancer Institute, Cairo University, Cairo 11796, Egypt
| | - Kunihiro Yoshida
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
- Department of Oral and Craniofacial Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
| | | | - Barbara Wegiel
- Division of Surgical Sciences, Department of Surgery, Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Stuart K. Calderwood
- Department of Radiation Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
- Correspondence: (T.E.); (S.K.C.); Tel.: +81-86-235-6661 (T.E.); +1-617-667-4240 (S.K.C.); Fax: +81-86-235-6664 (T.E.); +1-617-667-4245 (S.K.C.)
| |
Collapse
|
10
|
Lessey LR, Robinson SC, Chaudhary R, Daniel JM. Adherens junction proteins on the move—From the membrane to the nucleus in intestinal diseases. Front Cell Dev Biol 2022; 10:998373. [PMID: 36274850 PMCID: PMC9581404 DOI: 10.3389/fcell.2022.998373] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
The function and structure of the mammalian epithelial cell layer is maintained by distinct intercellular adhesion complexes including adherens junctions (AJs), tight junctions, and desmosomes. The AJ is most integral for stabilizing cell-cell adhesion and conserving the structural integrity of epithelial tissues. AJs are comprised of the transmembrane protein E-cadherin and cytoplasmic catenin cofactors (α, β, γ, and p120-catenin). One organ where malfunction of AJ is a major contributor to disease states is the mammalian intestine. In the intestine, cell-cell adhesion complexes work synergistically to maintain structural integrity and homeostasis of the epithelium and prevent its malfunction. Consequently, when AJ integrity is compromised in the intestinal epithelium, the ensuing homeostatic disruption leads to diseases such as inflammatory bowel disease and colorectal carcinoma. In addition to their function at the plasma membrane, protein components of AJs also have nuclear functions and are thus implicated in regulating gene expression and intracellular signaling. Within the nucleus, AJ proteins have been shown to interact with transcription factors such as TCF/LEF and Kaiso (ZBTB33), which converge on the canonical Wnt signaling pathway. The multifaceted nature of AJ proteins highlights their complexity in modulating homeostasis and emphasizes the importance of their subcellular localization and expression in the mammalian intestine. In this review, we summarize the nuclear roles of AJ proteins in intestinal tissues; their interactions with transcription factors and how this leads to crosstalk with canonical Wnt signaling; and how nuclear AJ proteins are implicated in intestinal homeostasis and disease.
Collapse
|
11
|
Hivare P, Gangrade A, Swarup G, Bhavsar K, Singh A, Gupta R, Thareja P, Gupta S, Bhatia D. Peptide functionalized DNA hydrogel enhances neuroblastoma cell growth and differentiation. NANOSCALE 2022; 14:8611-8620. [PMID: 35687044 DOI: 10.1039/d1nr07187d] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Designing programmable biomaterials that could act as extracellular matrices and permit functionalization is a current need for tissue engineering advancement. DNA based hydrogels are gaining significant attention owing to their self-assembling properties, biocompatibility, chemical robustness and low batch to batch variability. The real potential of DNA hydrogels in the biomedical domain remains to be explored. In this work, a DNA hydrogel was coated on a glass surface and coupled to a synthetic IKVAV peptide by a chemical crosslinker. We observe enhanced neuronal differentiation, prolonged neurite length, dynamic movement of microtubules and cytoskeleton, and altered endocytic mechanisms in neuroblastoma-based stem cells for the peptide modified DNA hydrogel compared to the unmodified DNA hydrogel and controls. We anticipate that a peptide-modified DNA hydrogel could emerge as a promising scaffold coating material to develop nerve tissue conduits in the future for application in neuroscience and neuroregeneration.
Collapse
Affiliation(s)
- Pravin Hivare
- Biological Engineering Discipline, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India.
| | - Ankit Gangrade
- Biological Engineering Discipline, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India.
| | - Gitanjali Swarup
- Biological Engineering Discipline, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India.
| | - Krishna Bhavsar
- Biological Engineering Discipline, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India.
| | - Ankur Singh
- Biological Engineering Discipline, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India.
| | - Ratnika Gupta
- Chemical Engineering Discipline, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India
| | - Prachi Thareja
- Chemical Engineering Discipline, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India
| | - Sharad Gupta
- Biological Engineering Discipline, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India.
- Center for Biomedical Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India
| | - Dhiraj Bhatia
- Biological Engineering Discipline, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India.
- Center for Biomedical Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India
| |
Collapse
|
12
|
Zheng Y, Wang L, Yin L, Yao Z, Tong R, Xue J, Lu Y. Lung Cancer Stem Cell Markers as Therapeutic Targets: An Update on Signaling Pathways and Therapies. Front Oncol 2022; 12:873994. [PMID: 35719973 PMCID: PMC9204354 DOI: 10.3389/fonc.2022.873994] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 04/25/2022] [Indexed: 02/05/2023] Open
Abstract
Cancer stem cells, a relatively small group of self-renewing cancer cells, were first isolated from acute myeloid leukemia. These cells can play a crucial role in tumor metastasis, relapse, and therapy resistance. The cancer stem cell theory may be applied to lung cancer and explain the inefficiency of traditional treatments and eventual recurrence. However, because of the unclear accuracy and illusive biological function of cancer stem cells, some researchers remain cautious about this theory. Despite the ongoing controversy, cancer stem cells are still being investigated, and their biomarkers are being discovered for application in cancer diagnosis, targeted therapy, and prognosis prediction. Potential lung cancer stem cell markers mainly include surface biomarkers such as CD44, CD133, epithelial cell adhesion molecule, and ATP-binding cassette subfamily G member 2, along with intracellular biomarkers such as aldehyde dehydrogenase, sex-determining region Y-box 2, NANOG, and octamer-binding transcription factor 4. These markers have different structures and functions but are closely associated with the stem potential and uncontrollable proliferation of tumor cells. The aberrant activation of major signaling pathways, such as Notch, Hedgehog, and Wnt, may be associated with the expression and regulation of certain lung cancer stem cell markers, thus leading to lung cancer stem cell maintenance, chemotherapy resistance, and cancer promotion. Treatments targeting lung cancer stem cell markers, including antibody drugs, nanoparticle drugs, chimeric antigen receptor T-cell therapy, and other natural or synthetic specific inhibitors, may provide new hope for patients who are resistant to conventional lung cancer therapies. This review provides comprehensive and updated data on lung cancer stem cell markers with regard to their structures, functions, signaling pathways, and promising therapeutic target approaches, aiming to elucidate potential new therapies for lung cancer.
Collapse
Affiliation(s)
- Yue Zheng
- Department of Thoracic Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Laduona Wang
- Department of Thoracic Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Limei Yin
- Department of Thoracic Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Zhuoran Yao
- Department of Thoracic Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Ruizhan Tong
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jianxin Xue
- Department of Thoracic Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, Chengdu, China
| | - You Lu
- Department of Thoracic Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
13
|
King RE, Ward-Shaw ET, Hu R, Lambert PF, Thibeault SL. Expanded Basal Compartment and Disrupted Barrier in Vocal Fold Epithelium Infected with Mouse Papillomavirus MmuPV1. Viruses 2022; 14:v14051059. [PMID: 35632798 PMCID: PMC9146965 DOI: 10.3390/v14051059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/07/2022] [Accepted: 05/11/2022] [Indexed: 02/01/2023] Open
Abstract
Laryngeal infection with low-risk human papillomaviruses can cause recurrent respiratory papillomatosis (RRP), a disease with severe effects on vocal fold epithelium resulting in impaired voice function and communication. RRP research has been stymied by limited preclinical models. We recently reported a murine model of laryngeal MmuPV1 infection and disease in immunodeficient mice. In the current study, we compare quantitative and qualitative measures of epithelial proliferation, apoptosis, differentiation, and barrier between mice with MmuPV1-induced disease of the larynx and surrounding tissues and equal numbers of uninfected controls. Findings supported our hypothesis that laryngeal MmuPV1 infection recapitulates many features of RRP. Like RRP, MmuPV1 increased proliferation in infected vocal fold epithelium, expanded the basal compartment of cells, decreased differentiated cells, and altered cell–cell junctions and basement membrane. Effects of MmuPV1 on apoptosis were equivocal, as with RRP. Barrier markers resembled human neoplastic disease in severe MmuPV1-induced disease. We conclude that MmuPV1 infection of the mouse larynx provides a useful, if imperfect, preclinical model for RRP that will facilitate further study and treatment development for this intractable and devastating disease.
Collapse
Affiliation(s)
- Renee E. King
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, WI 53705, USA; (R.E.K.); (E.T.W.-S.); (P.F.L.)
- Department of Surgery, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Ella T. Ward-Shaw
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, WI 53705, USA; (R.E.K.); (E.T.W.-S.); (P.F.L.)
| | - Rong Hu
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA;
| | - Paul F. Lambert
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, WI 53705, USA; (R.E.K.); (E.T.W.-S.); (P.F.L.)
| | - Susan L. Thibeault
- Department of Surgery, University of Wisconsin-Madison, Madison, WI 53705, USA
- Correspondence:
| |
Collapse
|
14
|
Chen CN, Wang JC, Chen YT, Yang TL. Exploration of the niche effect on tumor satellite budding of head and neck cancer with biomimicking modeling. Biomaterials 2022; 285:121471. [PMID: 35490561 DOI: 10.1016/j.biomaterials.2022.121471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 03/09/2022] [Accepted: 03/12/2022] [Indexed: 11/24/2022]
|
15
|
Delgado-Bellido D, Garcia-Diaz A, Oliver FJ. Co-immunoprecipitation of Protein Complexes from Different Subcellular Compartments in Vasculogenic Mimicry Studies. Methods Mol Biol 2022; 2514:61-72. [PMID: 35771419 DOI: 10.1007/978-1-0716-2403-6_7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Aberrant extravascular expression of VE-cadherin has been observed in metastasis associated with vasculogenic mimicry (VM); we have recently shown that in VM prone cells VE-cadherin (mainly in the form of phospho-VE-cadherin in Y658) is in part located in the cell nucleus, which associates with p120-catenin and the transcription factor kaiso allowing increased plasticity that potentiates VM development in malignant cells. In this chapter, we describe the protocol to analyze protein-protein interactions in subcellular fractions with particular focus in VE-cadherin. The verification of the subcellular interactome of VE-cadherin and other key proteins involved in VM shed light to novel functions of endothelial proteins aberrantly expressed in tumor cells and their consequences in cell plasticity during VM development.
Collapse
Affiliation(s)
- Daniel Delgado-Bellido
- Instituto de Parasitología y Biomedicina López Neyra, CSIC, Granada, Spain
- Instituto de salud Carlos III, CIBERONC, Madrid, Spain
| | - Angel Garcia-Diaz
- Instituto de Parasitología y Biomedicina López Neyra, CSIC, Granada, Spain
- Instituto de salud Carlos III, CIBERONC, Madrid, Spain
| | - Francisco Javier Oliver
- Instituto de Parasitología y Biomedicina López Neyra, CSIC, Granada, Spain.
- Instituto de salud Carlos III, CIBERONC, Madrid, Spain.
| |
Collapse
|
16
|
Bruun J, Eide PW, Bergsland CH, Bruck O, Svindland A, Arjama M, Välimäki K, Bjørnslett M, Guren MG, Kallioniemi O, Nesbakken A, Lothe RA, Pellinen T. E-cadherin is a robust prognostic biomarker in colorectal cancer and low expression is associated with sensitivity to inhibitors of topoisomerase, aurora, and HSP90 in preclinical models. Mol Oncol 2021; 16:2312-2329. [PMID: 34890102 PMCID: PMC9208074 DOI: 10.1002/1878-0261.13159] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 11/10/2021] [Accepted: 12/09/2021] [Indexed: 12/24/2022] Open
Abstract
Cell–cell and cell–matrix adhesion proteins that have been implicated in colorectal epithelial integrity and epithelial‐to‐mesenchymal transition could be robust prognostic and potential predictive biomarkers for standard and novel therapies. We analyzed in situ protein expression of E‐cadherin (ECAD), integrin β4 (ITGB4), zonula occludens 1 (ZO‐1), and cytokeratins in a single‐hospital series of Norwegian patients with colorectal cancer (CRC) stages I–IV (n = 922) using multiplex fluorescence‐based immunohistochemistry (mfIHC) on tissue microarrays. Pharmacoproteomic associations were explored in 35 CRC cell lines annotated with drug sensitivity data on > 400 approved and investigational drugs. ECAD, ITGB4, and ZO‐1 were positively associated with survival, while cytokeratins were negatively associated with survival. Only ECAD showed independent prognostic value in multivariable Cox models. Clinical and molecular associations for ECAD were technically validated on a different mfIHC platform, and the prognostic value was validated in another Norwegian series (n = 798). In preclinical models, low and high ECAD expression differentially associated with sensitivity to topoisomerase, aurora, and HSP90 inhibitors, and EGFR inhibitors. E‐cadherin protein expression is a robust prognostic biomarker with potential clinical utility in CRC.
