51
|
Volmer LL, Önder CE, Volz B, Singh AR, Brucker SY, Engler T, Hartkopf AD, Koch A. Microfluidic Isolation of Disseminated Tumor Cells from the Bone Marrow of Breast Cancer Patients. Int J Mol Sci 2023; 24:13930. [PMID: 37762233 PMCID: PMC10531360 DOI: 10.3390/ijms241813930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/04/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023] Open
Abstract
Disseminated tumor cells (DTCs) in the bone marrow (BM) of breast cancer (BC) patients are putative precursors of metastatic disease, and their presence is associated with an adverse clinical outcome. To achieve the personalization of therapy on a clinical routine level, the characterization of DTCs and in vitro drug testing on DTCs are of great interest. Therefore, biobanking methods, as well as novel approaches to DTC isolation, need to be developed. In this study, we established a protocol for the biobanking of BM samples and evaluated a microfluidic-based separation system (Parsortix®) for the enrichment of cryopreserved DTCs. We were able to successfully isolate viable DTCs after the prior cryopreservation of BM samples. We calculated a significant increase of up to 90-fold in harvested DTCs with the proposed method compared to the current standard techniques, opening up new analysis possibilities for DTCs. Our advanced method further presents options for 3D DTC cultures, enabling the individualized testing of targeted therapies for BC patients. In conclusion, we present a novel approach for DTC enrichment, with possibilities for future clinical implications.
Collapse
Affiliation(s)
- Léa L. Volmer
- Research Institute for Women’s Health, University of Tübingen, 72076 Tübingen, Germany
- Department of Women’s Health, University of Tübingen, 72076 Tübingen, Germany
| | - Cansu E. Önder
- Research Institute for Women’s Health, University of Tübingen, 72076 Tübingen, Germany
| | - Barbara Volz
- Research Institute for Women’s Health, University of Tübingen, 72076 Tübingen, Germany
| | - Anjali R. Singh
- Research Institute for Women’s Health, University of Tübingen, 72076 Tübingen, Germany
| | - Sara Y. Brucker
- Department of Women’s Health, University of Tübingen, 72076 Tübingen, Germany
| | - Tobias Engler
- Department of Women’s Health, University of Tübingen, 72076 Tübingen, Germany
| | - Andreas D. Hartkopf
- Research Institute for Women’s Health, University of Tübingen, 72076 Tübingen, Germany
- Department of Women’s Health, University of Tübingen, 72076 Tübingen, Germany
| | - André Koch
- Research Institute for Women’s Health, University of Tübingen, 72076 Tübingen, Germany
| |
Collapse
|
52
|
Siatis KE, Giannopoulou E, Manou D, Sarantis P, Karamouzis MV, Raftopoulou S, Fasseas K, Alzahrani FM, Kalofonos HP, Theocharis AD. Resistance to hormone therapy in breast cancer cells promotes autophagy and EGFR signaling pathway. Am J Physiol Cell Physiol 2023; 325:C708-C720. [PMID: 37575061 PMCID: PMC10625825 DOI: 10.1152/ajpcell.00199.2023] [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: 05/09/2023] [Revised: 07/13/2023] [Accepted: 07/26/2023] [Indexed: 08/15/2023]
Abstract
Breast cancer is the leading cause of cancer deaths for women worldwide. Endocrine therapies represent the cornerstone for hormone-dependent breast cancer treatment. However, in many cases, endocrine resistance is induced with poor prognosis for patients. In the current study, we have developed MCF-7 cell lines resistant to fulvestrant (MCF-7Fulv) and tamoxifen (MCF-7Tam) aiming at investigating mechanisms underlying resistance. Both resistant cell lines exerted lower proliferation capacity in two-dimensional (2-D) cultures but retain estrogen receptor α (ERα) expression and proliferate independent of the presence of estrogens. The established cell lines tend to be more aggressive exhibiting advanced capacity to form colonies, increased expression of epidermal growth factor receptor (EGFR), human epidermal growth factor receptor 2 (HER2), and heterodimerization of ERBB family receptors and activation of EGFR downstream pathways like MEK/ERK1/2 and PI3K/AKT. Tyrosine kinase inhibitors tested against resistant MCF-7Fulv and MCF-7Tam cells showed moderate efficacy to inhibit cell proliferation, except for lapatinib, which concomitantly inhibits both EGFR and HER2 receptors and strongly reduced cell proliferation. Furthermore, increased autophagy was observed in resistant MCF-7Fulv and MCF-7Tam cells as shown by the presence of autophagosomes and increased Beclin-1 levels. The increased autophagy in resistant cells is not associated with increased apoptosis, suggesting a cytoprotective role for autophagy that may favor cells' survival and aggressiveness. Thus, by exploiting those underlying mechanisms, new targets could be established to overcome endocrine resistance.NEW & NOTEWORTHY The development of resistance to hormone therapy caused by both fulvestrant and tamoxifen promotes autophagy with concomitant apoptosis evasion, rendering cells capable of surviving and growing. The fact that resistance also triggers ERBB family signaling pathways, which are poorly inhibited by tyrosine kinase inhibitors might attribute to cells' aggressiveness. It is obvious that the development of endocrine therapy resistance involves a complex interplay between deregulated ERBB signaling and autophagy that may be considered in clinical practice.
