1
|
Helweg LP, Storm J, Witte KE, Schulten W, Wrachtrup L, Janotte T, Kitke A, Greiner JFW, Knabbe C, Kaltschmidt B, Simon M, Kaltschmidt C. Targeting Key Signaling Pathways in Glioblastoma Stem Cells for the Development of Efficient Chemo- and Immunotherapy. Int J Mol Sci 2022; 23:12919. [PMID: 36361720 PMCID: PMC9659205 DOI: 10.3390/ijms232112919] [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] [Received: 09/02/2022] [Revised: 09/23/2022] [Accepted: 10/21/2022] [Indexed: 01/12/2024] Open
Abstract
Glioblastoma multiforme (GBM) is the most aggressive and most common malignant brain tumor with poor patient survival despite therapeutic intervention. On the cellular level, GBM comprises a rare population of glioblastoma stem cells (GSCs), driving therapeutic resistance, invasion, and recurrence. GSCs have thus come into the focus of therapeutic strategies, although their targeting remains challenging. In the present study, we took advantage of three GSCs-populations recently established in our lab to investigate key signaling pathways and subsequent therapeutic strategies targeting GSCs. We observed that NF-κB, a crucial transcription factor in GBM progression, was expressed in all CD44+/CD133+/Nestin+-GSC-populations. Exposure to TNFα led to activation of NF-κB-RELA and/or NF-κB-c-REL, depending on the GBM type. GSCs further expressed the proto-oncogene MYC family, with MYChigh GSCs being predominantly located in the tumor spheres ("GROW"-state) while NF-κB-RELAhigh GSCs were migrating out of the sphere ("GO"-state). We efficiently targeted GSCs by the pharmacologic inhibition of NF-κB using PTDC/Bortezomib or inhibition of MYC by KJ-Pyr-9, which significantly reduced GSC-viability, even in comparison to the standard chemotherapeutic drug temozolomide. As an additional cell-therapeutic strategy, we showed that NK cells could kill GSCs. Our findings offer new perspectives for developing efficient patient-specific chemo- and immunotherapy against GBM.
Collapse
Affiliation(s)
- Laureen P. Helweg
- Department of Cell Biology, University of Bielefeld, Universitätsstrasse 25, 33615 Bielefeld, Germany
- Forschungsverbund BioMedizin Bielefeld, OWL (FBMB e.V.), Maraweg 21, 33617 Bielefeld, Germany
| | - Jonathan Storm
- Department of Cell Biology, University of Bielefeld, Universitätsstrasse 25, 33615 Bielefeld, Germany
- Forschungsverbund BioMedizin Bielefeld, OWL (FBMB e.V.), Maraweg 21, 33617 Bielefeld, Germany
| | - Kaya E. Witte
- Department of Cell Biology, University of Bielefeld, Universitätsstrasse 25, 33615 Bielefeld, Germany
- Forschungsverbund BioMedizin Bielefeld, OWL (FBMB e.V.), Maraweg 21, 33617 Bielefeld, Germany
| | - Wiebke Schulten
- Department of Cell Biology, University of Bielefeld, Universitätsstrasse 25, 33615 Bielefeld, Germany
| | - Lennart Wrachtrup
- Department of Cell Biology, University of Bielefeld, Universitätsstrasse 25, 33615 Bielefeld, Germany
| | - Till Janotte
- Department of Cell Biology, University of Bielefeld, Universitätsstrasse 25, 33615 Bielefeld, Germany
| | - Angelika Kitke
- Department of Cell Biology, University of Bielefeld, Universitätsstrasse 25, 33615 Bielefeld, Germany
| | - Johannes F. W. Greiner
- Department of Cell Biology, University of Bielefeld, Universitätsstrasse 25, 33615 Bielefeld, Germany
- Forschungsverbund BioMedizin Bielefeld, OWL (FBMB e.V.), Maraweg 21, 33617 Bielefeld, Germany
| | - Cornelius Knabbe
- Forschungsverbund BioMedizin Bielefeld, OWL (FBMB e.V.), Maraweg 21, 33617 Bielefeld, Germany
- Heart and Diabetes Centre NRW, Institute for Laboratory and Transfusion Medicine, Ruhr-University Bochum, 32545 Bad Oeynhausen, Germany
| | - Barbara Kaltschmidt
- Department of Cell Biology, University of Bielefeld, Universitätsstrasse 25, 33615 Bielefeld, Germany
- Forschungsverbund BioMedizin Bielefeld, OWL (FBMB e.V.), Maraweg 21, 33617 Bielefeld, Germany
- Molecular Neurobiology, Faculty of Biology, Bielefeld University, Universitätsstrasse 25, 33615 Bielefeld, Germany
| | - Matthias Simon
- Forschungsverbund BioMedizin Bielefeld, OWL (FBMB e.V.), Maraweg 21, 33617 Bielefeld, Germany
- Department of Neurosurgery and Epilepsy Surgery, Protestant Hospital of Bethel Foundation, University Medical School OWL at Bielefeld, Bielefeld University, Campus Bielefeld-Bethel, Burgsteig 13, 33617 Bielefeld, Germany
| | - Christian Kaltschmidt
- Department of Cell Biology, University of Bielefeld, Universitätsstrasse 25, 33615 Bielefeld, Germany
- Forschungsverbund BioMedizin Bielefeld, OWL (FBMB e.V.), Maraweg 21, 33617 Bielefeld, Germany
| |
Collapse
|
2
|
Growth dynamics of breast cancer stem cells: effects of self-feedback and EMT mechanisms. Theory Biosci 2022; 141:297-311. [PMID: 35921025 DOI: 10.1007/s12064-022-00374-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 07/06/2022] [Indexed: 10/16/2022]
Abstract
Breast cancer stem cells (BCSCs) with the ability to self-renew and differentiate have been identified in primary breast cancer tissues and cell lines. The BCSCs are often resistant to traditional radiation and/or chemotherapies. Previous studies have also shown that successful therapy must eradicate cancer stem cells. The purpose of this paper is to develop a mathematical model with self-feedback mechanism to illustrate the issues regarding the difficulties of absolutely eliminating a breast cancer. In addition, we introduce the mechanism of the epithelial-mesenchymal transition (EMT) to investigate the influence of EMT on the effects of breast cancer growth and treatment. Results indicate that the EMT mechanism facilitates the growth of breast cancer and makes breast cancer more difficult to be cured. Therefore, targeting the signals involved in EMT can halt tumor progression in breast cancer. Finally, we apply the experimental data to carry out numerical simulations and validate our theoretical conclusions.
Collapse
|
3
|
Survivin Inhibition by Piperine Sensitizes Glioblastoma Cancer Stem Cells and Leads to Better Drug Response. Int J Mol Sci 2022; 23:ijms23147604. [PMID: 35886952 PMCID: PMC9323232 DOI: 10.3390/ijms23147604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 11/17/2022] Open
Abstract
Glioblastoma multiforme (GBM) cancer stem cells (GSCs) are one of the strongest contributing factors to treatment resistance in GBM. Identification of biomarkers capable of directly affecting these cells within the bulk tumor is a major challenge associated with the development of new targeting strategies. In this study, we focus on understanding the potential of the multifunctional extraordinaire survivin as a biomarker for GSCs. We analyzed the expression profiles of this gene using various publicly available datasets to understand its importance in stemness and other cancer processes. The findings from these studies were further validated using human GSCs isolated from a GBM cell line. In these GSCs, survivin was inhibited using the dietary phytochemical piperine (PIP) and the subsequent effects on stemness, cancer processes and Temozolomide were investigated. In silico analysis identified survivin to be one of the most significant differentially regulated gene in GSCs, in comparison to common stemness markers. Further validation studies on the isolated GSCs showed the importance of survivin in stemness, cancer progression and therapy resistance. Taken together, our study identifies survivin as a more consistent GSC marker and also suggests the possibility of using survivin inhibitors along with standard of care drugs for better therapeutic outcomes.
Collapse
|
4
|
Helweg LP, Windmöller BA, Burghardt L, Storm J, Förster C, Wethkamp N, Wilkens L, Kaltschmidt B, Banz-Jansen C, Kaltschmidt C. The Diminishment of Novel Endometrial Carcinoma-Derived Stem-like Cells by Targeting Mitochondrial Bioenergetics and MYC. Int J Mol Sci 2022; 23:ijms23052426. [PMID: 35269569 PMCID: PMC8910063 DOI: 10.3390/ijms23052426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/18/2022] [Accepted: 02/21/2022] [Indexed: 02/01/2023] Open
Abstract
Cancer stem cells (CSCs) are a small subpopulation of tumor cells harboring properties that include self-renewal, multi-lineage differentiation, tumor reconstitution, drug resistance and invasiveness, making them key players in tumor relapse. In the present paper, we develop new CSC models and analyze the molecular pathways involved in survival to identify targets for the establishment of novel therapies. Endometrial carcinoma-derived stem-like cells (ECSCs) were isolated from carcinogenic gynecological tissue and analyzed regarding their expression of prominent CSC markers. Further, they were treated with the MYC-signaling inhibitor KJ-Pyr-9, chemotherapeutic agent carboplatin and type II diabetes medication metformin. ECSC populations express common CSC markers, such as Prominin-1 and CD44 antigen as well as epithelial-to-mesenchymal transition markers, Twist, Snail and Slug, and exhibit the ability to form free-floating spheres. The inhibition of MYC signaling and treatment with carboplatin as well as metformin significantly reduced the cell survival of ECSC-like cells. Further, treatment with metformin significantly decreased the mitochondrial membrane potential of ECSC-like cells, while the extracellular lactate concentration was increased. The established ECSC-like populations represent promising in vitro models to further study the contribution of ECSCs to endometrial carcinogenesis. Targeting MYC signaling as well as mitochondrial bioenergetics has shown promising results in the diminishment of ECSCs, although molecular signaling pathways need further investigations.