Collapse
Affiliation(s)
- Jarle Bruun
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Norway.,K.G. Jebsen Colorectal Cancer Research Centre, Division for Cancer Medicine, Oslo University Hospital, Norway
| | - Peter W Eide
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Norway.,K.G. Jebsen Colorectal Cancer Research Centre, Division for Cancer Medicine, Oslo University Hospital, Norway
| | - Christian Holst Bergsland
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Norway.,K.G. Jebsen Colorectal Cancer Research Centre, Division for Cancer Medicine, Oslo University Hospital, Norway
| | - Oscar Bruck
- Hematology Research Unit Helsinki, University of Helsinki and Comprehensive Cancer Center, Helsinki University Hospital, Finland
| | - Aud Svindland
- K.G. Jebsen Colorectal Cancer Research Centre, Division for Cancer Medicine, Oslo University Hospital, Norway.,Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Norway.,Department of Pathology, Oslo University Hospital, Norway
| | - Mariliina Arjama
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Finland
| | - Katja Välimäki
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Finland
| | - Merete Bjørnslett
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Norway.,K.G. Jebsen Colorectal Cancer Research Centre, Division for Cancer Medicine, Oslo University Hospital, Norway
| | - Marianne G Guren
- K.G. Jebsen Colorectal Cancer Research Centre, Division for Cancer Medicine, Oslo University Hospital, Norway.,Department of Oncology, Oslo University Hospital, Norway
| | - Olli Kallioniemi
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Finland.,Science for Life Laboratory, Department of Oncology & Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Arild Nesbakken
- K.G. Jebsen Colorectal Cancer Research Centre, Division for Cancer Medicine, Oslo University Hospital, Norway.,Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Norway.,Department of Gastrointestinal Surgery, Oslo University Hospital, Norway
| | - Ragnhild A Lothe
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Norway.,K.G. Jebsen Colorectal Cancer Research Centre, Division for Cancer Medicine, Oslo University Hospital, Norway.,Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Norway
| | - Teijo Pellinen
- K.G. Jebsen Colorectal Cancer Research Centre, Division for Cancer Medicine, Oslo University Hospital, Norway.,Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Finland
| |
Collapse
|
17
|
Seebacher NA, Krchniakova M, Stacy AE, Skoda J, Jansson PJ. Tumour Microenvironment Stress Promotes the Development of Drug Resistance. Antioxidants (Basel) 2021; 10:1801. [PMID: 34829672 PMCID: PMC8615091 DOI: 10.3390/antiox10111801] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 10/29/2021] [Accepted: 11/08/2021] [Indexed: 01/18/2023] Open
Abstract
Multi-drug resistance (MDR) is a leading cause of cancer-related death, and it continues to be a major barrier to cancer treatment. The tumour microenvironment (TME) has proven to play an essential role in not only cancer progression and metastasis, but also the development of resistance to chemotherapy. Despite the significant advances in the efficacy of anti-cancer therapies, the development of drug resistance remains a major impediment to therapeutic success. This review highlights the interplay between various factors within the TME that collectively initiate or propagate MDR. The key TME-mediated mechanisms of MDR regulation that will be discussed herein include (1) altered metabolic processing and the reactive oxygen species (ROS)-hypoxia inducible factor (HIF) axis; (2) changes in stromal cells; (3) increased cancer cell survival via autophagy and failure of apoptosis; (4) altered drug delivery, uptake, or efflux and (5) the induction of a cancer stem cell (CSC) phenotype. The review also discusses thought-provoking ideas that may assist in overcoming the TME-induced MDR. We conclude that stressors from the TME and exposure to chemotherapeutic agents are strongly linked to the development of MDR in cancer cells. Therefore, there remains a vast area for potential research to further elicit the interplay between factors existing both within and outside the TME. Elucidating the mechanisms within this network is essential for developing new therapeutic strategies that are less prone to failure due to the development of resistance in cancer cells.
Collapse
Affiliation(s)
| | - Maria Krchniakova
- Department of Experimental Biology, Faculty of Science, Masaryk University, 62500 Brno, Czech Republic;
- International Clinical Research Center, St. Anne’s University Hospital, 65691 Brno, Czech Republic
| | - Alexandra E. Stacy
- Cancer Drug Resistance & Stem Cell Program, School of Medical Science, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia;
| | - Jan Skoda
- Department of Experimental Biology, Faculty of Science, Masaryk University, 62500 Brno, Czech Republic;
- International Clinical Research Center, St. Anne’s University Hospital, 65691 Brno, Czech Republic
| | - Patric J. Jansson
- Cancer Drug Resistance & Stem Cell Program, School of Medical Science, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia;
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St. Leonards, NSW 2065, Australia
| |
Collapse
|
18
|
Yuan E, Liu L, Huang M, Chang B, Qi C, Gou N, Ren J. Effects of complex extracts of traditional Chinese herbs on gastric mucosal injury in rats and potential underlying mechanism. FOOD FRONTIERS 2021; 2:305-315. [DOI: 10.1002/fft2.73] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2025] Open
Abstract
AbstractFive complex extracts (CEs) of seven Chinese herbs (Astragalus, Poria cocos, Alpinia officinarum Hance, Radix Puerariae, Ginseng, Licorice, Hericium erinaceus) were prepared by hot water extraction and evaluated for their effect on gastric ulcer in rats. In rats with acetic acid‐induced chronic gastric ulcer, gross and microscopic appearance showed that gastric mucosal injury index and lesion inhibition rate were improved after CEs gavage for 21 days. Pretreatment with CEs for 21 days in rats with acute gastric ulcer could also improve the gastric mucosal injury by ethanol. CE1, CE4, and CE5 showed more obvious effect in two models. The cell experiments results showed that CE1, CE4, and CE5 effectively inhibited Wnt signaling activity. Thus, they could protect gastric mucosa through inhibiting Wnt signaling pathway. These results indicated that CE1, CE4, and CE5 had significant protective effects on gastric mucosal injury by inhibiting Wnt signalling pathway and could be developed into safe functional products.
Collapse
Affiliation(s)
- Erdong Yuan
- School of Food Science and Technology South China University of Technology Wushan Road 381 Guangzhou Guangdong 510000 China
| | - Liangyun Liu
- School of Food Science and Technology South China University of Technology Wushan Road 381 Guangzhou Guangdong 510000 China
| | - Min Huang
- School of Food Science and Technology South China University of Technology Wushan Road 381 Guangzhou Guangdong 510000 China
| | - Bo Chang
- School of Food Science and Technology South China University of Technology Wushan Road 381 Guangzhou Guangdong 510000 China
| | - Chunli Qi
- School of Food Science and Technology South China University of Technology Wushan Road 381 Guangzhou Guangdong 510000 China
| | - Na Gou
- School of Food Science and Technology South China University of Technology Wushan Road 381 Guangzhou Guangdong 510000 China
| | - Jiaoyan Ren
- School of Food Science and Technology South China University of Technology Wushan Road 381 Guangzhou Guangdong 510000 China
| |
Collapse
|
19
|
Abreu de Oliveira WA, Moens S, El Laithy Y, van der Veer BK, Athanasouli P, Cortesi EE, Baietti MF, Koh KP, Ventura JJ, Amant F, Annibali D, Lluis F. Wnt/β-Catenin Inhibition Disrupts Carboplatin Resistance in Isogenic Models of Triple-Negative Breast Cancer. Front Oncol 2021; 11:705384. [PMID: 34367990 PMCID: PMC8340846 DOI: 10.3389/fonc.2021.705384] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 06/28/2021] [Indexed: 12/11/2022] Open
Abstract
Triple-Negative Breast Cancer (TNBC) is the most aggressive breast cancer subtype, characterized by limited treatment options and higher relapse rates than hormone-receptor-positive breast cancers. Chemotherapy remains the mainstay treatment for TNBC, and platinum salts have been explored as a therapeutic alternative in neo-adjuvant and metastatic settings. However, primary and acquired resistance to chemotherapy in general and platinum-based regimens specifically strongly hampers TNBC management. In this study, we used carboplatin-resistant in vivo patient-derived xenograft and isogenic TNBC cell-line models and detected enhanced Wnt/β-catenin activity correlating with an induced expression of stem cell markers in both resistant models. In accordance, the activation of canonical Wnt signaling in parental TNBC cell lines increases stem cell markers' expression, formation of tumorspheres and promotes carboplatin resistance. Finally, we prove that Wnt signaling inhibition resensitizes resistant models to carboplatin both in vitro and in vivo, suggesting the synergistic use of Wnt inhibitors and carboplatin as a therapeutic option in TNBC. Here we provide evidence for a prominent role of Wnt signaling in mediating resistance to carboplatin, and we establish that combinatorial targeting of Wnt signaling overcomes carboplatin resistance enhancing chemotherapeutic drug efficacy.
Collapse
Affiliation(s)
| | - Stijn Moens
- Leuven Cancer Institute (LKI), Department of Oncology, Gynecological Oncology Lab 3000, KU Leuven, Leuven, Belgium
| | - Youssef El Laithy
- Stem Cell Institute, Department of Development and Regeneration, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Bernard K van der Veer
- Stem Cell Institute, Department of Development and Regeneration, Laboratory for Stem Cell and Developmental Epigenetics, KU Leuven, Leuven, Belgium
| | - Paraskevi Athanasouli
- Stem Cell Institute, Department of Development and Regeneration, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Emanuela Elsa Cortesi
- Translational Cell and Tissue Research - Department of Imaging & Pathology, KU Leuven, Leuven, Belgium
| | | | - Kian Peng Koh
- Stem Cell Institute, Department of Development and Regeneration, Laboratory for Stem Cell and Developmental Epigenetics, KU Leuven, Leuven, Belgium
| | - Juan-Jose Ventura
- Translational Cell and Tissue Research - Department of Imaging & Pathology, KU Leuven, Leuven, Belgium
| | - Frédéric Amant
- Leuven Cancer Institute (LKI), Department of Oncology, Gynecological Oncology Lab 3000, KU Leuven, Leuven, Belgium.,Centre for Gynecologic Oncology Amsterdam (CGOA), Antoni Van Leeuwenhoek-Netherlands Cancer Institute (AvL-NKI), University Medical Center (UMC), Amsterdam, Netherlands
| | - Daniela Annibali
- Leuven Cancer Institute (LKI), Department of Oncology, Gynecological Oncology Lab 3000, KU Leuven, Leuven, Belgium.,Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Frederic Lluis
- Stem Cell Institute, Department of Development and Regeneration, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| |
Collapse
|
20
|
Jariyal H, Gupta C, Andhale S, Gadge S, Srivastava A. Comparative stemness and differentiation of luminal and basal breast cancer stem cell type under glutamine-deprivation. J Cell Commun Signal 2021; 15:207-222. [PMID: 33511560 PMCID: PMC7991029 DOI: 10.1007/s12079-020-00603-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Accepted: 12/20/2020] [Indexed: 12/12/2022] Open
Abstract
Glutamine (gln) metabolism has emerged as a cancer therapeutic target in past few years, however, the effect of gln-deprivation of bCSCs remains elusive in breast cancer. In this study, effect of glutamine on stemness and differentiation potential of bCSCs isolated from MCF-7 and MDAMB-231 were studied. We have shown that bCSCs differentiate into CD24+ epithelial population under gln-deprivation and demonstrated increased expression of epithelial markers such as e-cadherin, claudin-1 and decreased expression of mesenchymal protein n-cadherin. MCF-7-bCSCs showed a decrease in EpCAMhigh population whereas MDAMB-231-bCSCs increased CD44high population in response to gln-deprivation. The expression of intracellular stem cell markers such sox-2, oct-4 and nanog showed a drastic decrease in gene expression under gln-deprived MDAMB-231-bCSCs. Finally, localization of β-catenin in MCF-7 and MDAMB-231 cells showed its accumulation in cytosol or perinuclear space reducing its efficiency to transcribe downstream genes. Conclusively, our study demonstrated that gln-deprivation induces differentiation of bCSCs into epithelial subtypes and also reduces stemness of bCSCs mediated by reduced nuclear localization of β-catenin. It also suggests that basal and luminal bCSCs respond differentially towards changes in extracellular and intracellular gln. This study could significantly affect the gln targeting regimen of breast cancer therapeutics.