Collapse
Affiliation(s)
- Konstantinos E Siatis
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Rio, Greece
- Clinical Oncology Laboratory, Division of Oncology, Department of Medicine, University of Patras, Rio, Greece
| | - Efstathia Giannopoulou
- Clinical Oncology Laboratory, Division of Oncology, Department of Medicine, University of Patras, Rio, Greece
| | - Dimitra Manou
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Rio, Greece
| | - Panagiotis Sarantis
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Michalis V Karamouzis
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Sofia Raftopoulou
- Electron Microscopy Laboratory, Faculty of Crop Production, Agricultural University of Athens, Athens, Greece
| | - Konstantinos Fasseas
- Electron Microscopy Laboratory, Faculty of Crop Production, Agricultural University of Athens, Athens, Greece
| | - Fatimah Mohammed Alzahrani
- Department of Chemistry, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Haralabos P Kalofonos
- Clinical Oncology Laboratory, Division of Oncology, Department of Medicine, University of Patras, Rio, Greece
| | - Achilleas D Theocharis
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Rio, Greece
- Department of Chemistry, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| |
Collapse
|
53
|
Collodet C, Blust K, Gkouma S, Ståhl E, Chen X, Hartman J, Hedhammar M. Development and characterization of a recombinant silk network for 3D culture of immortalized and fresh tumor-derived breast cancer cells. Bioeng Transl Med 2023; 8:e10537. [PMID: 37693069 PMCID: PMC10487315 DOI: 10.1002/btm2.10537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 04/05/2023] [Accepted: 04/12/2023] [Indexed: 09/12/2023] Open
Abstract
Traditional cancer models rely on 2D cell cultures or 3D spheroids, which fail to recapitulate cell-extracellular matrix (ECM) interactions, a key element of tumor development. Existing hydrogel-based 3D alternatives lack mechanical support for cell growth and often suffer from low reproducibility. Here we report a novel strategy to make 3D models of breast cancer using a tissue-like, well-defined network environment based on recombinant spider silk, functionalized with a cell adhesion motif from fibronectin (FN-silk). With this approach, the canonical cancer cells SK-BR-3, MCF-7, and MDA-MB-231, maintain their characteristic expression of markers (i.e., ERα, HER2, and PGR) while developing distinct morphology. Transcriptomic analyses demonstrate how culture in the FN-silk networks modulates the biological processes of cell adhesion and migration while affecting physiological events involved in malignancy, such as inflammation, remodeling of the ECM, and resistance to anticancer drugs. Finally, we show that integration in FN-silk networks promotes the viability of cells obtained from the superficial scraping of patients' breast tumors.
Collapse
Affiliation(s)
- Caterina Collodet
- Division of Protein TechnologySchool of Biotechnology, KTH Royal Institute of TechnologyStockholmSweden
| | - Kelly Blust
- Division of Protein TechnologySchool of Biotechnology, KTH Royal Institute of TechnologyStockholmSweden
| | - Savvini Gkouma
- Division of Protein TechnologySchool of Biotechnology, KTH Royal Institute of TechnologyStockholmSweden
| | - Emmy Ståhl
- Division of Protein TechnologySchool of Biotechnology, KTH Royal Institute of TechnologyStockholmSweden
| | - Xinsong Chen
- Department of Oncology‐PathologyKarolinska InstitutetStockholmSweden
| | - Johan Hartman
- Department of Oncology‐PathologyKarolinska InstitutetStockholmSweden
- Department of Clinical Pathology and Cancer DiagnosticsKarolinska University HospitalStockholmSweden
| | - My Hedhammar
- Division of Protein TechnologySchool of Biotechnology, KTH Royal Institute of TechnologyStockholmSweden
| |
Collapse
|
54
|
Palmiero M, Cantarosso I, di Blasio L, Monica V, Peracino B, Primo L, Puliafito A. Collective directional migration drives the formation of heteroclonal cancer cell clusters. Mol Oncol 2023; 17:1699-1725. [PMID: 36587372 PMCID: PMC10483614 DOI: 10.1002/1878-0261.13369] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 11/11/2022] [Accepted: 12/30/2022] [Indexed: 01/02/2023] Open
Abstract
Metastasisation occurs through the acquisition of invasive and survival capabilities that allow tumour cells to colonise distant sites. While the role of multicellular aggregates in cancer dissemination is acknowledged, the mechanisms that drive the formation of multiclonal cell aggregates are not fully elucidated. Here, we show that cancer cells of different tissue of origins can perform collective directional migration and can actively form heteroclonal aggregates in 3D, through a proliferation-independent mechanism. Coalescence of distant cell clusters is mediated by subcellular actin-rich protrusions and multicellular outgrowths that extend towards neighbouring aggregates. Coherently, perturbation of cytoskeletal dynamics impairs collective migration while myosin II activation is necessary for multicellular movements. We put forward the hypothesis that cluster attraction is mediated by secreted soluble factors. Such a hypothesis is consistent with the abrogation of aggregation by inhibition of PI3K/AKT/mTOR and MEK/ERK, the chemoattracting activity of conditioned culture media and with a wide screening of secreted proteins. Our results present a novel collective migration model and shed light on the mechanisms of formation of heteroclonal aggregates in cancer.