Collapse
Affiliation(s)
- Laureen P. Helweg
- Department of Cell Biology, Faculty of Biology, University of Bielefeld, Universitätsstrasse 25, 33615 Bielefeld, Germany; (B.A.W.); (L.B.); (J.S.); (B.K.); (C.K.)
- Forschungsverbund BioMedizin Bielefeld/OWL FBMB e.V., 33615 Bielefeld, Germany; (C.F.); (L.W.); (C.B.-J.)
- Correspondence: ; Tel.: +49-0521-106-5619
| | - Beatrice A. Windmöller
- Department of Cell Biology, Faculty of Biology, University of Bielefeld, Universitätsstrasse 25, 33615 Bielefeld, Germany; (B.A.W.); (L.B.); (J.S.); (B.K.); (C.K.)
- Forschungsverbund BioMedizin Bielefeld/OWL FBMB e.V., 33615 Bielefeld, Germany; (C.F.); (L.W.); (C.B.-J.)
| | - Leonie Burghardt
- Department of Cell Biology, Faculty of Biology, University of Bielefeld, Universitätsstrasse 25, 33615 Bielefeld, Germany; (B.A.W.); (L.B.); (J.S.); (B.K.); (C.K.)
| | - Jonathan Storm
- Department of Cell Biology, Faculty of Biology, University of Bielefeld, Universitätsstrasse 25, 33615 Bielefeld, Germany; (B.A.W.); (L.B.); (J.S.); (B.K.); (C.K.)
- Forschungsverbund BioMedizin Bielefeld/OWL FBMB e.V., 33615 Bielefeld, Germany; (C.F.); (L.W.); (C.B.-J.)
| | - Christine Förster
- Forschungsverbund BioMedizin Bielefeld/OWL FBMB e.V., 33615 Bielefeld, Germany; (C.F.); (L.W.); (C.B.-J.)
- Institute of Pathology, KRH Hospital Nordstadt, Affiliated with the Protestant Hospital of Bethel Foundation, 30167 Hannover, Germany;
| | - Nils Wethkamp
- Institute of Pathology, KRH Hospital Nordstadt, Affiliated with the Protestant Hospital of Bethel Foundation, 30167 Hannover, Germany;
| | - Ludwig Wilkens
- Forschungsverbund BioMedizin Bielefeld/OWL FBMB e.V., 33615 Bielefeld, Germany; (C.F.); (L.W.); (C.B.-J.)
- Institute of Pathology, KRH Hospital Nordstadt, Affiliated with the Protestant Hospital of Bethel Foundation, 30167 Hannover, Germany;
| | - Barbara Kaltschmidt
- Department of Cell Biology, Faculty of Biology, University of Bielefeld, Universitätsstrasse 25, 33615 Bielefeld, Germany; (B.A.W.); (L.B.); (J.S.); (B.K.); (C.K.)
- Forschungsverbund BioMedizin Bielefeld/OWL FBMB e.V., 33615 Bielefeld, Germany; (C.F.); (L.W.); (C.B.-J.)
- Molecular Neurobiology, Faculty of Biology, Bielefeld University, 33615 Bielefeld, Germany
| | - Constanze Banz-Jansen
- Forschungsverbund BioMedizin Bielefeld/OWL FBMB e.V., 33615 Bielefeld, Germany; (C.F.); (L.W.); (C.B.-J.)
- Department of Gynecology and Obstetrics, and Perinatal Center, Protestant Hospital of Bethel Foundation, University Medical School OWL at Bielefeld, Bielefeld University, Campus Bielefeld-Bethel, 33615 Bielefeld, Germany
| | - Christian Kaltschmidt
- Department of Cell Biology, Faculty of Biology, University of Bielefeld, Universitätsstrasse 25, 33615 Bielefeld, Germany; (B.A.W.); (L.B.); (J.S.); (B.K.); (C.K.)
- Forschungsverbund BioMedizin Bielefeld/OWL FBMB e.V., 33615 Bielefeld, Germany; (C.F.); (L.W.); (C.B.-J.)
| |
Collapse
|
5
|
Tabebi M, Kumar Dutta R, Skoglund C, Söderkvist P, Gimm O. Loss of SDHB Induces a Metabolic Switch in the hPheo1 Cell Line toward Enhanced OXPHOS. Int J Mol Sci 2022; 23:560. [PMID: 35008989 PMCID: PMC8745660 DOI: 10.3390/ijms23010560] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 12/26/2021] [Accepted: 12/27/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Enzymes of tricarboxylic acid (TCA) have recently been recognized as tumor suppressors. Mutations in the SDHB subunit of succinate dehydrogenase (SDH) cause pheochromocytomas and paragangliomas (PCCs/PGLs) and predispose patients to malignant disease with poor prognosis. METHODS Using the human pheochromocytoma cell line (hPheo1), we knocked down SDHB gene expression using CRISPR-cas9 technology. RESULTS Microarray gene expression analysis showed that >500 differentially expressed gene targets, about 54%, were upregulated in response to SDHB knock down. Notably, genes involved in glycolysis, hypoxia, cell proliferation, and cell differentiation were up regulated, whereas genes involved in oxidative phosphorylation (OXPHOS) were downregulated. In vitro studies show that hPheo1 proliferation is not affected negatively and the cells that survive by shifting their metabolism to the use of glutamine as an alternative energy source and promote OXPHOS activity. Knock down of SDHB expression results in a significant increase in GLUD1 expression in hPheo1 cells cultured as monolayer or as 3D culture. Analysis of TCGA data confirms the enhancement of GLUD1 in SDHB mutated/low expressed PCCs/PGLs. CONCLUSIONS Our data suggest that the downregulation of SDHB in PCCs/PGLs results in increased GLUD1 expression and may represent a potential biomarker and therapeutic target in SDHB mutated tumors and SDHB loss of activity-dependent diseases.
Collapse
Affiliation(s)
- Mouna Tabebi
- Department of Biomedical and Clinical Sciences (BKV), Linköping University, 581 83 Linköping, Sweden; (R.K.D.); (C.S.); (P.S.)
| | - Ravi Kumar Dutta
- Department of Biomedical and Clinical Sciences (BKV), Linköping University, 581 83 Linköping, Sweden; (R.K.D.); (C.S.); (P.S.)
| | - Camilla Skoglund
- Department of Biomedical and Clinical Sciences (BKV), Linköping University, 581 83 Linköping, Sweden; (R.K.D.); (C.S.); (P.S.)
| | - Peter Söderkvist
- Department of Biomedical and Clinical Sciences (BKV), Linköping University, 581 83 Linköping, Sweden; (R.K.D.); (C.S.); (P.S.)
- Clinical Genomics Linköping, Science for Life Laboratory, Linköping University, 581 83 Linköping, Sweden
| | - Oliver Gimm
- Department of Surgery and Department of Biomedical and Clinical Sciences (BKV), Linköping University, 581 83 Linköping, Sweden;
| |
Collapse
|
6
|
Analysis of Several Pathways for Efficient Killing of Prostate Cancer Stem Cells: A Central Role of NF-κB RELA. Int J Mol Sci 2021; 22:ijms22168901. [PMID: 34445612 PMCID: PMC8396252 DOI: 10.3390/ijms22168901] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/13/2021] [Accepted: 08/15/2021] [Indexed: 12/12/2022] Open
Abstract
Prostate cancer is a common cause of death worldwide. Here, we isolated cancer stem cells (CSCs) from four adenocarcinomas of the prostate (Gleason scores from 3 + 3 up to 4 + 5). CSCs were characterized by the expression of the stem cell markers TWIST, the epithelial cell adhesion molecule (EPCAM), the transcription factors SNAI1 (SNAIL) and SNAI2 (SLUG) and cancer markers such as CD44 and prominin-1 (CD133). All investigated CSC populations contained a fraction highly positive for aldehyde dehydrogenase (ALDH) function and displayed robust expressions of programmed cell death 1 (PD-1) ligands. Furthermore, we investigated immunotherapeutic approaches but had no success even with the clinically used PD-1 inhibitor pembrolizumab. In addition, we studied another death-inducing pathway via interferon gamma signaling and detected high-level upregulations of human leukocyte antigen A (HLA-A) and beta 2-microglobulin (B2M) with only moderate killing efficacy. To examine further killing mechanisms in prostate cancer stem cells (PCSCs), we analyzed NF-κB signaling. Surprisingly, two patient-specific populations of PCSCs were found: one with canonical NF-κB signaling and another one with blunted NF-κB activation, which can be efficiently killed by tumor necrosis factor (TNF). Thus, culturing of PCSCs and analysis of respective NF-κB induction potency after surgery might be a powerful tool for optimizing patient-specific treatment options, such as the use of TNF-inducing chemotherapeutics and/or NF-κB inhibitors.