Collapse
Affiliation(s)
- Heena Jariyal
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Ahmedabad, Gandhinagar, Gujarat, India
| | - Chanchal Gupta
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Ahmedabad, Gandhinagar, Gujarat, India
| | - Shambhavi Andhale
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Ahmedabad, Gandhinagar, Gujarat, India
| | - Sonali Gadge
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Ahmedabad, Gandhinagar, Gujarat, India
| | - Akshay Srivastava
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research-Ahmedabad, Opposite Air force Station, Palaj, Gandhinagar, Gujarat, 382355, India.
| |
Collapse
|
21
|
Vilchez Larrea S, Valsecchi WM, Fernández Villamil SH, Lafon Hughes LI. First body of evidence suggesting a role of a tankyrase-binding motif (TBM) of vinculin (VCL) in epithelial cells. PeerJ 2021; 9:e11442. [PMID: 34123588 PMCID: PMC8164839 DOI: 10.7717/peerj.11442] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 04/21/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Adherens junctions (AJ) are involved in cancer, infections and neurodegeneration. Still, their composition has not been completely disclosed. Poly(ADP-ribose) polymerases (PARPs) catalyze the synthesis of poly(ADP-ribose) (PAR) as a posttranslational modification. Four PARPs synthesize PAR, namely PARP-1/2 and Tankyrase-1/2 (TNKS). In the epithelial belt, AJ are accompanied by a PAR belt and a subcortical F-actin ring. F-actin depolymerization alters the AJ and PAR belts while PARP inhibitors prevent the assembly of the AJ belt and cortical actin. We wondered which PARP synthesizes the belt and which is the PARylation target protein. Vinculin (VCL) participates in the anchorage of F-actin to the AJ, regulating its functions, and colocalized with the PAR belt. TNKS has been formerly involved in the assembly of epithelial cell junctions. HYPOTHESIS TNKS poly(ADP-ribosylates) (PARylates) epithelial belt VCL, affecting its functions in AJ, including cell shape maintenance. MATERIALS AND METHODS Tankyrase-binding motif (TBM) sequences in hVCL gene were identified and VCL sequences from various vertebrates, Drosophila melanogaster and Caenorhabditis elegans were aligned and compared. Plasma membrane-associated PAR was tested by immunocytofluorescence (ICF) and subcellular fractionation in Vero cells while TNKS role in this structure and cell junction assembly was evaluated using specific inhibitors. The identity of the PARylated proteins was tested by affinity precipitation with PAR-binding reagent followed by western blots. Finally, MCF-7 human breast cancer epithelial cells were subjected to transfection with Tol2-plasmids, carrying a dicistronic expression sequence including Gallus gallus wt VCL (Tol-2-GgVCL), or the same VCL gene with a point mutation in TBM-II (Tol2-GgVCL/*TBM) under the control of a β-actin promoter, plus green fluorescent protein following an internal ribosome entry site (IRES-GFP) to allow the identification of transfected cells without modifying the transfected protein of interest. RESULTS AND DISCUSSION In this work, some of the hypothesis predictions have been tested. We have demonstrated that: (1) VCL TBMs were conserved in vertebrate evolution while absent in C. elegans; (2) TNKS inhibitors disrupted the PAR belt synthesis, while PAR and an endogenous TNKS pool were associated to the plasma membrane; (3) a VCL pool was covalently PARylated; (4) transfection of MCF-7 cells leading to overexpression of Gg-VCL/*TBM induced mesenchymal-like cell shape changes. This last point deserves further investigation, bypassing the limits of our transient transfection and overexpression system. In fact, a 5th testable prediction would be that a single point mutation in VCL TBM-II under endogenous expression control would induce an epithelial to mesenchymal transition (EMT). To check this, a CRISPR/Cas9 substitution approach followed by migration, invasion, gene expression and chemo-resistance assays should be performed.
Collapse
Affiliation(s)
- Salomé Vilchez Larrea
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular “Dr Héctor N. Torres”, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Ciudad Autónoma de Buenos Aires, República Argentina
| | - Wanda Mariela Valsecchi
- Instituto de Química y Fisicoquímica Biológicas, “Prof. Alejandro C. Paladini” (IQUIFIB) Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Ciudad Autónoma de Buenos Aires, República Argentina
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Silvia H. Fernández Villamil
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular “Dr Héctor N. Torres”, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Ciudad Autónoma de Buenos Aires, República Argentina
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Laura I. Lafon Hughes
- Grupo de Biofisicoquímica, Departamento de Ciencias Biológicas, Centro Universitario Regional Litoral Norte (CENUR), Universidad de la República, Salto, Uruguay
- Departamento de Genética, Instituto de Investigaciones Biológicas Clemente Estable, Ministerio de Educación y Cultura, Montevideo, Uruguay
| |
Collapse
|
22
|
SFRPs Are Biphasic Modulators of Wnt-Signaling-Elicited Cancer Stem Cell Properties beyond Extracellular Control. Cell Rep 2020; 28:1511-1525.e5. [PMID: 31390565 DOI: 10.1016/j.celrep.2019.07.023] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 05/31/2019] [Accepted: 07/09/2019] [Indexed: 11/22/2022] Open
Abstract
Secreted frizzled-related proteins (SFRPs) are mainly known for their role as extracellular modulators and tumor suppressors that downregulate Wnt signaling. Using the established (CRISPR/Cas9 targeting promoters of SFRPs and targeting SFRPs transcript) system, we find that nuclear SFRPs interact with β-catenin and either promote or suppress TCF4 recruitment. SFRPs bind with β-catenin on both their N and C termini, which the repressive effects caused by SFRP-β-catenin-N-terminus binding overpower the promoting effects of their binding at the C terminus. By high Wnt activity, β-catenin and SFRPs only bind with their C termini, which results in the upregulation of β-catenin transcriptional activity and cancer stem cell (CSC)-related genes. Furthermore, we identify disulfide bonds of the cysteine-rich domain (CRD) and two threonine phosphorylation events of the netrin-related motif (NTR) domain of SFRPs that are essential for their role as biphasic modulators, suggesting that SFRPs are biphasic modulators of Wnt signaling-elicited CSC properties beyond extracellular control.
Collapse
|
23
|
Wang Y, Zhong Y, Zhang C, Liao J, Wang G. PM2.5 downregulates MicroRNA-139-5p and induces EMT in Bronchiolar Epithelium Cells by targeting Notch1. J Cancer 2020; 11:5758-5767. [PMID: 32913469 PMCID: PMC7477455 DOI: 10.7150/jca.46976] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 07/19/2020] [Indexed: 12/30/2022] Open
Abstract
PM2.5 was closely linked to lung cancer worldwide. However, the mechanism involved in PM2.5 induced lung cancer is still largely unknown. In this study, we performed chronic PM2.5 stimulation animal and cells model to investigate the carcinogenetic mechanisms of PM2.5 by targeting EMT through Notch1 signal pathway. Next, we focused on the miRNA involved in PM2.5 induced Notch1 pathway activation. We found chronic PM2.5 could induce EMT event in vivo and in vitro, while reducing miR-139-5p expression and activating Notch1 pathway meanwhile. And blocking Notch1 signal pathway by specific small molecule inhibitor could reverse PM2.5 induced EMT. Then, overexpression of miR-139-5p downregulated the expression of Notch1 protein in untreated 16HBE cells. Importantly, overexpression of miR-139-5p blocked Notch1 pathway activation and inhibited EMT event in PM2.5 treated cells. These results indicate that PM2.5 induces EMT event through Notch1 signal pathway and miR-139-5p is a novel regulator of PM2.5-induced EMT by targeting Notch1. Our conclusion is that overexpression of miR-139-5p can down-regulate the expression of Notch1 and reverse the occurrence of malignant lung events induced by chronic exposure to PM2.5.
Collapse
Affiliation(s)
- Yunxia Wang
- Department of Respiratory and Critical Care Medicine, Peking University First Hospital, Beijing, China
| | - Yijue Zhong
- Department of Geriatrics, Jiangsu Provincial Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Cheng Zhang
- Department of Respiratory and Critical Care Medicine, Peking University First Hospital, Beijing, China
| | - Jiping Liao
- Department of Respiratory and Critical Care Medicine, Peking University First Hospital, Beijing, China
| | - Guangfa Wang
- Department of Respiratory and Critical Care Medicine, Peking University First Hospital, Beijing, China
| |
Collapse
|
24
|
Li MN, Lu N, Wang YX, Zhang XQ, Zhou Y, Zhang MX. Regulatory mechanism of tumor suppressor gene miR-302b in malignant tumors. Shijie Huaren Xiaohua Zazhi 2020; 28:570-580. [DOI: 10.11569/wcjd.v28.i14.570] [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] [Indexed: 02/06/2023] Open
Abstract
MicroRNAs are a class of endogenous non-coding RNA molecules that regulate the translation of proteins. They play important regulatory roles in the growth, development, and aging of organisms, as well as cell proliferation, differentiation, apoptosis, and even cancer. miR-302b can participate in the regulation of the expression of a variety of genes, and functions as a tumor suppressor gene in the occurrence, development, invasion, and metastasis of malignant tumors. This article discusses the expression of miR-302b in malignant tumors and its biological functions and molecular mechanism.
Collapse
Affiliation(s)
- Min-Na Li
- Department of Gastroenterology, the First Affiliated Hospital of Xi'an Medical University, Xi'an 710077, Shaanxi Province, China
| | - Ning Lu
- Department of Gastroenterology, the First Affiliated Hospital of Xi'an Medical University, Xi'an 710077, Shaanxi Province, China
| | - Yi-Xuan Wang
- Department of Gastroenterology, the First Affiliated Hospital of Xi'an Medical University, Xi'an 710077, Shaanxi Province, China
| | - Xiao-Qing Zhang
- Department of Gastroenterology, the First Affiliated Hospital of Xi'an Medical University, Xi'an 710077, Shaanxi Province, China
| | - Ying Zhou
- Shaanxi University of Traditional Chinese Medicine, Xianyang 712046, Shaanxi Province, China
| | - Ming-Xin Zhang
- Department of Gastroenterology, the First Affiliated Hospital of Xi'an Medical University, Xi'an 710077, Shaanxi Province, China,the Second Clinical Medical College of Shaanxi University of Chinese Medicine, Xianyang 712046, Shaanxi Province, China
| |
Collapse
|
25
|
Yuan S, Gopal JV, Ren S, Chen L, Liu L, Gao Z. Anticancer fungal natural products: Mechanisms of action and biosynthesis. Eur J Med Chem 2020; 202:112502. [PMID: 32652407 DOI: 10.1016/j.ejmech.2020.112502] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 05/20/2020] [Accepted: 05/25/2020] [Indexed: 01/07/2023]
Abstract
Many fungal metabolites show promising anticancer properties both in vitro and in animal models, and some synthetic analogs of those metabolites have progressed into clinical trials. However, currently, there are still no fungi-derived agents approved as anticancer drugs. Two potential reasons could be envisioned: 1) lacking a clear understanding of their anticancer mechanism of action, 2) unable to supply enough materials to support the preclinical and clinic developments. In this review, we will summarize recent efforts on elucidating the anticancer mechanisms and biosynthetic pathways of several promising anticancer fungal natural products.
Collapse
Affiliation(s)
- Siwen Yuan
- School of Marine Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Jannu Vinay Gopal
- School of Marine Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Shuya Ren
- School of Marine Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Litong Chen
- School of Marine Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Lan Liu
- School of Marine Sciences, Sun Yat-sen University, Guangzhou, 510006, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, 519000, China
| | - Zhizeng Gao
- School of Marine Sciences, Sun Yat-sen University, Guangzhou, 510006, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, 519000, China.
| |
Collapse
|
26
|
Mirhashemi M, Ghazi N, Saghravanian N, Taghipour A, Mohajertehran F. Evaluation of CD24 and CD44 as cancer stem cell markers in squamous cell carcinoma and epithelial dysplasia of the oral cavity by q- RT-PCR. Dent Res J (Isfahan) 2020; 17:208-212. [PMID: 32774798 PMCID: PMC7386372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Squamous cell carcinoma (SCC) is the most common cancer of the oral cavity and may be preceded by dysplastic epithelial lesion. Oral SCC (OSCC) contains a rare subset of cancer cells with self-renewal ability, termed as cancer stem cells (CSCs). CD24 and CD44 as CSC markers are cell surface glycoproteins. These markers contribute to the onset, maintenance, and extension of tumor growth, as well as angiogenesis. In the present study, these two markers were simultaneously evaluated to provide a specific phenotype for carcinogenesis process in oral cavity. MATERIALS AND METHODS In this analytical-cross-sectional study, the expression of CD24 and CD44 genes was evaluated in 45 OSCCs (20 low-grade and 25 high-grade) and 15 oral epithelial dysplasia specimens by real-time quantitative reverse transcription polymerase chain reaction. Kruskal-Wallis and Mann-Whitney U-test, Kendall, and Spearman tests were used for statistical analysis. The significance level was considered <0.05. RESULTS High expression of both markers genes was reported in two-thirds of samples. There was no significant difference between studied groups in gene expression of CD24 and CD44 whereas statistically significant association between CD24 and CD44 was observed in all three groups. This correlation was more significant in OSCC groups (P < 0.001). CONCLUSION High expression of CSC markers in OSCC and oral epithelial dysplasia revealed the importance of accurate examination of dysplastic lesions with high expression of these markers and the possibility of malignant transformation. Regarding a significant association of two markers, further studies are necessary to provide a specific phenotype (CD44 high CD24 high) for these lesions.