Collapse
|
7
|
Witte KE, Hertel O, Windmöller BA, Helweg LP, Höving AL, Knabbe C, Busche T, Greiner JFW, Kalinowski J, Noll T, Mertzlufft F, Beshay M, Pfitzenmaier J, Kaltschmidt B, Kaltschmidt C, Banz-Jansen C, Simon M. Nanopore Sequencing Reveals Global Transcriptome Signatures of Mitochondrial and Ribosomal Gene Expressions in Various Human Cancer Stem-like Cell Populations. Cancers (Basel) 2021; 13:cancers13051136. [PMID: 33800955 PMCID: PMC7962028 DOI: 10.3390/cancers13051136] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/03/2021] [Accepted: 03/04/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Cancer is the leading cause of death in the industrialized world. In particular, so-called cancer stem cells (CSCs) play a crucial role in disease progression, as they are known to contribute to tumor growth and metastasis. Thus, CSCs are heavily investigated in a broad range of cancers. Nevertheless, global transcriptomic profiling of CSC populations derived from different tumor types is rare. We established three CSC populations from tumors in the uterus, brain, lung, and prostate and assessed their global transcriptomes using nanopore full-length cDNA sequencing, a new technique to assess insights into global gene profile. We observed common expression in all CSCs for distinct genes encoding proteins for organelles, such as ribosomes, mitochondria, and proteasomes. Additionally, we detected high expressions of inflammation- and immunity-related genes. Conclusively, we observed high similarities between all CSCs independent of their tumor of origin, which may build the basis for identifying novel therapeutic strategies targeting CSCs. Abstract Cancer stem cells (CSCs) are crucial mediators of tumor growth, metastasis, therapy resistance, and recurrence in a broad variety of human cancers. Although their biology is increasingly investigated within the distinct types of cancer, direct comparisons of CSCs from different tumor types allowing comprehensive mechanistic insights are rarely assessed. In the present study, we isolated CSCs from endometrioid carcinomas, glioblastoma multiforme as well as adenocarcinomas of lung and prostate and assessed their global transcriptomes using full-length cDNA nanopore sequencing. Despite the expression of common CSC markers, principal component analysis showed a distinct separation of the CSC populations into three clusters independent of the specific type of tumor. However, GO-term and KEGG pathway enrichment analysis revealed upregulated genes related to ribosomal biosynthesis, the mitochondrion, oxidative phosphorylation, and glycolytic pathways, as well as the proteasome, suggesting a great extent of metabolic flexibility in CSCs. Interestingly, the GO term “NF-kB binding” was likewise found to be elevated in all investigated CSC populations. In summary, we here provide evidence for high global transcriptional similarities between CSCs from various tumors, which particularly share upregulated gene expression associated with mitochondrial and ribosomal activity. Our findings may build the basis for identifying novel therapeutic strategies targeting CSCs.
Collapse
Affiliation(s)
- Kaya E. Witte
- Department of Cell Biology, Faculty of Biology, University of Bielefeld, Universitätsstrasse 25, 33699 Bielefeld, Germany; (B.A.W.); (L.P.H.); (A.L.H.); (J.F.W.G.); (B.K.); (C.K.)
- Forschungsverbund BioMedizin Bielefeld, OWL (FBMB e.V.), Maraweg 21, 33699 Bielefeld, Germany; (C.K.); (F.M.); (M.B.); (J.P.); (C.B.-J.); (M.S.)
- Correspondence: ; Tel.: +49-521-106-5629
| | - Oliver Hertel
- Department of Cell Culture Technology, Faculty of Technology, University of Bielefeld, Universitätsstrasse 25, 33699 Bielefeld, Germany; (O.H.); (T.N.)
- Center for Biotechnology-CeBiTec, University of Bielefeld, Universitätsstrasse 27, 33699 Bielefeld, Germany; (T.B.); (J.K.)
| | - Beatrice A. Windmöller
- Department of Cell Biology, Faculty of Biology, University of Bielefeld, Universitätsstrasse 25, 33699 Bielefeld, Germany; (B.A.W.); (L.P.H.); (A.L.H.); (J.F.W.G.); (B.K.); (C.K.)
- Forschungsverbund BioMedizin Bielefeld, OWL (FBMB e.V.), Maraweg 21, 33699 Bielefeld, Germany; (C.K.); (F.M.); (M.B.); (J.P.); (C.B.-J.); (M.S.)
| | - Laureen P. Helweg
- Department of Cell Biology, Faculty of Biology, University of Bielefeld, Universitätsstrasse 25, 33699 Bielefeld, Germany; (B.A.W.); (L.P.H.); (A.L.H.); (J.F.W.G.); (B.K.); (C.K.)
- Forschungsverbund BioMedizin Bielefeld, OWL (FBMB e.V.), Maraweg 21, 33699 Bielefeld, Germany; (C.K.); (F.M.); (M.B.); (J.P.); (C.B.-J.); (M.S.)
| | - Anna L. Höving
- Department of Cell Biology, Faculty of Biology, University of Bielefeld, Universitätsstrasse 25, 33699 Bielefeld, Germany; (B.A.W.); (L.P.H.); (A.L.H.); (J.F.W.G.); (B.K.); (C.K.)
- Heart and Diabetes Centre NRW, Institute for Laboratory and Transfusion Medicine, Ruhr-University Bochum, 32545 Bad Oeynhausen, Germany
| | - Cornelius Knabbe
- Forschungsverbund BioMedizin Bielefeld, OWL (FBMB e.V.), Maraweg 21, 33699 Bielefeld, Germany; (C.K.); (F.M.); (M.B.); (J.P.); (C.B.-J.); (M.S.)
- Heart and Diabetes Centre NRW, Institute for Laboratory and Transfusion Medicine, Ruhr-University Bochum, 32545 Bad Oeynhausen, Germany
| | - Tobias Busche
- Center for Biotechnology-CeBiTec, University of Bielefeld, Universitätsstrasse 27, 33699 Bielefeld, Germany; (T.B.); (J.K.)
| | - Johannes F. W. Greiner
- Department of Cell Biology, Faculty of Biology, University of Bielefeld, Universitätsstrasse 25, 33699 Bielefeld, Germany; (B.A.W.); (L.P.H.); (A.L.H.); (J.F.W.G.); (B.K.); (C.K.)
- Forschungsverbund BioMedizin Bielefeld, OWL (FBMB e.V.), Maraweg 21, 33699 Bielefeld, Germany; (C.K.); (F.M.); (M.B.); (J.P.); (C.B.-J.); (M.S.)
| | - Jörn Kalinowski
- Center for Biotechnology-CeBiTec, University of Bielefeld, Universitätsstrasse 27, 33699 Bielefeld, Germany; (T.B.); (J.K.)
| | - Thomas Noll
- Department of Cell Culture Technology, Faculty of Technology, University of Bielefeld, Universitätsstrasse 25, 33699 Bielefeld, Germany; (O.H.); (T.N.)
- Center for Biotechnology-CeBiTec, University of Bielefeld, Universitätsstrasse 27, 33699 Bielefeld, Germany; (T.B.); (J.K.)
| | - Fritz Mertzlufft
- Forschungsverbund BioMedizin Bielefeld, OWL (FBMB e.V.), Maraweg 21, 33699 Bielefeld, Germany; (C.K.); (F.M.); (M.B.); (J.P.); (C.B.-J.); (M.S.)
- Scientific Director of the Protestant Hospital of Bethel Foundation, University Medical School OWL at Bielefeld, Bielefeld University, Campus Bielefeld-Bethel, Maraweg 21, 33699 Bielefeld, Germany
| | - Morris Beshay
- Forschungsverbund BioMedizin Bielefeld, OWL (FBMB e.V.), Maraweg 21, 33699 Bielefeld, Germany; (C.K.); (F.M.); (M.B.); (J.P.); (C.B.-J.); (M.S.)
- Department for Thoracic Surgery and Pneumology, Protestant Hospital of Bethel Foundation, University Medical School OWL at Bielefeld, Bielefeld University, Campus Bielefeld-Bethel, Burgsteig 13, 33699 Bielefeld, Germany
| | - Jesco Pfitzenmaier
- Forschungsverbund BioMedizin Bielefeld, OWL (FBMB e.V.), Maraweg 21, 33699 Bielefeld, Germany; (C.K.); (F.M.); (M.B.); (J.P.); (C.B.-J.); (M.S.)
- Department of Urology and Center for Computer-Assisted and Robotic Urology, Protestant Hospital of Bethel Foundation, University Medical School OWL at Bielefeld, Bielefeld University, Campus Bielefeld-Bethel, Burgsteig 13, 33699 Bielefeld, Germany
| | - Barbara Kaltschmidt
- Department of Cell Biology, Faculty of Biology, University of Bielefeld, Universitätsstrasse 25, 33699 Bielefeld, Germany; (B.A.W.); (L.P.H.); (A.L.H.); (J.F.W.G.); (B.K.); (C.K.)
- Forschungsverbund BioMedizin Bielefeld, OWL (FBMB e.V.), Maraweg 21, 33699 Bielefeld, Germany; (C.K.); (F.M.); (M.B.); (J.P.); (C.B.-J.); (M.S.)
- Molecular Neurobiology, Faculty of Biology, Bielefeld University, Universitätsstrasse 25, 33699 Bielefeld, Germany
| | - Christian Kaltschmidt
- Department of Cell Biology, Faculty of Biology, University of Bielefeld, Universitätsstrasse 25, 33699 Bielefeld, Germany; (B.A.W.); (L.P.H.); (A.L.H.); (J.F.W.G.); (B.K.); (C.K.)
- Forschungsverbund BioMedizin Bielefeld, OWL (FBMB e.V.), Maraweg 21, 33699 Bielefeld, Germany; (C.K.); (F.M.); (M.B.); (J.P.); (C.B.-J.); (M.S.)
| | - Constanze Banz-Jansen
- Forschungsverbund BioMedizin Bielefeld, OWL (FBMB e.V.), Maraweg 21, 33699 Bielefeld, Germany; (C.K.); (F.M.); (M.B.); (J.P.); (C.B.-J.); (M.S.)
- Department of Gynecology and Obstetrics, and Perinatal Center, Protestant Hospital of Bethel Foundation, University Medical School OWL at Bielefeld, Bielefeld University, Campus Bielefeld-Bethel, Burgsteig 13, 33699 Bielefeld, Germany
| | - Matthias Simon
- Forschungsverbund BioMedizin Bielefeld, OWL (FBMB e.V.), Maraweg 21, 33699 Bielefeld, Germany; (C.K.); (F.M.); (M.B.); (J.P.); (C.B.-J.); (M.S.)
- Department of Neurosurgery and Epilepsy Surgery, Protestant Hospital of Bethel Foundation, University Medical School OWL at Bielefeld, Bielefeld University, Campus Bielefeld-Bethel, Burgsteig 13, 33699 Bielefeld, Germany
| |
Collapse
|
8
|
Activation of RAS Signalling is Associated with Altered Cell Adhesion in Phaeochromocytoma. Int J Mol Sci 2020; 21:ijms21218072. [PMID: 33138083 PMCID: PMC7663737 DOI: 10.3390/ijms21218072] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 10/25/2020] [Accepted: 10/27/2020] [Indexed: 02/06/2023] Open
Abstract
Phaeochromocytomas and paragangliomas (PPGLs) are neuroendocrine catecholamine-producing tumours that may progress into inoperable metastatic disease. Treatment options for metastatic disease are limited, indicating a need for functional studies to identify pharmacologically targetable pathophysiological mechanisms, which require biologically relevant experimental models. Recently, a human progenitor phaeochromocytoma cell line named “hPheo1” was established, but its genotype has not been characterised. Performing exome sequencing analysis, we identified a KIF1B T827I mutation, and the oncogenic NRAS Q61K mutation. While KIF1B mutations are recurring somatic events in PPGLs, NRAS mutations have hitherto not been detected in PPGLs. Therefore, we aimed to assess its implications for the hPheo1 cell line, and possible relevance for the pathophysiology of PPGLs. We found that transient downregulation of NRAS in hPheo1 led to elevated expression of genes associated with cell adhesion, and enhanced adhesion to hPheo1 cells’ extracellular matrix. Analyses of previously published mRNA data from two independent PPGL patient cohorts (212 tissue samples) revealed a subcluster of PPGLs featuring hyperactivated RAS pathway-signalling and under-expression of cell adhesion-related gene expression programs. Thus, we conclude that NRAS activity in hPheo1 decreases adhesion to their own extracellular matrix and mirrors a transcriptomic RAS-signalling-related phenomenon in PPGLs.
Collapse
|
9
|
Label-free platform on pH-responsive chitosan: Adhesive heterogeneity for cancer stem-like cell isolation from A549 cells via integrin β4. Carbohydr Polym 2020; 239:116168. [PMID: 32414450 DOI: 10.1016/j.carbpol.2020.116168] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 03/11/2020] [Accepted: 03/12/2020] [Indexed: 12/25/2022]
Abstract
Great efforts have been paid to develop methodologies for cancer stem-like cell (CSLC) isolation in anti-cancer research. The major obstacle lies in the lack of generic biomarkers for different cancer types and the requirement of complicated immuno-labeling procedures. The purpose of this study is to establish a label-free platform for CSLC isolation using pH-responsive chitosan. Based on the adhesive heterogeneity, 15.7 ± 1.9 % of human non-small cell lung cancer (NSCLC) cell line A549 detached from the chitosan substrate following medium pH elevation from 6.99 to 7.65 within 1 h. As a result, this subpopulation of cells with low adhesiveness exhibited superior CSLC hallmarks, including self-renewal, invasive and metastatic potential, therapeutic-resistance, colony formation in vitro, as well as nude mice xenograft in vivo for tumorigenesis, in comparison with their high-adhesive counterpart. Furthermore, integrin β4 is decisive in controlling CSLC detachment of NSCLC. Conclusively, this pH-dependent isolation provides new insights into biomaterial-based CSLC isolation.
Collapse
|
10
|
Wang X, Zhao Z, Zhang H, Hou J, Feng W, Zhang M, Guo J, Xia J, Ge Q, Chen X, Wu X. Simultaneous isolation of mesenchymal stem cells and endothelial progenitor cells derived from murine bone marrow. Exp Ther Med 2018; 16:5171-5177. [PMID: 30542473 PMCID: PMC6257072 DOI: 10.3892/etm.2018.6844] [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: 11/12/2017] [Accepted: 09/20/2018] [Indexed: 12/28/2022] Open
Abstract
Mesenchymal stem or stromal cells (MSCs) are identified as sources of pluripotent stem cells with varying degrees of plasticity. Endothelial progenitor cells (EPCs) originate from either bone marrow (BM) or peripheral blood and can mature into cells that line the lumen of blood vessels. MSC and EPC therapies exhibit promising results in a variety of diseases. The current study described the simultaneous isolation of EPCs and MSCs from murine BM using a straightforward approach. The method is based on differences in attachment time and trypsin sensitivity of MSCs and EPCs. The proposed method revealed characteristics of isolated cells. Isolated MSCs were positive for cell surface markers, cluster of differentiation (CD)29, CD44 and stem cell antigen-1 (Sca-1), and negative for hematopoietic surface markers, CD45 and CD11b. Isolated EPCs were positive for Sca-1 and vascular endothelial growth factor receptor 2 and CD133. The results indicate that the proposed method ensured simultaneous isolation of homogenous populations of MSCs and EPCs from murine BM.
Collapse
Affiliation(s)
- Xiaoyi Wang
- Laboratory of Translational Medicine, School of Medicine, Shihezi University, Shihezi, Xinjiang 832008, P.R. China
| | - Zongsheng Zhao
- Department of Animal Genetic Breeding and Reproduction, College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832008, P.R. China
| | - Hongwei Zhang
- Department of General Surgery, First Affiliated Hospital, Shihezi University, Shihezi, Xinjiang 832008, P.R. China
| | - Jixue Hou
- Department of General Surgery, First Affiliated Hospital, Shihezi University, Shihezi, Xinjiang 832008, P.R. China
| | - Wenlei Feng
- Laboratory of Translational Medicine, School of Medicine, Shihezi University, Shihezi, Xinjiang 832008, P.R. China
| | - Meng Zhang
- Laboratory of Translational Medicine, School of Medicine, Shihezi University, Shihezi, Xinjiang 832008, P.R. China
| | - Jun Guo
- Laboratory of Translational Medicine, School of Medicine, Shihezi University, Shihezi, Xinjiang 832008, P.R. China
| | - Jie Xia
- Laboratory of Translational Medicine, School of Medicine, Shihezi University, Shihezi, Xinjiang 832008, P.R. China
| | - Quanhu Ge
- Laboratory of Translational Medicine, School of Medicine, Shihezi University, Shihezi, Xinjiang 832008, P.R. China
| | - Xueling Chen
- Department of Immunology, School of Medicine, Shihezi University, Shihezi, Xinjiang 832008, P.R. China
| | - Xiangwei Wu
- Laboratory of Translational Medicine, School of Medicine, Shihezi University, Shihezi, Xinjiang 832008, P.R. China.,Department of General Surgery, First Affiliated Hospital, Shihezi University, Shihezi, Xinjiang 832008, P.R. China
| |
Collapse
|
11
|
Nallanthighal S, Elmaliki KM, Reliene R. Pomegranate Extract Alters Breast Cancer Stem Cell Properties in Association with Inhibition of Epithelial-to-Mesenchymal Transition. Nutr Cancer 2017; 69:1088-1098. [PMID: 28976208 DOI: 10.1080/01635581.2017.1359318] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Cancer stem cells (CSCs) have become an important target population in cancer therapy and prevention due to their ability to self-renew, initiate tumors, and resist therapy. We examined whether pomegranate extract (PE) alters characteristics of breast CSCs. Ability to grow as mammospheres is a hallmark of breast CSCs. PE inhibited mammosphere formation in two different cell lines, neoplastic mammary epithelial HMLER and breast cancer Hs578T. In addition, mammosphere-derived cells from PE treatment groups showed reduced mammosphere formation for at least two serial passages. These data indicate that PE inhibits CSC's ability to self-renew. In addition, incubation of mammospheres with PE reversed them into adherent cultures, indicating promotion of CSC differentiation. Epithelial-to-mesenchymal transition (EMT) is a key program in generating CSCs and maintaining their characteristics. Thus, we examined the effect of PE on EMT. PE reduced cell migration, a major feature of the EMT phenotype. In addition, PE downregulated genes involved in EMT, including the EMT-inducing transcription factor Twist family basic helix-loop-helix transcription factor 1 (TWIST1). This suggests that PE suppresses CSC characteristics in part due to inhibition of EMT. The ability of PE to suppress CSCs can be exploited in the prevention of breast cancer.