Collapse
Affiliation(s)
- Majid Mirhashemi
- Department of Oral and Maxillofacial Pathology, School of Dentistry, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Narges Ghazi
- Department of Oral and Maxillofacial Pathology, School of Dentistry, Mashhad University of Medical Sciences, Mashhad, Iran,Oral and Maxillofacial Diseases Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Nasrollah Saghravanian
- Department of Oral and Maxillofacial Pathology, School of Dentistry, Mashhad University of Medical Sciences, Mashhad, Iran,Oral and Maxillofacial Diseases Research Center, Mashhad University of Medical Sciences, Mashhad, Iran,Address for correspondence: Dr. Nasrollah Saghravanian, Department of Oral and Maxillofacial Pathology, School of Dentistry, Mashhad University of Medical Sciences, Mashhad, Iran. E-mail:
| | - Ali Taghipour
- Department of Biostatistics and Epidemiology, School of Health, Mashhad University of Medical Sciences, Mashhad, Iran
| | | |
Collapse
|
27
|
Saghravanian N, Mirhashemi M, Ghazi N, Taghipour A, Mohajertehran F. Evaluation of CD24 and CD44 as cancer stem cell markers in squamous cell carcinoma and epithelial dysplasia of the oral cavity by q- RT-PCR. Dent Res J (Isfahan) 2020. [DOI: 10.4103/1735-3327.284727] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
28
|
Li F, Dai L, Niu J. GPX2 silencing relieves epithelial-mesenchymal transition, invasion, and metastasis in pancreatic cancer by downregulating Wnt pathway. J Cell Physiol 2019; 235:7780-7790. [PMID: 31774184 DOI: 10.1002/jcp.29391] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 09/27/2019] [Indexed: 12/13/2022]
Abstract
Glutathione peroxidase 2 (GPX2) participates in many cancers including pancreatic cancer (PC), and overexpression of GPX2 promotes tumor growth. Herein, we identified the role of GPX2 in epithelial-mesenchymal transformation (EMT), invasion, and metastasis in PC. Bioinformatics prediction was applied to select PC-related genes. The regulatory function of GPX2 in PC was explored by treatment with short hairpin RNA against GPX2 or LiCl (activator of wingless-type MMTV integration site [Wnt] pathway) in PC cells. GPX2 level in PC tissues, the levels of GPX2, β-catenin, Vimentin, Snail, epithelial-cadherin (E-cadherin), matrix metalloproteinase 2 (MMP2), MMP9, and Wnt2 in cells were determined. Subsequently, cell proliferation, invasion, and metastasis were assayed. Bioinformatics analysis revealed that GPX2 was involved in PC development mediated by the Wnt pathway. GPX2 was highly expressed in PC tissues. GPX2 silencing downregulated levels of β-catenin, Vimentin, Snail, MMP2, MMP9, and Wnt2 but upregulated levels of E-cadherin. It was confirmed that GPX2 silencing suppressed PC cell proliferation, metastasis, and invasion. Furthermore, the trend of EMT and invasion and metastasis of PC induced by the LiCl-activated Wnt pathway was reversed when the GPX2 was silenced. GPX2 silencing could inhibit the Wnt pathway, subsequently suppress PC development.
Collapse
Affiliation(s)
- Fuzhou Li
- Department of Imaging, Linyi People's Hospital, Linyi, China
| | - Lan Dai
- Department of Gynaecology and Obstetrics, Chinese Medicine Hospital of Linyi City, Linyi, China
| | - Jixiang Niu
- Department of General Surgery, Linyi People's Hospital, Linyi, China
| |
Collapse
|
29
|
Sun HR, Wang S, Yan SC, Zhang Y, Nelson PJ, Jia HL, Qin LX, Dong QZ. Therapeutic Strategies Targeting Cancer Stem Cells and Their Microenvironment. Front Oncol 2019; 9:1104. [PMID: 31709180 PMCID: PMC6821685 DOI: 10.3389/fonc.2019.01104] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 10/07/2019] [Indexed: 12/12/2022] Open
Abstract
Cancer stem cells (CSCs) have been demonstrated in a variety of tumors and are thought to act as a clonogenic core for the genesis of new tumor growth. This small subpopulation of cancer cells has been proposed to help drive tumorigenesis, metastasis, recurrence and conventional therapy resistance. CSCs show self-renewal and flexible clonogenic properties and help define specific tumor microenvironments (TME). The interaction between CSCs and TME is thought to function as a dynamic support system that fosters the generation and maintenance of CSCs. Investigation of the interaction between CSCs and the TME is shedding light on the biologic mechanisms underlying the process of tumor malignancy, metastasis, and therapy resistance. We summarize recent advances in CSC biology and their environment, and discuss the challenges and future strategies for targeting this biology as a new therapeutic approach.
Collapse
Affiliation(s)
- Hao-Ran Sun
- Department of General Surgery, Cancer Metastasis Institute, Institutes of Biomedical Sciences, Huashan Hospital, Fudan University, Shanghai, China
| | - Shun Wang
- Department of General Surgery, Cancer Metastasis Institute, Institutes of Biomedical Sciences, Huashan Hospital, Fudan University, Shanghai, China
| | - Shi-Can Yan
- Department of General Surgery, Cancer Metastasis Institute, Institutes of Biomedical Sciences, Huashan Hospital, Fudan University, Shanghai, China
| | - Yu Zhang
- Department of General Surgery, Cancer Metastasis Institute, Institutes of Biomedical Sciences, Huashan Hospital, Fudan University, Shanghai, China
| | - Peter J. Nelson
- Medizinische Klinik und Poliklinik IV, Ludwig-Maximilian-University (LMU), Munich, Germany
| | - Hu-Liang Jia
- Department of General Surgery, Cancer Metastasis Institute, Institutes of Biomedical Sciences, Huashan Hospital, Fudan University, Shanghai, China
| | - Lun-Xiu Qin
- Department of General Surgery, Cancer Metastasis Institute, Institutes of Biomedical Sciences, Huashan Hospital, Fudan University, Shanghai, China
| | - Qiong-Zhu Dong
- Department of General Surgery, Cancer Metastasis Institute, Institutes of Biomedical Sciences, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
30
|
Wang Y, Zhong Y, Hou T, Liao J, Zhang C, Sun C, Wang G. PM2.5 induces EMT and promotes CSC properties by activating Notch pathway in vivo and vitro. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2019; 178:159-167. [PMID: 31002970 DOI: 10.1016/j.ecoenv.2019.03.086] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 03/12/2019] [Accepted: 03/20/2019] [Indexed: 05/20/2023]
Abstract
Fine particulate matter (PM2.5) has been closely linked to increased morbidity and mortality of lung cancer worldwide. However, the role of PM2.5 in the etiology of lung cancer and the mechanism involved in PM2.5 induced lung cancer are largely unknown. In this study, we performed chronic exposure animal model to investigate the carcinogenetic mechanisms of PM2.5 by targeting the induction of epithelial-mesenchymal transition (EMT) and cancer stem cells (CSC) properties through Notch1 signal pathway. The antagonism of Notch1 signal pathway was carried out in vitro cell lines of A549 and BEAS-2B to block EMT and CSC. We found that chronic PM2.5 exposure mice lung tissue pathology showed atypical hyperplasia of bronchiolar epithelium. Then, we discovered that chronic PM2.5 exposure induced notable EMT event and obvious CSC properties indicating the developing process of cell malignant behaviors. EMT characterized with decreased protein expression of E-cadherin and increased protein expression of Vimentin. CSC properties induced by chronic PM2.5 exposure characterized with increased cell-surface markers (ABCG2 and ALDH1A1) and self-renewal genes (SOX2 and OCT4). Furthermore, PM2.5 exposure activate Notch signal pathway by increasing expression of Notch1 and Hes1. At last, we blocked Notch signal pathway by inhibitor RO4929097 in vitro to explore the underlying mechanism mediating PM2.5 induced EMT and CSC. We found that blocking Notch1 could prevent PM2.5 induced malignant behaviors including EMT and CSC in A549 and BEAS-2B. These data revealed that the induction of EMT and CSC properties were involved in the lung cancer risk of PM2.5 in vivo, and blocking-up Notch1 may negatively regulate EMT and CSC to suppress the invasion and migration in vitro, thereby putatively serving as a novel therapeutic target for PM2.5 induced lung cancer.
Collapse
Affiliation(s)
- Yunxia Wang
- Department of Respiratory and Critical Care Medicine, Peking University First Hospital, Beijing, China.
| | - Yijue Zhong
- Department of Respiratory and Critical Care Medicine, Peking University First Hospital, Beijing, China.
| | - Tianfang Hou
- Department of Respiratory and Critical Care Medicine, Peking University First Hospital, Beijing, China.
| | - Jiping Liao
- Department of Respiratory and Critical Care Medicine, Peking University First Hospital, Beijing, China.
| | - Cheng Zhang
- Department of Respiratory and Critical Care Medicine, Peking University First Hospital, Beijing, China.
| | - Chao Sun
- Department of Respiratory and Critical Care Medicine, Peking University First Hospital, Beijing, China.
| | - Guangfa Wang
- Department of Respiratory and Critical Care Medicine, Peking University First Hospital, Beijing, China.
| |
Collapse
|
31
|
Essex A, Pineda J, Acharya G, Xin H, Evans J. Replication Study: Wnt activity defines colon cancer stem cells and is regulated by the microenvironment. eLife 2019; 8:e45426. [PMID: 31215867 PMCID: PMC6584130 DOI: 10.7554/elife.45426] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 06/07/2019] [Indexed: 01/05/2023] Open
Abstract
As part of the Reproducibility Project: Cancer Biology we published a Registered Report (Evans et al., 2015), that described how we intended to replicate selected experiments from the paper 'Wnt activity defines colon cancer stem cells and is regulated by the microenvironment' (Vermeulen et al., 2010). Here, we report the results. Using three independent primary spheroidal colon cancer cultures that expressed a Wnt reporter construct we observed high Wnt activity was associated with the cell surface markers CD133, CD166, and CD29, but not CD24 and CD44, while the original study found all five markers were correlated with high Wnt activity (Figure 2F; Vermeulen et al., 2010). Clonogenicity was highest in cells with high Wnt activity and clonogenic potential of cells with low Wnt activity were increased by myofibroblast-secreted factors, including HGF. While the effects were in the same direction as the original study (Figure 6D; Vermeulen et al., 2010) whether statistical significance was reached among the different conditions varied. When tested in vivo, we did not find a difference in tumorigenicity between high and low Wnt activity, while the original study found cells with high Wnt activity were more effective in inducing tumors (Figure 7E; Vermeulen et al., 2010). Tumorigenicity, however, was increased with myofibroblast-secreted factors, which was in the same direction as the original study (Figure 7E; Vermeulen et al., 2010), but not statistically significant. Finally, we report meta-analyses for each results where possible.
Collapse
Affiliation(s)
| | | | | | - Hong Xin
- Explora BioLabs Inc, San Diego, United States
| | - James Evans
- PhenoVista Biosciences, San Diego, United States
| |
Collapse
|
32
|
Combination of 5-fluorouracil and thymoquinone targets stem cell gene signature in colorectal cancer cells. Cell Death Dis 2019; 10:379. [PMID: 31097715 PMCID: PMC6522523 DOI: 10.1038/s41419-019-1611-4] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 04/06/2019] [Accepted: 04/17/2019] [Indexed: 12/12/2022]
Abstract
Cancer stem cells (CSCs) residing in colorectal cancer tissues have tumorigenic capacity and contribute to chemotherapeutic resistance and disease relapse. It is well known that the survival of colorectal CSCs after 5-fluorouracil (5-FU)-based therapy leads to cancer recurrence. Thus CSCs represent a promising drug target. Here, we designed and synthesized novel hybrid molecules linking 5-FU with the plant-derived compound thymoquinone (TQ) and tested the potential of individual compounds and their combination to eliminate colorectal CSCs. Both, Combi and SARB hybrid showed augmented cytotoxicity against colorectal cancer cells, but were non-toxic to organoids prepared from healthy murine small intestine. NanoString analysis revealed a unique signature of deregulated gene expression in response to the combination of TQ and 5-FU (Combi) and SARB treatment. Importantly, two principle stem cell regulatory pathways WNT/ß-Catenin and PI3K/AKT were found to be downregulated after Combi and hybrid treatment. Furthermore, both treatments strikingly eliminated CD133+ CSC population, accompanying the depleted self-renewal capacity by eradicating long-term propagated 3D tumor cell spheres at sub-toxic doses. In vivo xenografts on chicken eggs of SARB-treated HCT116 cells showed a prominent nuclear ß-Catenin and E-cadherin staining. This was in line with the reduced transcriptional activity of ß-Catenin and diminished cell adhesion under SARB exposure. In contrast to 5-FU, both, Combi and SARB treatment effectively reduced the angiogenic capacity of the remaining resistant tumor cells. Taken together, combination or hybridization of single compounds target simultaneously a broader spectrum of oncogenic pathways leading to an effective eradication of colorectal cancer cells.