Collapse
Affiliation(s)
- Sameera Nallanthighal
- a Cancer Research Center , University at Albany, State University of New York , Rensselaer , New York , USA.,b Department of Biomedical Sciences , University at Albany, State University of New York , Albany , New York , USA
| | - Kristine M Elmaliki
- a Cancer Research Center , University at Albany, State University of New York , Rensselaer , New York , USA
| | - Ramune Reliene
- a Cancer Research Center , University at Albany, State University of New York , Rensselaer , New York , USA.,c Department of Environmental Health Sciences , University at Albany, State University of New York , Albany , New York , USA
| |
Collapse
|
12
|
Gentile LB, Nagamine MK, Biondi LR, Sanches DS, Toyota F, Giovani TM, de Jesus IP, da Fonseca IIM, Queiroz-Hazarbassanov N, Diaz BL, Salles Gomes CDOM, Dagli MLZ. Establishment of primary mixed cell cultures from spontaneous canine mammary tumors: Characterization of classic and new cancer-associated molecules. PLoS One 2017; 12:e0184228. [PMID: 28945747 PMCID: PMC5612463 DOI: 10.1371/journal.pone.0184228] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 08/21/2017] [Indexed: 11/21/2022] Open
Abstract
There are many factors which make canine cancer like cancer in humans. The occurrence of spontaneous mammary tumors in pet dogs, tumor genetics, molecular targets and exposure to the same environmental risk factors are among these factors. Therefore, the study of canine cancer can provide useful information to the oncology field. This study aimed to establish and characterize a panel of primary mixed cell cultures obtained from spontaneous canine mammary tumors. Eight established cell cultures obtained from one normal mammary gland, one complex adenoma, one mixed adenoma, two complex carcinomas and two mixed carcinomas were analyzed. The gene expression levels of classic molecular cancer players such as fibroblast growth factor receptor (FGFR) 2, breast cancer (BRCA) 1, BRCA2 and estrogen receptor (ESR) 1 were evaluated. For the first time, three orphan nuclear receptors, estrogen-related receptors (ERRs) α, β and γ were studied in canine mammary cancer. The highest expression level of ERRα was observed in complex carcinoma-derived cell culture, while the highest levels of ERRβ and γ were observed in cells derived from a mixed carcinoma. Meanwhile, complex carcinomas presented the highest levels of expression of ESR1, BRCA1 and FGFR2 among all samples. BRCA2 was found exclusively in complex adenoma. The transcription factor GATA3 had its highest levels in mixed carcinoma samples and its lowest levels in complex adenoma. Proliferation assays were also performed to evaluate the mixed cell cultures response to ER ligands, genistein and DES, both in normoxia and hypoxic conditions. Our results demonstrate that morphological and functional studies of primary mixed cell cultures derived from spontaneous canine mammary tumors are possible and provide valuable tool for the study of various stages of mammary cancer development.
Collapse
Affiliation(s)
- Luciana B. Gentile
- Laboratory of Experimental and Comparative Oncology, Department of Pathology, School of Veterinary Medicine and Animal Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Marcia K. Nagamine
- Laboratory of Experimental and Comparative Oncology, Department of Pathology, School of Veterinary Medicine and Animal Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Luiz R. Biondi
- Laboratory of Experimental and Comparative Oncology, Department of Pathology, School of Veterinary Medicine and Animal Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Daniel S. Sanches
- Laboratory of Experimental and Comparative Oncology, Department of Pathology, School of Veterinary Medicine and Animal Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Fábio Toyota
- Veterinary Hospital Cães e Gatos, Osasco, São Paulo, Brazil
| | - Tatiane M. Giovani
- Department of Pathology, School of Veterinary Medicine and Animal Sciences, University of São Paulo (USP), São Paulo, São Paulo, Brazil
| | - Isis P. de Jesus
- Laboratory of Experimental and Comparative Oncology, Department of Pathology, School of Veterinary Medicine and Animal Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Ivone I. M. da Fonseca
- Laboratory of Experimental and Comparative Oncology, Department of Pathology, School of Veterinary Medicine and Animal Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Nicolle Queiroz-Hazarbassanov
- Applied Pharmacology and Toxicology Laboratory, School of Veterinary Medicine and Animal Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Bruno L. Diaz
- Laboratory of Inflammation, Carlos Chagas Filho Biophysics Institute (IBCCF), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Cristina de O. Massoco Salles Gomes
- Applied Pharmacology and Toxicology Laboratory, School of Veterinary Medicine and Animal Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Maria Lucia Z. Dagli
- Laboratory of Experimental and Comparative Oncology, Department of Pathology, School of Veterinary Medicine and Animal Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| |
Collapse
|
13
|
Zhu YT, Pang SY, Luo Y, Chen W, Bao JM, Tan WL. A modified method by differential adhesion for enrichment of bladder cancer stem cells. Int Braz J Urol 2017; 42:817-24. [PMID: 27564296 PMCID: PMC5006781 DOI: 10.1590/s1677-5538.ibju.2015.0409] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 10/15/2015] [Indexed: 12/18/2022] Open
Abstract
Purpose: In a previous study the vaccine was effective against bladder cancer in a mouse model. However, a small portion of tumors regrew because the vaccine could not eliminate bladder cancer stem cells (CSCs). In this study, we showed a modified method for the isolation of bladder CSCs using a combination of differential adhesion method and serum-free culture medium (SFM) method. Materials and Methods: Trypsin-resistant cells and trypsin-sensitive cells were isolated from MB49, EJ and 5637 cells by a combination of differential adhesion method and SFM method. The CSCs characterizations of trypsin-resistant cells were verified by the flow cytometry, the western blotting, the quantitative polymerase chain reaction, the resistance to chemotherapy assay, the transwell assay, and the tumor xenograft formation assay. Results: Trypsin-resistant cells were isolated and identified in CSCs characters, with high expression of CSCs markers, higher resistance to chemotherapy, greater migration in vitro, and stronger tumorigenicity in vivo. Conclusion: Trypsin-resistant cells displayed specific CSCs properties. Our study showed trypsin-resistant cells were isolated successfully with a modified method using a combination of differential adhesion method and SFM method.
Collapse
Affiliation(s)
- Yong-Tong Zhu
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shi-Yu Pang
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yang Luo
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wei Chen
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ji-Ming Bao
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wan-Long Tan
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
14
|
Chen J, Kumar S. Biophysical Regulation of Cancer Stem/Initiating Cells: Implications for Disease Mechanisms and Translation. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2017; 1:87-95. [PMID: 29082354 DOI: 10.1016/j.cobme.2017.02.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cancer stem/initiating cells (CSCs) are a subset of tumor cells proposed to play privileged roles in seeding tumors and driving metastasis. CSCs have emerged as an increasingly important target of interest in cancer biology and therapy. Recent work has suggested that CSC maintenance and metastatic potential may be modulated by physical inputs within the tissue microenvironment, including interstitial pressure and extracellular matrix stiffness. Here we review recent progress in our understanding of CSC regulation by biophysical signals within the tumor microenvironment. While the mechanistic basis of this signaling remains incompletely understood, we discuss emerging evidence that mechanical inputs can epigenetically regulate CSC behavior and that some CSCs can evade mechanotransductive signals to more efficiently infiltrate tissue. We also describe efforts to leverage these findings to engineer culture platforms for the characterization of CSC mechanics for discovery and screening.
Collapse
Affiliation(s)
- Joseph Chen
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720
| | - Sanjay Kumar
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720
| |
Collapse
|
15
|
Lin MC, Chen SY, Tsai HM, He PL, Lin YC, Herschman H, Li HJ. PGE 2 /EP 4 Signaling Controls the Transfer of the Mammary Stem Cell State by Lipid Rafts in Extracellular Vesicles. Stem Cells 2016; 35:425-444. [PMID: 27506158 DOI: 10.1002/stem.2476] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 07/05/2016] [Accepted: 07/25/2016] [Indexed: 01/09/2023]
Abstract
Prostaglandin E2 (PGE2 )-initiated signaling contributes to stem cell homeostasis and regeneration. However, it is unclear how PGE2 signaling controls cell stemness. This study identifies a previously unknown mechanism by which PGE2 /prostaglandin E receptor 4 (EP4 ) signaling regulates multiple signaling pathways (e.g., PI3K/Akt signaling, TGFβ signaling, Wnt signaling, EGFR signaling) which maintain the basal mammary stem cell phenotype. A shift of basal mammary epithelial stem cells (MaSCs) from a mesenchymal/stem cell state to a non-basal-MaSC state occurs in response to prostaglandin E receptor 4 (EP4 ) antagonism. EP4 antagonists elicit release of signaling components, by controlling their trafficking into extracellular vesicles/exosomes in a lipid raft/caveolae-dependent manner. Consequently, EP4 antagonism indirectly inactivates, through induced extracellular vesicle/exosome release, pathways required for mammary epithelial stem cell homeostasis, e.g. canonical/noncanonical Wnt, TGFβ and PI3K/Akt pathways. EP4 antagonism causes signaling receptors and signaling components to shift from non-lipid raft fractions to lipid raft fractions, and to then be released in EP4 antagonist-induced extracellular vesicles/exosomes, resulting in the loss of the stem cell state by mammary epithelial stem cells. In contrast, luminal mammary epithelial cells can acquire basal stem cell properties following ingestion of EP4 antagonist-induced stem cell extracellular vesicles/exosomes, and can then form mammary glands. These findings demonstrate that PGE2 /EP4 signaling controls homeostasis of mammary epithelial stem cells through regulating extracellular vesicle/exosome release. Reprogramming of mammary epithelial cells can result from EP4 -mediated stem cell property transfer by extracellular vesicles/exosomes containing caveolae-associated proteins, between mammary basal and luminal epithelial cells. Stem Cells 2017;35:425-444.
Collapse
Affiliation(s)
- Meng-Chieh Lin
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Shih-Yin Chen
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Ho-Min Tsai
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Pei-Lin He
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Yen-Chun Lin
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan.,Department of Biochemistry, University of Washington, Seattle, Washington, USA
| | - Harvey Herschman
- Department of Molecular & Medical Pharmacology, University of California, Los Angeles, Los Angeles, California, USA.,Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, California, USA
| | - Hua-Jung Li
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan
| |
Collapse
|
16
|
Eyking A, Reis H, Frank M, Gerken G, Schmid KW, Cario E. MiR-205 and MiR-373 Are Associated with Aggressive Human Mucinous Colorectal Cancer. PLoS One 2016; 11:e0156871. [PMID: 27271572 PMCID: PMC4894642 DOI: 10.1371/journal.pone.0156871] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 05/20/2016] [Indexed: 02/07/2023] Open
Abstract
Mucinous adenocarcinoma (MAC) represents a distinct histopathological entity of colorectal cancer (CRC), which is associated with disease progression and poor prognosis. Here, we found that expression levels of miR-205 and miR-373 were specifically upregulated only in patients with mucinous colon cancers, but not in CRC that lack mucinous components. To investigate the effects of miR-205 and miR-373 on intestinal epithelial cell (IEC) biology by gain- and loss-of-function experiments in a proof-of-concept approach, we chose previously established in-vitro human Caco-2-based models of differentiated, non-invasive (expressing TLR4 wild-type; termed Caco-2[WT]) versus undifferentiated, invasive (expressing TLR4 mutant D299G; termed Caco-2[D299G]) IEC. Enterocyte-like Caco-2[WT] showed low levels of miR-205 and miR-373 expression, while both miRNAs were significantly upregulated in colorectal carcinoma-like Caco-2[D299G], thus resembling the miRNA expression pattern of paired normal versus tumor samples from MAC patients. Using stable transfection, we generated miR-205- or miR-373-expressing and miR-205- or miR-373-inhibiting subclones of these IEC lines. We found that introduction of miR-205 into Caco-2[WT] led to expansion of mucus-secreting goblet cell-like cells, which was associated with induction of KLF4, MUC2 and TGFβ1 expression. Activation of miR-205 in Caco-2[WT] induced chemoresistance, while inhibition of miR-205 in Caco-2[D299G] promoted chemosensitivity. Caco-2[WT] overexpressing miR-373 showed mitotic abnormalities and underwent morphologic changes (loss of epithelial polarity, cytoskeletal reorganization, and junctional disruption) associated with epithelial-mesenchymal transition and progression to inflammation-associated colonic carcinoma, which correlated with induction of phosphorylated STAT3 and N-CADHERIN expression. Functionally, introduction of miR-373 into Caco-2[WT] mediated loss of cell-cell adhesion and increased proliferation and invasion. Reversely, inhibition of miR-373 allowed mesenchymal IEC to regain epithelial properties, which correlated with absence of neoplastic progression. Using xenografts in mice demonstrated miR-373-mediated acceleration of malignant intestinal tumor growth. In conclusion, our results provide first evidence that miR-205 and miR-373 may differentially contribute to the aggressive phenotype of MAC in CRC.
Collapse
Affiliation(s)
- Annette Eyking
- Department of Gastroenterology and Hepatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Henning Reis
- Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Magdalena Frank
- Department of Gastroenterology and Hepatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Guido Gerken
- Department of Gastroenterology and Hepatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Kurt W. Schmid
- Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Elke Cario
- Department of Gastroenterology and Hepatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- * E-mail:
| |
Collapse
|
17
|
Ramena G, Yin Y, Yu Y, Walia V, Elble RC. CLCA2 Interactor EVA1 Is Required for Mammary Epithelial Cell Differentiation. PLoS One 2016; 11:e0147489. [PMID: 26930581 PMCID: PMC4773014 DOI: 10.1371/journal.pone.0147489] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 01/05/2016] [Indexed: 12/12/2022] Open
Abstract
CLCA2 is a p53-, p63-inducible transmembrane protein that is frequently downregulated in breast cancer. It is induced during differentiation of human mammary epithelial cells, and its knockdown causes epithelial-to-mesenchymal transition (EMT). To determine how CLCA2 promotes epithelial differentiation, we searched for interactors using membrane dihybrid screening. We discovered a strong interaction with the cell junctional protein EVA1 (Epithelial V-like Antigen 1) and confirmed it by co-immunoprecipitation. Like CLCA2, EVA1 is a type I transmembrane protein that is regulated by p53 and p63. It is thought to mediate homophilic cell-cell adhesion in diverse epithelial tissues. We found that EVA1 is frequently downregulated in breast tumors and breast cancer cell lines, especially those of mesenchymal phenotype. Moreover, knockdown of EVA1 in immortalized human mammary epithelial cells (HMEC) caused EMT, implying that EVA1 is essential for epithelial differentiation. Both EVA1 and CLCA2 co-localized with E-cadherin at cell-cell junctions. The interacting domains were delimited by deletion analysis, revealing the site of interaction to be the transmembrane segment (TMS). The primary sequence of the CLCA2 TMS was found to be conserved in CLCA2 orthologs throughout mammals, suggesting that its interaction with EVA1 co-evolved with the mammary gland. A screen for other junctional interactors revealed that CLCA2 was involved in two different complexes, one with EVA1 and ZO-1, the other with beta catenin. Overexpression of CLCA2 caused downregulation of beta catenin and beta catenin-activated genes. Thus, CLCA2 links a junctional adhesion molecule to cytosolic signaling proteins that modulate proliferation and differentiation. These results may explain how attenuation of CLCA2 causes EMT and why CLCA2 and EVA1 are frequently downregulated in metastatic breast cancer cell lines.
Collapse
Affiliation(s)
- Grace Ramena
- Dept of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University School of Medicine, Springfield, Illinois, 62794, United States of America
| | - Yufang Yin
- Dept of Pharmacology, Southern Illinois University School of Medicine, Springfield, Illinois, 62794, United States of America
| | - Yang Yu
- Dept of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University School of Medicine, Springfield, Illinois, 62794, United States of America
| | - Vijay Walia
- Laboratory of Cell and Developmental Signaling, National Cancer Institute-Frederick, Frederick, Maryland, 21702, United States of America
| | - Randolph C. Elble
- Dept of Pharmacology, Southern Illinois University School of Medicine, Springfield, Illinois, 62794, United States of America
- Simmons Cancer Institute, Southern Illinois University School of Medicine, Springfield, Illinois, 62794, United States of America
- * E-mail:
| |
Collapse
|
18
|
Homeostatic Signaling by Cell-Cell Junctions and Its Dysregulation during Cancer Progression. J Clin Med 2016; 5:jcm5020026. [PMID: 26901232 PMCID: PMC4773782 DOI: 10.3390/jcm5020026] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 02/05/2016] [Accepted: 02/05/2016] [Indexed: 12/16/2022] Open
Abstract
The transition of sessile epithelial cells to a migratory, mesenchymal phenotype is essential for metazoan development and tissue repair, but this program is exploited by tumor cells in order to escape the confines of the primary organ site, evade immunosurveillance, and resist chemo-radiation. In addition, epithelial-to-mesenchymal transition (EMT) confers stem-like properties that increase efficiency of colonization of distant organs. This review evaluates the role of cell–cell junctions in suppressing EMT and maintaining a quiescent epithelium. We discuss the conflicting data on junctional signaling in cancer and recent developments that resolve some of these conflicts. We focus on evidence from breast cancer, but include other organ sites where appropriate. Current and potential strategies for inhibition of EMT are discussed.
Collapse
|
19
|
Morata-Tarifa C, Jiménez G, García MA, Entrena JM, Griñán-Lisón C, Aguilera M, Picon-Ruiz M, Marchal JA. Low adherent cancer cell subpopulations are enriched in tumorigenic and metastatic epithelial-to-mesenchymal transition-induced cancer stem-like cells. Sci Rep 2016; 6:18772. [PMID: 26752044 PMCID: PMC4707518 DOI: 10.1038/srep18772] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 11/19/2015] [Indexed: 01/16/2023] Open
Abstract
Cancer stem cells are responsible for tumor progression, metastasis, therapy resistance and cancer recurrence, doing their identification and isolation of special relevance. Here we show that low adherent breast and colon cancer cells subpopulations have stem-like properties. Our results demonstrate that trypsin-sensitive (TS) breast and colon cancer cells subpopulations show increased ALDH activity, higher ability to exclude Hoechst 33342, enlarged proportion of cells with a cancer stem-like cell phenotype and are enriched in sphere- and colony-forming cells in vitro. Further studies in MDA-MB-231 breast cancer cells reveal that TS subpopulation expresses higher levels of SLUG, SNAIL, VIMENTIN and N-CADHERIN while show a lack of expression of E-CADHERIN and CLAUDIN, being this profile characteristic of the epithelial-to-mesenchymal transition (EMT). The TS subpopulation shows CXCL10, BMI-1 and OCT4 upregulation, differing also in the expression of several miRNAs involved in EMT and/or cell self-renewal such as miR-34a-5p, miR-34c-5p, miR-21-5p, miR-93-5p and miR-100-5p. Furthermore, in vivo studies in immunocompromised mice demonstrate that MDA-MB-231 TS cells form more and bigger xenograft tumors with shorter latency and have higher metastatic potential. In conclusion, this work presents a new, non-aggressive, easy, inexpensive and reproducible methodology to isolate prospectively cancer stem-like cells for subsequent biological and preclinical studies.