Collapse
|
33
|
Hoppe T, Kraus D, Probstmeier R, Jepsen S, Winter J. Stimulation with Porphyromonas gingivalis enhances malignancy and initiates anoikis resistance in immortalized oral keratinocytes. J Cell Physiol 2019; 234:21903-21914. [PMID: 31026063 DOI: 10.1002/jcp.28754] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 04/01/2019] [Accepted: 04/11/2019] [Indexed: 12/13/2022]
Abstract
The aim of this study was to get new insights into molecular processes involved in tumor propagation of immortalized oral keratinocytes induced by the keystone pathogen Porphyromonas gingivalis. Cell culture experiments with immortalized OKF6 cells were performed to analyze cellular effects caused by bacterial stimulation focusing on altered gene expression, signaling pathways, proliferation rate, cell viability, migration and invasion behavior, and on the development of antiapoptotic pathways. Gene and protein expression were analyzed using real-time polymerase chain reaction, enzyme-linked immunosorbent assay, western blot, and protein arrays. Trypan blue staining was used to analyze proliferation and viability, transwell assays for cellular migration, Matrigel assays for invasion, and anoikis-assays for evaluating anoikis resistance. Stimulation of OKF6 cells with Porphyromonas gingivalis led to an alteration in the molecular repertoire of proteins which are involved in cell proliferation, epithelial-mesenchymal transition, stem cell formation, migration, invasion, and anoikis resistance. Higher proliferation rates were detected in conjunction with an activation of PI3K/Akt signaling and the mTOR-pathway. Additionally, inhibition of glycogen-synthase-kinase3-β led to stabilization of β-catenin and Snail, which resulted in a switch from predominant E-cadherin to N-cadherin expression and increased expression of the stem cell markers Oct3/4, Sox2, and Nanog. Enhanced biosynthesis and enzyme activity of matrix metalloproteinase-9 was accompanied by elevated invasion behavior. Finally, anoikis resistance was detected in stimulated keratinocytes by decreased apoptosis of nonadherent cells and elevated expression of epidermal growth factor receptor and c-Met. Hence, Porphyromonas gingivalis is able to induce a more aggressive tumor-like phenotype in immortalized oral keratinocytes, thus contributing to enhanced tumor features.
Collapse
Affiliation(s)
- T Hoppe
- Department of Periodontology, Operative and Preventive Dentistry, University of Bonn, Bonn, Germany
| | - D Kraus
- Department of Prosthodontics, Preclinical Education, and Material Sciences, University of Bonn, Bonn, Germany
| | - R Probstmeier
- Department of Nuclear Medicine, Neuro- and Tumor Cell Biology Group, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - S Jepsen
- Department of Periodontology, Operative and Preventive Dentistry, University of Bonn, Bonn, Germany
| | - J Winter
- Department of Periodontology, Operative and Preventive Dentistry, University of Bonn, Bonn, Germany
| |
Collapse
|
34
|
Abstract
Cancer metastasis is the second leading cause of death in the United States. Despite its morbidity, metastasis is an inefficient process that few cells can survive. However, cancer cells can overcome these metastatic barriers via cellular responses to microenvironmental cues, such as through mechanotransduction. This review focuses on the mechanosensitive ion channels TRPV4 and P2X7, and their roles in metastasis, as both channels have been shown to significantly affect tumor cell dissemination. Upon activation, these channels help form tumor neovasculature, promote transendothelial migration, and increase cell motility. Conversely, they have also been linked to forms of cancer cell death dependent upon levels of activation, implying the complex functionality of mechanosensitive ion channels. Understanding the roles of TRPV4, P2X7 and other mechanosensitive ion channels in these processes may reveal new possible drug targets that modify channel function to reduce a tumor's metastatic potential.
Collapse
|
35
|
Druzhkova I, Ignatova N, Prodanets N, Kiselev N, Zhukov I, Shirmanova M, Zagainov V, Zagaynova E. E-Cadherin in Colorectal Cancer: Relation to Chemosensitivity. Clin Colorectal Cancer 2019; 18:e74-e86. [PMID: 30415989 DOI: 10.1016/j.clcc.2018.10.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Revised: 04/13/2018] [Accepted: 10/09/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND The conventional chemotherapy of colorectal cancer with irinotecan, 5-fluorouracil, and oxaliplatin remains one of the front-line treatments worldwide. However, its efficacy is quite low. Recently studies of the epithelial-mesenchymal transition (EMT) have become the focus of investigations into the cause of chemoresistance in several types of cancer, including colorectal cancer. The data about the role of EMT in chemosensitivity are controversial. MATERIALS AND METHODS Human colon adenocarcinoma cell lines HT29 and HCT116 and 14 primary short-term cultures established from patient tumors were used. The chemosensitivity to irinotecan, 5-fluorouracil, and oxaliplatin was assessed using the (4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) test. Immunocytochemistry, immunohistochemistry, and Western blot test were used to investigate the E-cadherin expression, the loss of which is a major hallmark of EMT. RESULTS Elevated chemosensitivity of the cell line with EMT phenotype, HCT116, was demonstrated. Increased chemosensitivity was revealed in HT29 cell line upon EMT induction. E-cadherin-positive short-term cultures were more resistant to all the drugs tested, whereas each of E-cadherin-negative cultures showed sensitivity to at least one drug. The statistically significant dependency of cells viability on the E-cadherin expression (P < .04) was demonstrated on the short-term cultures using 2 concentrations of each drug. CONCLUSION The data obtained may serve as a basis for the analysis of colon cancer chemosensitivity using short-term cultures and the assay of E-cadherin expression.
Collapse
Affiliation(s)
- Irina Druzhkova
- Privolzhsky Research Medical University (PRMU), Nizhny Novgorod, Russia.
| | - Nadezhda Ignatova
- Privolzhsky Research Medical University (PRMU), Nizhny Novgorod, Russia
| | - Natalia Prodanets
- Privolzhsky Research Medical University (PRMU), Nizhny Novgorod, Russia
| | | | - Iliya Zhukov
- Privolzhsky Research Medical University (PRMU), Nizhny Novgorod, Russia
| | - Marina Shirmanova
- Privolzhsky Research Medical University (PRMU), Nizhny Novgorod, Russia
| | | | - Elena Zagaynova
- Privolzhsky Research Medical University (PRMU), Nizhny Novgorod, Russia
| |
Collapse
|
36
|
Zhao Y, Yu T, Zhang N, Chen J, Zhang P, Li S, Luo L, Cui Z, Qin Y, Liu F. Nuclear E-Cadherin Acetylation Promotes Colorectal Tumorigenesis via Enhancing β-Catenin Activity. Mol Cancer Res 2018; 17:655-665. [PMID: 30401720 DOI: 10.1158/1541-7786.mcr-18-0637] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 09/23/2018] [Accepted: 10/18/2018] [Indexed: 11/16/2022]
Abstract
The E-cadherin/β-catenin signaling pathway plays a critical role in the maintenance of epithelial architecture and regulation of tumor progression. Normally, E-cadherin locates on the cell surface with its cytosolic domain linking to the actin cytoskeleton through interaction with catenins. Although the nuclear localization of E-cadherin has been frequently observed in various types of cancers, little is known regarding the functional consequences of its nuclear translocation. Here, we showed that in colorectal cancer samples and cell lines, E-cadherin localized in the nucleus; and the nuclear localization was mediated through protein interaction with CTNND1. In the nucleus, E-cadherin was acetylated by CREB-binding protein at Lysine870 and Lysine871 in its β-catenin-binding domain, and the acetylation can be reversed by SIRT2. Acetylation of nuclear E-cadherin attenuated its interaction with β-catenin, which therefore released β-catenin from the complex, resulting in increased expression of its downstream genes and accelerated tumor growth and migration. Further study showed that acetylation level of nuclear E-cadherin had high prognostic significance in clinical colorectal samples. Taken together, our findings reveal a novel mechanism of tumor progression through posttranslational modification of E-cadherin, which may serve as a potential drug target of tumor therapy. IMPLICATIONS: This finding that acetylation of nuclear E-cadherin regulates β-catenin activity expands our understanding of the acetylation of E-cadherin promotes colorectal cancer cell growth and suggests novel therapeutic approaches of targeting acetylation in tumors.
Collapse
Affiliation(s)
- Yongxu Zhao
- Key Laboratory of Stem Cell Biology, Shanghai Jiao Tong University School of Medicine (SJTUSM) and Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai, P.R. China
| | - Tao Yu
- Key Laboratory of Stem Cell Biology, Shanghai Jiao Tong University School of Medicine (SJTUSM) and Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai, P.R. China
| | - Nan Zhang
- Department of Oncology, Jinan Central Hospital afflicted to Shandong University, Shandong, P.R. China
| | - Jianxia Chen
- Shanghai Key Laboratory of Mycobacterium Tuberculosis, Shanghai Pulmonary Hospital, Tongji University Medical School, Shanghai, P.R. China
| | - Peng Zhang
- Key Laboratory of Stem Cell Biology, Shanghai Jiao Tong University School of Medicine (SJTUSM) and Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai, P.R. China
| | - Shuang Li
- Key Laboratory of Stem Cell Biology, Shanghai Jiao Tong University School of Medicine (SJTUSM) and Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai, P.R. China
| | - Lijun Luo
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Department of Periodontology, School of Stomatology, Tongji University, Shanghai, P.R. China
| | - Zhenling Cui
- Shanghai Key Laboratory of Mycobacterium Tuberculosis, Shanghai Pulmonary Hospital, Tongji University Medical School, Shanghai, P.R. China
| | - Yue Qin
- Key Laboratory of Stem Cell Biology, Shanghai Jiao Tong University School of Medicine (SJTUSM) and Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai, P.R. China.
| | - Feng Liu
- Key Laboratory of Stem Cell Biology, Shanghai Jiao Tong University School of Medicine (SJTUSM) and Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai, P.R. China. .,Department of Oncology, Jinan Central Hospital afflicted to Shandong University, Shandong, P.R. China.,Shanghai Key Laboratory of Mycobacterium Tuberculosis, Shanghai Pulmonary Hospital, Tongji University Medical School, Shanghai, P.R. China
| |
Collapse
|
37
|
Liou GY. CD133 as a regulator of cancer metastasis through the cancer stem cells. Int J Biochem Cell Biol 2018; 106:1-7. [PMID: 30399449 DOI: 10.1016/j.biocel.2018.10.013] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 10/28/2018] [Accepted: 10/31/2018] [Indexed: 02/06/2023]
Abstract
Cancer stem cells are the cancer cells that have abilities to self-renew, differentiate into defined progenies, and initiate and maintain tumor growth. They also contribute to cancer metastasis and therapeutic resistance, both of which are the major causes of cancer mortality. Among the reported makers of the cancer stem cells, CD133 is the most well-known marker for isolating and studying cancer stem cells in different types of cancer. The CD133high population of cancer cells are not only capable of self-renewal, proliferation, but also highly metastatic and resistant to therapy. Despite very limited information on physiological functions of CD133, many ongoing studies are aimed to reveal the mechanisms that CD133 utilizes to modulate cancer dissemination and drug resistance with a long-term goal for bringing down the number of cancer deaths. In this review, in addition to the regulation of CD133, and its involvement in cancer initiation, and development, the recent updates on how CD133 modulates cancer dissemination, and therapeutic resistance are provided. The key signaling pathways that are upstream or downstream of CD133 during these processes are summarized. A comprehensive understanding of CD133-mediated cancer initiation, development, and dissemination through its pivotal role in cancer stem cells will offer new strategies in cancer therapy.
Collapse
Affiliation(s)
- Geou-Yarh Liou
- Clark Atlanta University, Center for Cancer Research & Therapeutic Development, and Department of Biological Sciences, 223 James P. Brawley Drive SW, Atlanta, GA 30314, USA.
| |
Collapse
|
38
|
Liu W, Wu J, Shi G, Yue X, Liu D, Zhang Q. Aberrant promoter methylation of PCDH10 as a potential diagnostic and prognostic biomarker for patients with breast cancer. Oncol Lett 2018; 16:4462-4470. [PMID: 30214581 PMCID: PMC6126325 DOI: 10.3892/ol.2018.9214] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Accepted: 06/07/2018] [Indexed: 12/15/2022] Open
Abstract
Protocadherin-10 (PCDH10) is a tumor suppressor gene. Its expression level is downregulated by promoter methylation in certain types of human tumors. The aim of the present study was to examine the expression level and promoter methylation status of PCDH10 in breast cancer cells and to evaluate the association of PCDH10 methylation and tumor progression and prognosis. MethyLight was used to detect the methylation status of PCDH10 in breast cancer tissues and healthy breast tissues. Reverse transcription-quantitative polymerase chain reaction was used to assess the mRNA expression level of PCDH10, as well as to evaluate the association between PCDH10 methylation and clinicopathological features, along with patients' overall survival (OS). PCDH10 5'-C-phosphate-G-3' (CpG) methylated sites were identified in tumor tissues and matched healthy tissues (n=392). Tumor tissues and matched healthy tissues exhibited identifiable PCR results, with PCDH10 gene promoter methylation identified in ductal carcinoma in situ (66%), invasive ductal carcinoma (82%), invasive ductal carcinoma with lymph node metastasis (85.32%) and hereditary breast cancer tissues (72.37%). PCDH10 mRNA expression was significantly decreased in breast cancer tissues compared with healthy breast tissues (P=0.032). PCDH10 methylation was associated with tumor size (P=0.004), but not associated with other clinical factors. Survival analysis revealed that the patients exhibiting methylated-PCDH10 had significantly poorer OS times than patients exhibiting unmethylated-PCDH10 (P<0.0001). Receiver operating characteristic analysis indicated a sensitivity of 75%, a specificity of 62.5%, and an area under the curve of 0.682 for PCDH10. Additionally, the results of the present study indicated that PCDH10 methylation status may be a useful diagnostic and prognostic evaluation biomarker for breast cancer. The results suggested that PCDH10 methylation is a common occurrence in primary breast cancer and is associated with poor survival rates among patients with breast cancer.
Collapse
Affiliation(s)
- Wentao Liu
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, P.R. China
| | - Jin Wu
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, P.R. China
| | - Guangyue Shi
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, P.R. China
| | - Xiaolong Yue
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, P.R. China
| | - Dan Liu
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, P.R. China
| | - Qingyuan Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, P.R. China
| |
Collapse
|
39
|
Tang Y, Berlind J, Mavila N. Inhibition of CREB binding protein-beta-catenin signaling down regulates CD133 expression and activates PP2A-PTEN signaling in tumor initiating liver cancer cells. Cell Commun Signal 2018. [PMID: 29530069 PMCID: PMC5848530 DOI: 10.1186/s12964-018-0222-5] [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: 12/23/2022] Open
Abstract
Background The WNT-beta-catenin pathway is known to regulate cellular homeostasis during development and tissue regeneration. Activation of WNT signaling increases the stability of cytoplasmic beta-catenin and enhances its nuclear translocation. Nuclear beta-catenin function is regulated by transcriptional co-factors such as CREB binding protein (CBP) and p300. Hyper-activated WNT-beta-catenin signaling is associated with many cancers. However, its role in inducing stemness to liver cancer cells, its autoregulation and how it regulates tumor suppressor pathways are not well understood. Here we have investigated the role of CBP-beta-catenin signaling on the expression of CD133, a known stem cell antigen and PP2A-PTEN pathway in tumor initiating liver cancer cells. Methods Human hepatoblastoma cell line HepG2 and clonally expanded CD133 expressing tumor initiating liver cells (TICs) from premalignant murine liver were used in this study. CBP-beta-catenin inhibitor ICG001 was used to target CBP-beta catenin signaling in liver cancer cells in vitro. Western blotting and real time PCR (qPCR) were used to quantify protein expression/phosphorylation and mRNA levels, respectively. CBP and CD133 gene silencing was performed by siRNA transfection. Fluorescence Activated Cell Sorting (FACS) was performed to quantify CD133 positive cells. Protein Phosphatase (PP2A) activity was measured after PP2AC immunoprecipitation. Results CBP inhibitor ICG001 and CBP silencing significantly reduced CD133 expression and anchorage independent growth in HepG2 and murine TICs. CD133 silencing in TICs decreased cell proliferation and expression levels of cell cycle regulatory genes, CyclinD1 and CyclinA2. ICG001 treatment and CBP silencing reduced the levels of phosphoSer380/Tyr382/383PTEN, phosphoSer473-AKT, Phospho-Ser552beta-catenin in TICs. ICG001 mediated de-phosphorylation of PTEN in TICs was PP2A dependent and partly prevented by co-treatment with PP2A inhibitor okadaic acid. Conclusions CBP-beta-catenin signaling promotes stemness via CD133 induction and cell proliferation in TICs. We found a novel functional link between CBP-beta-catenin and PP2A-PTEN-AKT pathway in liver TICs. Therefore, CBP-beta-catenin-PP2A-PTEN-AKT signaling axis could be a novel therapeutic target to prevent liver tumor initiation and cancer recurrence. Electronic supplementary material The online version of this article (10.1186/s12964-018-0222-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yuanyuan Tang
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.,Division of Digestive and Liver Diseases, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Joshua Berlind
- Division of Digestive and Liver Diseases, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Nirmala Mavila
- Division of Digestive and Liver Diseases, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA. .,Division of Applied Cell Biology and Physiology, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA.
| |
Collapse
|
40
|
Wang Y, Mo Y, Wang L, Su P, Xie Y. Let-7b contributes to hepatocellular cancer progression through Wnt/β-catenin signaling. Saudi J Biol Sci 2018; 25:953-958. [PMID: 30108446 PMCID: PMC6087813 DOI: 10.1016/j.sjbs.2018.03.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 03/01/2018] [Accepted: 03/05/2018] [Indexed: 12/11/2022] Open
Abstract
Elevated evidences show that microRNAs (miRNAs) play vital roles in tumor progression regulation. However, the functional role of let-7b in hepatocellular carcinoma (HCC) is still largely unknown. In this study, we try to investigate the biological activity of let-7b in human HCC cells and try to find the potential regulatory signaling pathway. Our results indicate that let- 7b was remarkably down-regulated in human HCC tissues by qRT-PCR. In addition, let-7b overexpression decreased the expression of β-catenin and c-Myc, while upregulated E-cadherin expression in HCC cells which was verified by quantitative real-time PCR (qRT-PCR) and western blotting. Furthermore, Wnt/β-catenin was involved in let-7b biological activity which was revealed by luciferase assay. Moreover, Wnt/β-catenin signaling inhibitor blocks HCC cell proliferation which is as the same pattern as let-7b overexpression inhibits in HCC cells proliferation. In conclusion, down-regulated let-7b promotes HCC cell proliferation through Wnt/β-catenin signaling in HCC cells. These results suggested that appropriate manipulation of let-7b might be a new treatment of human HCC in the future.
Collapse
Affiliation(s)
- Yiwei Wang
- North China University of Science and Technology Affiliated Hospital, Tangshan 063000, PR China
| | - Yanbo Mo
- North China University of Science and Technology Affiliated Hospital, Tangshan 063000, PR China
| | - Lin Wang
- North China University of Science and Technology Affiliated Hospital, Tangshan 063000, PR China
| | - Peng Su
- North China University of Technology, Tangshan 063000, PR China
| | - Yuxi Xie
- North China University of Science and Technology Affiliated Hospital, Tangshan 063000, PR China
| |
Collapse
|
41
|
Huang J, He Y, Mcleod HL, Xie Y, Xiao D, Hu H, Chen P, Shen L, Zeng S, Yin X, Ge J, Li L, Tang L, Ma J, Chen Z. miR-302b inhibits tumorigenesis by targeting EphA2 via Wnt/ β-catenin/EMT signaling cascade in gastric cancer. BMC Cancer 2017; 17:886. [PMID: 29273006 PMCID: PMC5741943 DOI: 10.1186/s12885-017-3875-3] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 12/04/2017] [Indexed: 02/08/2023] Open
Abstract
Background EphA2 is a crucial oncogene in gastric cancer (GC) development and metastasis, this study aims to identify microRNAs that target it and serve as key regulators of gastric carcinogenesis. Methods We identified several potential microRNAs targeting EphA2 by bioinformatics websites and then analyzed the role of miR-302b in modulating EphA2 in vitro and in vivo of GC, and it’s mechanism. Results Our analysis identified miR-302b, a novel regulator of EphA2, as one of the most significantly downregulated microRNA (miRNA) in GC tissues. Overexpression of miR-302b impaired GC cell migratory and invasive properties robustly and suppressed cell proliferation by arresting cells at G0–G1 phase in vitro. miR-302b exhibited anti-tumor activity by reversing EphA2 regulation, which relayed a signaling transduction cascade that attenuated the functions of N-cadherin, β-catenin, and Snail (markers of Wnt/β-catenin and epithelial-mesenchymal transition, EMT). This modulation of EphA2 also had distinct effects on cell proliferation and migration in GC in vivo. Conclusions miR-302b serves as a critical suppressor of GC cell tumorigenesis and metastasis by targeting the EphA2/Wnt/β-catenin/EMT pathway. Electronic supplementary material The online version of this article (10.1186/s12885-017-3875-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jin Huang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yijing He
- Department of Dermatology, XiangYa Hospital, Central South University, Changsha, 410008, China
| | - Howard L Mcleod
- Department of Clinical Pharmacology, XiangYa Hospital, Central South University, Changsha, 410008, China.,Hunan Key Laboratory of Pharmacogenetics, Changsha, 410008, China.,Moffitt Cancer Center, DeBartolo Family Personalized Medicine Institute, Tampa, FL, 33612, USA
| | - Yanchun Xie
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Desheng Xiao
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Huabin Hu
- The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510655, China
| | - Pan Chen
- Department of Hepatobiliary Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China
| | - Liangfang Shen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Shan Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Xianli Yin
- Department of gastroenterology and urology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China
| | - Jie Ge
- Department of General Surgery, Xiangya Hospital of Central South University, No.87 Xiangya Road, Changsha, 410008, People's Republic of China
| | - Li Li
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Lanhua Tang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Jian Ma
- Cancer Research Institute, Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Central South University, No.138 Tongzipo Road, Changsha, China.
| | - Zihua Chen
- Department of General Surgery, Xiangya Hospital of Central South University, No.87 Xiangya Road, Changsha, 410008, People's Republic of China.
| |
Collapse
|
42
|
Li N, Shi H, Zhang L, Li X, Gao L, Zhang G, Shi Y, Guo S. miR-188 Inhibits Glioma Cell Proliferation and Cell Cycle Progression Through Targeting β-Catenin. Oncol Res 2017; 26:785-794. [PMID: 29268818 PMCID: PMC7844764 DOI: 10.3727/096504017x15127309628257] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
MicroRNAs (miRNAs) play important roles in several human cancers. Although miR-188 has been suggested to function as a tumor repressor in cancers, its precise role in glioma and the molecular mechanism remain unknown. In the present study, we investigated the effect of miR-188 on glioma and explored its relevant mechanisms. We found that the expression of miR-188 is dramatically downregulated in glioma tissues and cell lines. Subsequent investigation revealed that miR-188 expression was inversely correlated with β-catenin expression in glioma tissue samples. Using a luciferase reporter assay, β-catenin was determined to be a direct target of miR-188. Overexpression of miR-188 reduced β-catenin expression at both the mRNA and protein levels, and inhibition of miR-188 increased β-catenin expression. Moreover, we found that overexpression of miR-188 suppressed glioma cell proliferation and cell cycle G1–S transition, whereas inhibition of miR-188 promoted glioma cell proliferation. Importantly, silencing β-catenin recapitulated the cellular and molecular effects seen upon miR-188 overexpression, which included inhibiting glioma cell proliferation and G1–S transition. Taken together, our results demonstrated that miR-188 inhibits glioma cell proliferation by targeting β-catenin, representing an effective therapeutic strategy for glioma.
Collapse
Affiliation(s)
- Nan Li
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P.R. China
| | - Hangyu Shi
- Department of Neurosurgery, Xi'an Children's Hospital, Xi'an, Shaanxi, P.R. China
| | - Lu Zhang
- Department of Foreign Languages, Ming De College of Northwestern Polytechnical University, Xi'an, Shaanxi, P.R. China
| | - Xu Li
- Department of First Internal Medicine, Shaanxi Province Tumor Hospital, Xi'an, Shaanxi, P.R. China
| | - Lu Gao
- Department of Neurosurgery, Xi'an Children's Hospital, Xi'an, Shaanxi, P.R. China
| | - Gang Zhang
- Department of Neurosurgery, Xi'an Children's Hospital, Xi'an, Shaanxi, P.R. China
| | - Yongqiang Shi
- Department of Neurosurgery, Xi'an Children's Hospital, Xi'an, Shaanxi, P.R. China
| | - Shiwen Guo
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P.R. China
| |
Collapse
|
43
|
Xie D, Zheng GZ, Xie P, Zhang QH, Lin FX, Chang B, Hu QX, Du SX, Li XD. Antitumor activity of resveratrol against human osteosarcoma cells: a key role of Cx43 and Wnt/β-catenin signaling pathway. Oncotarget 2017; 8:111419-111432. [PMID: 29340064 PMCID: PMC5762332 DOI: 10.18632/oncotarget.22810] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Accepted: 11/13/2017] [Indexed: 12/14/2022] Open
Abstract
Osteosarcoma is a high-grade bone sarcoma with strong invasive ability. However, treatment with traditional chemotherapeutic drugs is limited by low tolerability and side effects. Resveratrol has been reported previously to have selective antitumor effect on various tumor cells while little is known about its effects and underlying mechanism in osteosarcoma biology. In this study, we found that resveratrol inhibits proliferation and glycolysis, induces apoptosis and reduces the invasiveness of U2-OS cells in vitro. After treatment with resveratrol, the expression of related Wnt/β-catenin signaling pathway target genes, such as β-catenin, c-myc, cyclin D1, MMP-2 and MMP-9, was downregulated and an increased E-cadherin level was observed as well. Additionally, the dual luciferase assay results also indicated that resveratrol suppressed the activity of Wnt/β-catenin signaling pathway. Interestingly, we noticed that the expression of connexin 43 (Cx43) increased with the prolongation of resveratrol treatment time. To further investigate the relationship between Cx43 and the Wnt/β-catenin signaling pathway in osteosarcoma, we used lentiviral-mediated shRNA to knockdown the expression of Cx43. Knockdown of Cx43 activated the Wnt/β-catenin signaling pathway, promoted proliferation and invasion, and inhibited apoptosis of U2-OS cells. Taken together, our results demonstrate that the antitumor activity of resveratrol against U2-OS cells in vitro occurs through up-regulating Cx43 and E-cadherin, and suppressing the Wnt/β-catenin signaling pathway. Moreover, Cx43 expression is negatively related to the activity of the Wnt/β-catenin pathway in U2-OS cells.