Collapse
Affiliation(s)
- Cynthia Morata-Tarifa
- Biopathology and Medicine Regenerative Institute (IBIMER), University of Granada, Granada, Spain
- Biosanitary Institute of Granada (ibs.GRANADA), University Hospitals of Granada-Univesity of Granada, Granada, Spain
| | - Gema Jiménez
- Biopathology and Medicine Regenerative Institute (IBIMER), University of Granada, Granada, Spain
- Biosanitary Institute of Granada (ibs.GRANADA), University Hospitals of Granada-Univesity of Granada, Granada, Spain
- Department of Human Anatomy and Embryology, University of Granada, Granada, Spain
| | - María A. García
- Biopathology and Medicine Regenerative Institute (IBIMER), University of Granada, Granada, Spain
- Biosanitary Institute of Granada (ibs.GRANADA), University Hospitals of Granada-Univesity of Granada, Granada, Spain
- Department of Oncology, University Hospital Virgen de las Nieves, Granada, Spain
| | - José M. Entrena
- Institute of Neuroscience, Biomedical Research Center, University of Granada, Granada, Spain
- Animal Behavior Research Unit, Scientific Instrumentation Center, University of Granada, Granada, Spain
| | - Carmen Griñán-Lisón
- Biopathology and Medicine Regenerative Institute (IBIMER), University of Granada, Granada, Spain
- Biosanitary Institute of Granada (ibs.GRANADA), University Hospitals of Granada-Univesity of Granada, Granada, Spain
| | - Margarita Aguilera
- Biosanitary Institute of Granada (ibs.GRANADA), University Hospitals of Granada-Univesity of Granada, Granada, Spain
- Department of Microbiology, University of Granada, Granada, Spain
| | - Manuel Picon-Ruiz
- Biopathology and Medicine Regenerative Institute (IBIMER), University of Granada, Granada, Spain
- Biosanitary Institute of Granada (ibs.GRANADA), University Hospitals of Granada-Univesity of Granada, Granada, Spain
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Juan A. Marchal
- Biopathology and Medicine Regenerative Institute (IBIMER), University of Granada, Granada, Spain
- Biosanitary Institute of Granada (ibs.GRANADA), University Hospitals of Granada-Univesity of Granada, Granada, Spain
- Department of Human Anatomy and Embryology, University of Granada, Granada, Spain
| |
Collapse
|
20
|
Translational regulation of inhibin βA by TGFβ via the RNA-binding protein hnRNP E1 enhances the invasiveness of epithelial-to-mesenchymal transitioned cells. Oncogene 2015; 35:1725-35. [PMID: 26096938 PMCID: PMC4688046 DOI: 10.1038/onc.2015.238] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 04/24/2015] [Accepted: 05/22/2015] [Indexed: 12/27/2022]
Abstract
The epithelial-to-mesenchymal transition (EMT) is a cellular process that functions during embryonic development and tissue regeneration, thought to be aberrantly activated in epithelial-derived cancer and play an important role in the process of metastasis. The TGFβ signaling pathway is a key inducer of EMT and we have elucidated a post-transcriptional mechanism by which TGFβ modulates expression of select transcripts via the RNA binding protein hnRNP E1 during EMT. One such transcript inhibin βA is a member of the TGFβ superfamily. Here, we show by polysome profiling that inhibin βA is translationally regulated by TGFβ via hnRNP E1. TGFβ treatment or knockdown of hnRNP E1 relieves silencing of the inhibin βA transcript, resulting in increased protein expression and secreted levels of the inhibin βA homodimer, activin A. Our data indicates that the translational up-regulation of inhibin βA enhances the migration and invasion of cells that have undergone an EMT and promotes cancer progression in vivo.
Collapse
|
21
|
Li SY, Sun R, Wang HX, Shen S, Liu Y, Du XJ, Zhu YH, Jun W. Combination therapy with epigenetic-targeted and chemotherapeutic drugs delivered by nanoparticles to enhance the chemotherapy response and overcome resistance by breast cancer stem cells. J Control Release 2015; 205:7-14. [PMID: 25445694 DOI: 10.1016/j.jconrel.2014.11.011] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Revised: 10/28/2014] [Accepted: 11/09/2014] [Indexed: 12/17/2022]
Abstract
Aberrant DNA hypermethylation is critical in the regulation of renewal and maintenance of cancer stem cells (CSCs), which represent targets for carcinogenic initiation by chemical and environmental agents. The administration of decitabine (DAC), which is a DNA hypermethylation inhibitor, is an attractive approach to enhancing the chemotherapeutic response and overcoming drug resistance by CSCs. In this study, we investigated whether low-dose DAC encapsulated in nanoparticles could be used to sensitize bulk breast cancer cells and CSCs to chemotherapy. In vitro studies revealed that treatment with nanoparticles loaded with low-dose DAC (NPDAC) combined with nanoparticles loaded with doxorubicin (NPDOX) better reduced the proportion of CSCs with high aldehyde dehydrogenase activity (ALDH(hi)) in the mammospheres of MDA-MB-231 cells, and better overcame the drug resistance by ALDH(hi) cells. Subsequently, systemic delivery of NPDAC significantly down-regulated the expression of DNMT1 and DNMT3b in a MB-MDA-231 xenograft murine model and induced increased caspase-9 expression, which contributed to the increased sensitivity of the bulk cancer cells and CSCs to NPDOX treatment. Importantly, the combined treatment of NPDAC and NPDOX resulted in the lowest proportion of ALDH(hi) CSCs and the highest proportion of apoptotic tumor cells, and the best tumor suppressive effects in inhibiting breast cancer growth.
Collapse
MESH Headings
- Aldehyde Dehydrogenase/genetics
- Aldehyde Dehydrogenase/metabolism
- Animals
- Antineoplastic Combined Chemotherapy Protocols/administration & dosage
- Apoptosis/drug effects
- Azacitidine/administration & dosage
- Azacitidine/analogs & derivatives
- Azacitidine/chemistry
- Breast Neoplasms/drug therapy
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Caspase 9/genetics
- Caspase 9/metabolism
- Cell Line, Tumor
- Chemistry, Pharmaceutical
- DNA (Cytosine-5-)-Methyltransferase 1
- DNA (Cytosine-5-)-Methyltransferases/genetics
- DNA (Cytosine-5-)-Methyltransferases/metabolism
- DNA Methylation/drug effects
- Decitabine
- Dose-Response Relationship, Drug
- Doxorubicin/administration & dosage
- Doxorubicin/chemistry
- Drug Carriers
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Epigenesis, Genetic/drug effects
- Female
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Mice, Inbred NOD
- Mice, SCID
- Nanomedicine
- Nanoparticles
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Polyesters/chemistry
- Polyethylene Glycols/chemistry
- Spheroids, Cellular
- Technology, Pharmaceutical/methods
- Time Factors
- Tumor Burden/drug effects
- Xenograft Model Antitumor Assays
- DNA Methyltransferase 3B
Collapse
Affiliation(s)
- Shi-Yong Li
- Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, PR China; The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, PR China
| | - Rong Sun
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, PR China
| | - Hong-Xia Wang
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, PR China
| | - Song Shen
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, PR China
| | - Yang Liu
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, PR China
| | - Xiao-Jiao Du
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, PR China
| | - Yan-Hua Zhu
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, PR China
| | - Wang Jun
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, PR China; Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, PR China; High Magnetic Field Laboratory of CAS, University of Science and Technology of China, Hefei, Anhui 230026, PR China.
| |
Collapse
|
22
|
|
23
|
Härmä V, Schukov HP, Happonen A, Ahonen I, Virtanen J, Siitari H, Åkerfelt M, Lötjönen J, Nees M. Quantification of dynamic morphological drug responses in 3D organotypic cell cultures by automated image analysis. PLoS One 2014; 9:e96426. [PMID: 24810913 PMCID: PMC4014501 DOI: 10.1371/journal.pone.0096426] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2013] [Accepted: 04/07/2014] [Indexed: 02/03/2023] Open
Abstract
Glandular epithelial cells differentiate into complex multicellular or acinar structures, when embedded in three-dimensional (3D) extracellular matrix. The spectrum of different multicellular morphologies formed in 3D is a sensitive indicator for the differentiation potential of normal, non-transformed cells compared to different stages of malignant progression. In addition, single cells or cell aggregates may actively invade the matrix, utilizing epithelial, mesenchymal or mixed modes of motility. Dynamic phenotypic changes involved in 3D tumor cell invasion are sensitive to specific small-molecule inhibitors that target the actin cytoskeleton. We have used a panel of inhibitors to demonstrate the power of automated image analysis as a phenotypic or morphometric readout in cell-based assays. We introduce a streamlined stand-alone software solution that supports large-scale high-content screens, based on complex and organotypic cultures. AMIDA (Automated Morphometric Image Data Analysis) allows quantitative measurements of large numbers of images and structures, with a multitude of different spheroid shapes, sizes, and textures. AMIDA supports an automated workflow, and can be combined with quality control and statistical tools for data interpretation and visualization. We have used a representative panel of 12 prostate and breast cancer lines that display a broad spectrum of different spheroid morphologies and modes of invasion, challenged by a library of 19 direct or indirect modulators of the actin cytoskeleton which induce systematic changes in spheroid morphology and differentiation versus invasion. These results were independently validated by 2D proliferation, apoptosis and cell motility assays. We identified three drugs that primarily attenuated the invasion and formation of invasive processes in 3D, without affecting proliferation or apoptosis. Two of these compounds block Rac signalling, one affects cellular cAMP/cGMP accumulation. Our approach supports the growing needs for user-friendly, straightforward solutions that facilitate large-scale, cell-based 3D assays in basic research, drug discovery, and target validation.