Collapse
Affiliation(s)
- Da Xie
- Department of Orthopedics, The Third Affiliated Hospital (The Affiliated Luohu Hospital) of Shenzhen University, Shenzhen 518000, Guangdong, P. R. China
| | - Gui-Zhou Zheng
- Department of Orthopedics, The Third Affiliated Hospital (The Affiliated Luohu Hospital) of Shenzhen University, Shenzhen 518000, Guangdong, P. R. China
| | - Peng Xie
- Department of Orthopedics, The Third Affiliated Hospital (The Affiliated Luohu Hospital) of Shenzhen University, Shenzhen 518000, Guangdong, P. R. China
| | - Qi-Hao Zhang
- Department of Orthopedics, The Third Affiliated Hospital (The Affiliated Luohu Hospital) of Shenzhen University, Shenzhen 518000, Guangdong, P. R. China
| | - Fei-Xiang Lin
- Department of Orthopedics, The Third Affiliated Hospital (The Affiliated Luohu Hospital) of Shenzhen University, Shenzhen 518000, Guangdong, P. R. China
| | - Bo Chang
- Department of Orthopedics, The Third Affiliated Hospital (The Affiliated Luohu Hospital) of Shenzhen University, Shenzhen 518000, Guangdong, P. R. China
| | - Qin-Xiao Hu
- Department of Orthopedics, The Third Affiliated Hospital (The Affiliated Luohu Hospital) of Shenzhen University, Shenzhen 518000, Guangdong, P. R. China
| | - Shi-Xin Du
- Department of Orthopedics, The Third Affiliated Hospital (The Affiliated Luohu Hospital) of Shenzhen University, Shenzhen 518000, Guangdong, P. R. China
| | - Xue-Dong Li
- Department of Orthopedics, The Third Affiliated Hospital (The Affiliated Luohu Hospital) of Shenzhen University, Shenzhen 518000, Guangdong, P. R. China
| |
Collapse
|
44
|
Wei H, Liang F, Cheng W, Zhou R, Wu X, Feng Y, Wang Y. The mechanisms for lung cancer risk of PM 2.5 : Induction of epithelial-mesenchymal transition and cancer stem cell properties in human non-small cell lung cancer cells. ENVIRONMENTAL TOXICOLOGY 2017; 32:2341-2351. [PMID: 28846189 DOI: 10.1002/tox.22437] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 05/06/2017] [Accepted: 05/14/2017] [Indexed: 05/20/2023]
Abstract
Fine particulate matter (PM2.5 ) is a major component of air pollutions that are closely associated with increased risk of lung cancer. However, the role of PM2.5 in the etiology of lung cancer is largely unknown. In this study, we performed acute (24 hours) and chronic (five passages) exposure models to investigate the carcinogenetic mechanisms of PM2.5 by targeting the induction of epithelial-mesenchymal transition (EMT) and cancer stem cells (CSC) properties in human non-small cell lung cancer cell line A549. We found that both acute and chronic PM2.5 exposure enhanced cell migration and invasion, decreased mRNA expression of epithelial markers and increased mRNA expression of mesenchymal markers. Chronic PM2.5 exposure further induced notable EMT morphology and CSC properties, indicating the developing process of cell malignant behaviors from acute to chronic PM2.5 exposure. CSC properties induced by chronic PM2.5 exposure characterized with increased cell-surface markers (CD44, ABCG2), self-renewal genes (SOX2 and OCT4), side population cells and neoplastic capacity. Furthermore, the levels of three stemness-associated microRNAs, Let-7a, miR-16 and miR-34a, were found to be significantly downregulated by chronic PM2.5 exposure, with microarray data analysis from TCGA database showing their lower expression in human lung adenocarcinoma tissues than that in the adjacent normal lung tissues. These data revealed that the induction of EMT and CSC properties were involved in the lung cancer risk of PM2.5 , and implicated CSC properties and related microRNAs as possible biomarkers for carcinogenicity prediction of PM2.5 .
Collapse
Affiliation(s)
- Hongying Wei
- The Ninth People Hospital of Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Fan Liang
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Wei Cheng
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ren Zhou
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiaomeng Wu
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yan Feng
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yan Wang
- The Ninth People Hospital of Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Shanghai, 200092, China
| |
Collapse
|
45
|
Rosso M, Majem B, Devis L, Lapyckyj L, Besso MJ, Llauradó M, Abascal MF, Matos ML, Lanau L, Castellví J, Sánchez JL, Pérez Benavente A, Gil-Moreno A, Reventós J, Santamaria Margalef A, Rigau M, Vazquez-Levin MH. E-cadherin: A determinant molecule associated with ovarian cancer progression, dissemination and aggressiveness. PLoS One 2017. [PMID: 28934230 DOI: 10.1371/journal.pone.0184439] [] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Ovarian cancer (OC) is the fifth cancer death cause in women worldwide. The malignant nature of this disease stems from its unique dissemination pattern. Epithelial-to-mesenchymal transition (EMT) has been reported in OC and downregulation of Epithelial cadherin (E-cadherin) is a hallmark of this process. However, findings on the relationship between E-cadherin levels and OC progression, dissemination and aggressiveness are controversial. In this study, the evaluation of E-cadherin expression in an OC tissue microarray revealed its prognostic value to discriminate between advanced- and early-stage tumors, as well as serous tumors from other histologies. Moreover, E-cadherin, Neural cadherin (N-cadherin), cytokeratins and vimentin expression was assessed in TOV-112, SKOV-3, OAW-42 and OV-90 OC cell lines grown in monolayers and under anchorage-independent conditions to mimic ovarian tumor cell dissemination, and results were associated with cell aggressiveness. According to these EMT-related markers, cell lines were classified as mesenchymal (M; TOV-112), intermediate mesenchymal (IM; SKOV-3), intermediate epithelial (IE; OAW-42) and epithelial (E; OV-90). M- and IM-cells depicted the highest migration capacity when grown in monolayers, and aggregates derived from M- and IM-cell lines showed lower cell death, higher adhesion to extracellular matrices and higher invasion capacity than E- and IE-aggregates. The analysis of E-cadherin, N-cadherin, cytokeratin 19 and vimentin mRNA levels in 20 advanced-stage high-grade serous human OC ascites showed an IM phenotype in all cases, characterized by higher proportions of N- to E-cadherin and vimentin to cytokeratin 19. In particular, higher E-cadherin mRNA levels were associated with cancer antigen 125 levels more than 500 U/mL and platinum-free intervals less than 6 months. Altogether, E-cadherin expression levels were found relevant for the assessment of OC progression and aggressiveness.
Collapse
Affiliation(s)
- Marina Rosso
- Laboratorio de Estudios de la Interacción Celular en Reproducción y Cáncer, Instituto de Biología y Medicina Experimental (IBYME; CONICET-FIBYME), Buenos Aires, Argentina
| | - Blanca Majem
- Biomedical Research Unit in Gynecology, Vall Hebron Research Institute and University Hospital, Barcelona, Spain
| | - Laura Devis
- Biomedical Research Unit in Gynecology, Vall Hebron Research Institute and University Hospital, Barcelona, Spain
| | - Lara Lapyckyj
- Laboratorio de Estudios de la Interacción Celular en Reproducción y Cáncer, Instituto de Biología y Medicina Experimental (IBYME; CONICET-FIBYME), Buenos Aires, Argentina
| | - María José Besso
- Laboratorio de Estudios de la Interacción Celular en Reproducción y Cáncer, Instituto de Biología y Medicina Experimental (IBYME; CONICET-FIBYME), Buenos Aires, Argentina
| | - Marta Llauradó
- Biomedical Research Unit in Gynecology, Vall Hebron Research Institute and University Hospital, Barcelona, Spain
| | - María Florencia Abascal
- Laboratorio de Estudios de la Interacción Celular en Reproducción y Cáncer, Instituto de Biología y Medicina Experimental (IBYME; CONICET-FIBYME), Buenos Aires, Argentina
| | - María Laura Matos
- Laboratorio de Estudios de la Interacción Celular en Reproducción y Cáncer, Instituto de Biología y Medicina Experimental (IBYME; CONICET-FIBYME), Buenos Aires, Argentina
| | - Lucia Lanau
- Biomedical Research Unit in Gynecology, Vall Hebron Research Institute and University Hospital, Barcelona, Spain
| | - Josep Castellví
- Pathology Department, Vall Hebron University Hospital, Barcelona, Spain
| | - José Luis Sánchez
- Gynecology Oncology Department, Vall Hebron University Hospital, Barcelona, Spain
| | | | - Antonio Gil-Moreno
- Biomedical Research Unit in Gynecology, Vall Hebron Research Institute and University Hospital, Barcelona, Spain
- Gynecology Oncology Department, Vall Hebron University Hospital, Barcelona, Spain
| | - Jaume Reventós
- Biomedical Research Unit in Gynecology, Vall Hebron Research Institute and University Hospital, Barcelona, Spain
| | - Anna Santamaria Margalef
- Biomedical Research Unit in Gynecology, Vall Hebron Research Institute and University Hospital, Barcelona, Spain
| | - Marina Rigau
- Biomedical Research Unit in Gynecology, Vall Hebron Research Institute and University Hospital, Barcelona, Spain
| | - Mónica Hebe Vazquez-Levin
- Laboratorio de Estudios de la Interacción Celular en Reproducción y Cáncer, Instituto de Biología y Medicina Experimental (IBYME; CONICET-FIBYME), Buenos Aires, Argentina
| |
Collapse
|
46
|
Rosso M, Majem B, Devis L, Lapyckyj L, Besso MJ, Llauradó M, Abascal MF, Matos ML, Lanau L, Castellví J, Sánchez JL, Pérez Benavente A, Gil-Moreno A, Reventós J, Santamaria Margalef A, Rigau M, Vazquez-Levin MH. E-cadherin: A determinant molecule associated with ovarian cancer progression, dissemination and aggressiveness. PLoS One 2017. [PMID: 28934230 DOI: 10.1371/journal.pone.0184439]+[] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Ovarian cancer (OC) is the fifth cancer death cause in women worldwide. The malignant nature of this disease stems from its unique dissemination pattern. Epithelial-to-mesenchymal transition (EMT) has been reported in OC and downregulation of Epithelial cadherin (E-cadherin) is a hallmark of this process. However, findings on the relationship between E-cadherin levels and OC progression, dissemination and aggressiveness are controversial. In this study, the evaluation of E-cadherin expression in an OC tissue microarray revealed its prognostic value to discriminate between advanced- and early-stage tumors, as well as serous tumors from other histologies. Moreover, E-cadherin, Neural cadherin (N-cadherin), cytokeratins and vimentin expression was assessed in TOV-112, SKOV-3, OAW-42 and OV-90 OC cell lines grown in monolayers and under anchorage-independent conditions to mimic ovarian tumor cell dissemination, and results were associated with cell aggressiveness. According to these EMT-related markers, cell lines were classified as mesenchymal (M; TOV-112), intermediate mesenchymal (IM; SKOV-3), intermediate epithelial (IE; OAW-42) and epithelial (E; OV-90). M- and IM-cells depicted the highest migration capacity when grown in monolayers, and aggregates derived from M- and IM-cell lines showed lower cell death, higher adhesion to extracellular matrices and higher invasion capacity than E- and IE-aggregates. The analysis of E-cadherin, N-cadherin, cytokeratin 19 and vimentin mRNA levels in 20 advanced-stage high-grade serous human OC ascites showed an IM phenotype in all cases, characterized by higher proportions of N- to E-cadherin and vimentin to cytokeratin 19. In particular, higher E-cadherin mRNA levels were associated with cancer antigen 125 levels more than 500 U/mL and platinum-free intervals less than 6 months. Altogether, E-cadherin expression levels were found relevant for the assessment of OC progression and aggressiveness.