Collapse
Affiliation(s)
- Ville Härmä
- Medical Biotechnology Knowledge Centre, VTT Technical Research Centre of Finland, Turku, Finland
- Turku Centre for Biotechnology, University of Turku, Turku, Finland
| | - Hannu-Pekka Schukov
- Medical Biotechnology Knowledge Centre, VTT Technical Research Centre of Finland, Turku, Finland
- Turku Centre for Biotechnology, University of Turku, Turku, Finland
| | - Antti Happonen
- Department of Signal Processing, Tampere University of Technology, Tampere, Finland
| | - Ilmari Ahonen
- Department of Information Technology, University of Turku, Turku, Finland
| | - Johannes Virtanen
- Medical Biotechnology Knowledge Centre, VTT Technical Research Centre of Finland, Turku, Finland
- Turku Centre for Biotechnology, University of Turku, Turku, Finland
| | - Harri Siitari
- Medical Biotechnology Knowledge Centre, VTT Technical Research Centre of Finland, Turku, Finland
| | - Malin Åkerfelt
- Medical Biotechnology Knowledge Centre, VTT Technical Research Centre of Finland, Turku, Finland
- Turku Centre for Biotechnology, University of Turku, Turku, Finland
| | - Jyrki Lötjönen
- Knowledge Intensive Services, VTT Technical Research Centre of Finland, Tampere, Finland
| | - Matthias Nees
- Medical Biotechnology Knowledge Centre, VTT Technical Research Centre of Finland, Turku, Finland
- Turku Centre for Biotechnology, University of Turku, Turku, Finland
- * E-mail:
| |
Collapse
|
24
|
Loss of CLCA4 promotes epithelial-to-mesenchymal transition in breast cancer cells. PLoS One 2013; 8:e83943. [PMID: 24386311 PMCID: PMC3873418 DOI: 10.1371/journal.pone.0083943] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Accepted: 11/19/2013] [Indexed: 02/07/2023] Open
Abstract
The epithelial to mesenchymal transition (EMT) is a developmental program in which epithelial cells downregulate their cell-cell junctions, acquire spindle cell morphology and exhibit cellular motility. In breast cancer, EMT facilitates invasion of surrounding tissues and correlates closely with cancer metastasis and relapse. We found previously that the candidate tumor suppressor CLCA2 is expressed in differentiated, growth-arrested mammary epithelial cells but is downregulated during tumor progression and EMT. We further demonstrated that CLCA2 is a p53-inducible proliferation-inhibitor whose loss indicates an increased risk of metastasis. We show here that another member of the CLCA gene family, CLCA4, is expressed in mammary epithelial cells and is similarly downregulated in breast tumors and in breast cancer cell lines. Like CLCA2, the gene is stress-inducible, and ectopic expression inhibits colony formation. Transcriptional profiling studies revealed that CLCA4 and CLCA2 together are markers for mammary epithelial differentiation, and both are downregulated by TGF beta. Moreover, knockdown of CLCA4 in immortalized cells by shRNAs caused downregulation of epithelial marker E-cadherin and CLCA2, while mesenchymal markers N-cadherin, vimentin, and fibronectin were upregulated. Double knockdown of CLCA2 and CLCA4 enhanced the mesenchymal profile. These findings suggest that CLCA4 and CLCA2 play complementary but distinct roles in epithelial differentiation. Clinically, low expression of CLCA4 signaled lower relapse-free survival in basal and luminal B breast cancers.
Collapse
|
25
|
Duan JJ, Qiu W, Xu SL, Wang B, Ye XZ, Ping YF, Zhang X, Bian XW, Yu SC. Strategies for isolating and enriching cancer stem cells: well begun is half done. Stem Cells Dev 2013; 22:2221-39. [PMID: 23540661 DOI: 10.1089/scd.2012.0613] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cancer stem cells (CSCs) constitute a subpopulation of cancer cells that have the potential for self-renewal, multipotent differentiation, and tumorigenicity. Studies on CSC biology and CSC-targeted therapies depend on CSC isolation and/or enrichment methodologies. Scientists have conducted extensive research in this field since John Dick's group successfully isolated CSCs based on the expression of the CD34 and CD38 surface markers. Progress in CSC research has been greatly facilitated by the enrichment and isolation of these cells. In this review, we summarize the current strategies used in our and other laboratories for CSC isolation and enrichment, including methods based on stem cell surface markers, intracellular enzyme activity, the concentration of reactive oxygen species, the mitochondrial membrane potential, promoter-driven fluorescent protein expression, autofluorescence, suspension/adherent culture, cell division, the identification of side population cells, resistance to cytotoxic compounds or hypoxia, invasiveness/adhesion, immunoselection, and physical property. Although many challenges remain to be overcome, it is reasonable to believe that more reliable, efficient, and convenient methods will be developed in the near future.
Collapse
Affiliation(s)
- Jiang-Jie Duan
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Sarrio D, Franklin CK, Mackay A, Reis-Filho JS, Isacke CM. Epithelial and mesenchymal subpopulations within normal basal breast cell lines exhibit distinct stem cell/progenitor properties. Stem Cells 2012; 30:292-303. [PMID: 22102611 DOI: 10.1002/stem.791] [Citation(s) in RCA: 109] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
It has been proposed that epithelial-mesenchymal transition (EMT) in mammary epithelial cells and breast cancer cells generates stem cell features, and that the presence of EMT characteristics in claudin-low breast tumors reveals their origin in basal stem cells. It remains to be determined, however, whether EMT is an inherent property of normal basal stem cells, and if the presence of a mesenchymal-like phenotype is required for the maintenance of all their stem cell properties. We used nontumorigenic basal cell lines as models of normal stem cells/progenitors and demonstrate that these cell lines contain an epithelial subpopulation ("EpCAM+," epithelial cell adhesion molecule positive [EpCAM(pos)]/CD49f(high)) that spontaneously generates mesenchymal-like cells ("Fibros," EpCAM(neg)/CD49f(med/low)) through EMT. Importantly, stem cell/progenitor properties such as regenerative potential, high aldehyde dehydrogenase 1 activity, and formation of three-dimensional acini-like structures predominantly reside within EpCAM+ cells, while Fibros exhibit invasive behavior and mammosphere-forming ability. A gene expression profiling meta-analysis established that EpCAM+ cells show a luminal progenitor-like expression pattern, while Fibros most closely resemble stromal fibroblasts but not stem cells. Moreover, Fibros exhibit partial myoepithelial traits and strong similarities with claudin-low breast cancer cells. Finally, we demonstrate that Slug and Zeb1 EMT-inducers control the progenitor and mesenchymal-like phenotype in EpCAM+ cells and Fibros, respectively, by inhibiting luminal differentiation. In conclusion, nontumorigenic basal cell lines have intrinsic capacity for EMT, but a mesenchymal-like phenotype does not correlate with the acquisition of global stem cell/progenitor features. Based on our findings, we propose that EMT in normal basal cells and claudin-low breast cancers reflects aberrant/incomplete myoepithelial differentiation.
Collapse
Affiliation(s)
- David Sarrio
- Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, United Kingdom.
| | | | | | | | | |
Collapse
|
27
|
Walia V, Yu Y, Cao D, Sun M, McLean JR, Hollier BG, Cheng J, Mani SA, Rao K, Premkumar L, Elble RC. Loss of breast epithelial marker hCLCA2 promotes epithelial-to-mesenchymal transition and indicates higher risk of metastasis. Oncogene 2011; 31:2237-46. [PMID: 21909135 PMCID: PMC4154589 DOI: 10.1038/onc.2011.392] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Transition between epithelial and mesenchymal states is a feature of both normal development and tumor progression. We report that expression of chloride channel accessory protein hCLCA2 is a characteristic of epithelial differentiation in the immortalized MCF10A and HMLE models, while induction of EMT by cell dilution, TGFbeta, or mesenchymal transcription factors sharply reduces hCLCA2 levels. Attenuation of hCLCA2 expression by lentiviral shRNA caused cell overgrowth and focus formation, enhanced migration and invasion, and increased mammosphere formation in methylcellulose. These changes were accompanied by downregulation of E-cadherin and upregulation of mesenchymal markers such as vimentin and fibronectin. Moreover, hCLCA2 expression is greatly downregulated in breast cancer cells with a mesenchymal or claudin-low profile. These observations suggest that loss of hCLCA2 may promote metastasis. We find that higher-than-median expression of hCLCA2 is associated with a one-third lower rate of metastasis over an 18 year period among breast cancer patients compared to lower-than-median (n=344, unfiltered for subtype). Thus, hCLCA2 is required for epithelial differentiation, and its loss during tumor progression contributes to metastasis. Overexpression of hCLCA2 has been reported to inhibit cell proliferation and is accompanied by increases in chloride current at the plasma membrane and reduced intracellular pH (pHi). We found that knockdown cells have sharply reduced chloride current and higher pHi, both characteristics of tumor cells. These results suggest a mechanism for the effects on differentiation. Loss of hCLCA2 may allow escape from pHi homeostatic mechanisms, permitting the higher intracellular and lower extracellular pH that are characteristic of aggressive tumor cells.
Collapse
Affiliation(s)
- V Walia
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL 19629, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Tumour-microenvironmental interactions: paths to progression and targets for treatment. Semin Cancer Biol 2010; 20:128-38. [DOI: 10.1016/j.semcancer.2010.06.004] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2010] [Accepted: 06/24/2010] [Indexed: 01/01/2023]
|