Collapse
Affiliation(s)
- Marina Rosso
- Laboratorio de Estudios de la Interacción Celular en Reproducción y Cáncer, Instituto de Biología y Medicina Experimental (IBYME; CONICET-FIBYME), Buenos Aires, Argentina
| | - Blanca Majem
- Biomedical Research Unit in Gynecology, Vall Hebron Research Institute and University Hospital, Barcelona, Spain
| | - Laura Devis
- Biomedical Research Unit in Gynecology, Vall Hebron Research Institute and University Hospital, Barcelona, Spain
| | - Lara Lapyckyj
- Laboratorio de Estudios de la Interacción Celular en Reproducción y Cáncer, Instituto de Biología y Medicina Experimental (IBYME; CONICET-FIBYME), Buenos Aires, Argentina
| | - María José Besso
- Laboratorio de Estudios de la Interacción Celular en Reproducción y Cáncer, Instituto de Biología y Medicina Experimental (IBYME; CONICET-FIBYME), Buenos Aires, Argentina
| | - Marta Llauradó
- Biomedical Research Unit in Gynecology, Vall Hebron Research Institute and University Hospital, Barcelona, Spain
| | - María Florencia Abascal
- Laboratorio de Estudios de la Interacción Celular en Reproducción y Cáncer, Instituto de Biología y Medicina Experimental (IBYME; CONICET-FIBYME), Buenos Aires, Argentina
| | - María Laura Matos
- Laboratorio de Estudios de la Interacción Celular en Reproducción y Cáncer, Instituto de Biología y Medicina Experimental (IBYME; CONICET-FIBYME), Buenos Aires, Argentina
| | - Lucia Lanau
- Biomedical Research Unit in Gynecology, Vall Hebron Research Institute and University Hospital, Barcelona, Spain
| | - Josep Castellví
- Pathology Department, Vall Hebron University Hospital, Barcelona, Spain
| | - José Luis Sánchez
- Gynecology Oncology Department, Vall Hebron University Hospital, Barcelona, Spain
| | | | - Antonio Gil-Moreno
- Biomedical Research Unit in Gynecology, Vall Hebron Research Institute and University Hospital, Barcelona, Spain
- Gynecology Oncology Department, Vall Hebron University Hospital, Barcelona, Spain
| | - Jaume Reventós
- Biomedical Research Unit in Gynecology, Vall Hebron Research Institute and University Hospital, Barcelona, Spain
| | - Anna Santamaria Margalef
- Biomedical Research Unit in Gynecology, Vall Hebron Research Institute and University Hospital, Barcelona, Spain
| | - Marina Rigau
- Biomedical Research Unit in Gynecology, Vall Hebron Research Institute and University Hospital, Barcelona, Spain
| | - Mónica Hebe Vazquez-Levin
- Laboratorio de Estudios de la Interacción Celular en Reproducción y Cáncer, Instituto de Biología y Medicina Experimental (IBYME; CONICET-FIBYME), Buenos Aires, Argentina
| |
Collapse
|
47
|
Rosso M, Majem B, Devis L, Lapyckyj L, Besso MJ, Llauradó M, Abascal MF, Matos ML, Lanau L, Castellví J, Sánchez JL, Pérez Benavente A, Gil-Moreno A, Reventós J, Santamaria Margalef A, Rigau M, Vazquez-Levin MH. E-cadherin: A determinant molecule associated with ovarian cancer progression, dissemination and aggressiveness. PLoS One 2017; 12:e0184439. [PMID: 28934230 PMCID: PMC5608212 DOI: 10.1371/journal.pone.0184439] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Accepted: 08/23/2017] [Indexed: 12/21/2022] Open
Abstract
Ovarian cancer (OC) is the fifth cancer death cause in women worldwide. The malignant nature of this disease stems from its unique dissemination pattern. Epithelial-to-mesenchymal transition (EMT) has been reported in OC and downregulation of Epithelial cadherin (E-cadherin) is a hallmark of this process. However, findings on the relationship between E-cadherin levels and OC progression, dissemination and aggressiveness are controversial. In this study, the evaluation of E-cadherin expression in an OC tissue microarray revealed its prognostic value to discriminate between advanced- and early-stage tumors, as well as serous tumors from other histologies. Moreover, E-cadherin, Neural cadherin (N-cadherin), cytokeratins and vimentin expression was assessed in TOV-112, SKOV-3, OAW-42 and OV-90 OC cell lines grown in monolayers and under anchorage-independent conditions to mimic ovarian tumor cell dissemination, and results were associated with cell aggressiveness. According to these EMT-related markers, cell lines were classified as mesenchymal (M; TOV-112), intermediate mesenchymal (IM; SKOV-3), intermediate epithelial (IE; OAW-42) and epithelial (E; OV-90). M- and IM-cells depicted the highest migration capacity when grown in monolayers, and aggregates derived from M- and IM-cell lines showed lower cell death, higher adhesion to extracellular matrices and higher invasion capacity than E- and IE-aggregates. The analysis of E-cadherin, N-cadherin, cytokeratin 19 and vimentin mRNA levels in 20 advanced-stage high-grade serous human OC ascites showed an IM phenotype in all cases, characterized by higher proportions of N- to E-cadherin and vimentin to cytokeratin 19. In particular, higher E-cadherin mRNA levels were associated with cancer antigen 125 levels more than 500 U/mL and platinum-free intervals less than 6 months. Altogether, E-cadherin expression levels were found relevant for the assessment of OC progression and aggressiveness.
Collapse
Affiliation(s)
- Marina Rosso
- Laboratorio de Estudios de la Interacción Celular en Reproducción y Cáncer, Instituto de Biología y Medicina Experimental (IBYME; CONICET-FIBYME), Buenos Aires, Argentina
| | - Blanca Majem
- Biomedical Research Unit in Gynecology, Vall Hebron Research Institute and University Hospital, Barcelona, Spain
| | - Laura Devis
- Biomedical Research Unit in Gynecology, Vall Hebron Research Institute and University Hospital, Barcelona, Spain
| | - Lara Lapyckyj
- Laboratorio de Estudios de la Interacción Celular en Reproducción y Cáncer, Instituto de Biología y Medicina Experimental (IBYME; CONICET-FIBYME), Buenos Aires, Argentina
| | - María José Besso
- Laboratorio de Estudios de la Interacción Celular en Reproducción y Cáncer, Instituto de Biología y Medicina Experimental (IBYME; CONICET-FIBYME), Buenos Aires, Argentina
| | - Marta Llauradó
- Biomedical Research Unit in Gynecology, Vall Hebron Research Institute and University Hospital, Barcelona, Spain
| | - María Florencia Abascal
- Laboratorio de Estudios de la Interacción Celular en Reproducción y Cáncer, Instituto de Biología y Medicina Experimental (IBYME; CONICET-FIBYME), Buenos Aires, Argentina
| | - María Laura Matos
- Laboratorio de Estudios de la Interacción Celular en Reproducción y Cáncer, Instituto de Biología y Medicina Experimental (IBYME; CONICET-FIBYME), Buenos Aires, Argentina
| | - Lucia Lanau
- Biomedical Research Unit in Gynecology, Vall Hebron Research Institute and University Hospital, Barcelona, Spain
| | - Josep Castellví
- Pathology Department, Vall Hebron University Hospital, Barcelona, Spain
| | - José Luis Sánchez
- Gynecology Oncology Department, Vall Hebron University Hospital, Barcelona, Spain
| | | | - Antonio Gil-Moreno
- Biomedical Research Unit in Gynecology, Vall Hebron Research Institute and University Hospital, Barcelona, Spain
- Gynecology Oncology Department, Vall Hebron University Hospital, Barcelona, Spain
| | - Jaume Reventós
- Biomedical Research Unit in Gynecology, Vall Hebron Research Institute and University Hospital, Barcelona, Spain
| | - Anna Santamaria Margalef
- Biomedical Research Unit in Gynecology, Vall Hebron Research Institute and University Hospital, Barcelona, Spain
| | - Marina Rigau
- Biomedical Research Unit in Gynecology, Vall Hebron Research Institute and University Hospital, Barcelona, Spain
| | - Mónica Hebe Vazquez-Levin
- Laboratorio de Estudios de la Interacción Celular en Reproducción y Cáncer, Instituto de Biología y Medicina Experimental (IBYME; CONICET-FIBYME), Buenos Aires, Argentina
- * E-mail: ,
| |
Collapse
|
48
|
Powan P, Luanpitpong S, He X, Rojanasakul Y, Chanvorachote P. Detachment-induced E-cadherin expression promotes 3D tumor spheroid formation but inhibits tumor formation and metastasis of lung cancer cells. Am J Physiol Cell Physiol 2017; 313:C556-C566. [PMID: 28931539 DOI: 10.1152/ajpcell.00096.2017] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 09/15/2017] [Accepted: 09/15/2017] [Indexed: 12/12/2022]
Abstract
The epithelial-to-mesenchymal transition is proposed to be a key mechanism responsible for metastasis-related deaths. Similarly, cancer stem cells (CSCs) have been proposed to be a key driver of tumor metastasis. However, the link between the two events and their control mechanisms is unclear. We used a three-dimensional (3D) tumor spheroid assay and other CSC-indicating assays to investigate the role of E-cadherin in CSC regulation and its association to epithelial-to-mesenchymal transition in lung cancer cells. Ectopic overexpression and knockdown of E-cadherin were found to promote and retard, respectively, the formation of tumor spheroids in vitro but had opposite effects on tumor formation and metastasis in vivo in a xenograft mouse model. We explored the discrepancy between the in vitro and in vivo results and demonstrated, for the first time, that E-cadherin is required as a component of a major survival pathway under detachment conditions. Downregulation of E-cadherin increased the stemness of lung cancer cells but had an adverse effect on their survival, particularly on non-CSCs. Such downregulation also promoted anoikis resistance and invasiveness of lung cancer cells. These results suggest that anoikis assay could be used as an alternative method for in vitro assessment of CSCs that involves dysregulated adhesion proteins. Our data also suggest that agents that restore E-cadherin expression may be used as therapeutic agents for metastatic cancers.
Collapse
Affiliation(s)
- Phattrakorn Powan
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand.,Cell-Based Drug and Health Products Development Research Unit, Chulalongkorn University, Bangkok, Thailand.,Department of Pharmaceutical Sciences, West Virginia University, Morgantown, West Virginia; and
| | - Sudjit Luanpitpong
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Xiaoqing He
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, West Virginia; and
| | - Yon Rojanasakul
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, West Virginia; and.,Mary Babb Randolph Cancer Center, West Virginia University, Morgantown, West Virginia
| | - Pithi Chanvorachote
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand; .,Cell-Based Drug and Health Products Development Research Unit, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
49
|
Park SY, Shin JH, Kee SH. E-cadherin expression increases cell proliferation by regulating energy metabolism through nuclear factor-κB in AGS cells. Cancer Sci 2017; 108:1769-1777. [PMID: 28699254 PMCID: PMC5581528 DOI: 10.1111/cas.13321] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 07/04/2017] [Accepted: 07/08/2017] [Indexed: 12/18/2022] Open
Abstract
β-Catenin is a central player in Wnt signaling, and activation of Wnt signaling is associated with cancer development. E-cadherin in complex with β-catenin mediates cell-cell adhesion, which suppresses β-catenin-dependent Wnt signaling. Recently, a tumor-suppressive role for E-cadherin has been reconsidered, as re-expression of E-cadherin was reported to enhance the metastatic potential of malignant tumors. To explore the role of E-cadherin, we established an E-cadherin-expressing cell line, EC96, from AGS cells that featured undetectable E-cadherin expression and a high level of Wnt signaling. In EC96 cells, E-cadherin re-expression enhanced cell proliferation, although Wnt signaling activity was reduced. Subsequent analysis revealed that nuclear factor-κB (NF-κB) activation and consequent c-myc expression might be involved in E-cadherin expression-mediated cell proliferation. To facilitate rapid proliferation, EC96 cells enhance glucose uptake and produce ATP using both mitochondria oxidative phosphorylation and glycolysis, whereas AGS cells use these mechanisms less efficiently. These events appeared to be mediated by NF-κB activation. Therefore, E-cadherin re-expression and subsequent induction of NF-κB signaling likely enhance energy production and cell proliferation.
Collapse
Affiliation(s)
- Song Yi Park
- Department of Microbiology, College of Medicine, Korea University, Seoul, Korea
| | - Jee-Hye Shin
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Sun-Ho Kee
- Department of Microbiology, College of Medicine, Korea University, Seoul, Korea
| |
Collapse
|
50
|
Yuan H, Yu S, Cui Y, Men C, Yang D, Gao Z, Zhu Z, Wu J. Knockdown of mediator subunit Med19 suppresses bladder cancer cell proliferation and migration by downregulating Wnt/β-catenin signalling pathway. J Cell Mol Med 2017. [PMID: 28631286 PMCID: PMC5706513 DOI: 10.1111/jcmm.13229] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Mediator complex subunit 19 (Med19), a RNA polymerase II‐embedded coactivator, is reported to be involved in bladder cancer (BCa) progression, but its functional contribution to this process is poorly understood. Here, we investigate the effects of Med19 on malignant behaviours of BCa, as well as to elucidate the possible mechanisms. Med19 expression in 15 BCa tissues was significantly higher than adjacent paired normal tissues using real‐time PCR and Western blot analysis. Immunohistochemical staining of 167 paraffin‐embedded BCa tissues was performed, and the results showed that high Med19 protein level was positively correlated with clinical stages and histopathological grade. Med19 was knocked down in BCa cells using short‐hairpin RNA. Functional assays showed that knocking‐down of Med19 can suppress cell proliferation and migration in T24, UM‐UC3 cells and 5637 in vitro, and inhibited BCa tumour growth in vivo. TOP/FOPflash reporter assay revealed that Med19 knockdown decreased the activity of Wnt/β‐catenin pathway, and the target genes of Wnt/β‐catenin pathway were down‐regulated, including Wnt2, β‐catenin, Cyclin‐D1 and MMP‐9. However, protein levels of Gsk3β and E‐cadherin were elevated. Our data suggest that Med19 expression correlates with aggressive characteristics of BCa and Med19 knockdown suppresses the proliferation and migration of BCa cells through down‐regulating the Wnt/β‐catenin pathway, thereby highlighting Med19 as a potential therapeutic target for BCa treatment.
Collapse
Affiliation(s)
- Hejia Yuan
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Shengqiang Yu
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Yuanshan Cui
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Changping Men
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Diandong Yang
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Zhenli Gao
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Zhe Zhu
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Jitao Wu
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| |
Collapse
|