Collapse
Affiliation(s)
- Miriam Palmiero
- Candiolo Cancer Institute, FPO – IRCCSCandioloItaly
- Department of OncologyUniversity of TurinCandioloItaly
| | - Isabel Cantarosso
- Candiolo Cancer Institute, FPO – IRCCSCandioloItaly
- Department of OncologyUniversity of TurinCandioloItaly
| | - Laura di Blasio
- Candiolo Cancer Institute, FPO – IRCCSCandioloItaly
- Department of OncologyUniversity of TurinCandioloItaly
| | - Valentina Monica
- Candiolo Cancer Institute, FPO – IRCCSCandioloItaly
- Department of OncologyUniversity of TurinCandioloItaly
| | - Barbara Peracino
- Department of Clinical and Biological SciencesSan Luigi Hospital, University of TurinOrbassanoItaly
| | - Luca Primo
- Candiolo Cancer Institute, FPO – IRCCSCandioloItaly
- Department of OncologyUniversity of TurinCandioloItaly
| | - Alberto Puliafito
- Candiolo Cancer Institute, FPO – IRCCSCandioloItaly
- Department of OncologyUniversity of TurinCandioloItaly
| |
Collapse
|
55
|
Gandidzanwa S, Beukes N, Joseph SV, Janse Van Vuuren A, Mashazi P, Britton J, Kilian G, Roux S, Nyokong T, Lee ME, Frost CL, Tshentu ZR. The development of folate-functionalised palladium nanoparticles for folate receptor targeting in breast cancer cells. NANOTECHNOLOGY 2023; 34:465705. [PMID: 37527629 DOI: 10.1088/1361-6528/acec52] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 08/01/2023] [Indexed: 08/03/2023]
Abstract
Folate receptor-targeted therapy has excellent prospects for the treatment of breast cancer. A non-toxic concentration of folate-conjugated palladium-based nanoparticles was used to target the overexpressed folate receptor on breast cancer cells. The folate-conjugated nanoparticles were tailored to accumulate selectively in cancer cells relative to normal cells via the folate receptor. The MDA-MB-231, MDA-MB-468, MCF-7 breast cancer cell lines, and MCF-10A normal cell lines were used in the study. Qualitative and quantitative analysis of nanoparticle cellular uptake and accumulation was conducted using transmission electron microscopy and inductively coupled plasma-optical emission spectroscopy. The findings proved that folate-conjugated palladium nanoparticles successfully and preferentially accumulated in breast cancer cells. We conclude that folate-conjugated palladium nanoparticles can be potentially used to target breast cancer cells for radiopharmaceutical applications.
Collapse
Affiliation(s)
| | - Natasha Beukes
- Department of Biochemistry and Microbiology, Nelson Mandela University, Gqeberha 6001, South Africa
| | - Sinelizwi V Joseph
- Department of Chemistry, Nelson Mandela University, Gqeberha 6001, South Africa
| | - Arno Janse Van Vuuren
- Center for High Resolution Transmission Electron Microscopy, Nelson Mandela University, Gqeberha 6001, South Africa
| | - Philani Mashazi
- Department of Chemistry, Rhodes University, Makhanda 6140, South Africa
- Institute of Nanotechnology Innovation, Rhodes University, Makhanda 6140, South Africa
| | - Jonathan Britton
- Institute of Nanotechnology Innovation, Rhodes University, Makhanda 6140, South Africa
| | - Gareth Kilian
- Department of Pharmacy, Nelson Mandela University, Gqeberha 6001, South Africa
| | - Saartjie Roux
- Department of Human Physiology, Nelson Mandela University, Gqeberha 6001, South Africa
| | - Tebello Nyokong
- Institute of Nanotechnology Innovation, Rhodes University, Makhanda 6140, South Africa
| | - Michael E Lee
- Center for High Resolution Transmission Electron Microscopy, Nelson Mandela University, Gqeberha 6001, South Africa
| | - Carminita L Frost
- Department of Biochemistry and Microbiology, Nelson Mandela University, Gqeberha 6001, South Africa
| | - Zenixole R Tshentu
- Department of Chemistry, Nelson Mandela University, Gqeberha 6001, South Africa
| |
Collapse
|
56
|
Watzling M, Klaus L, Weidemeier T, Horder H, Ebert R, Blunk T, Bauer-Kreisel P. Three-Dimensional Breast Cancer Model to Investigate CCL5/CCR1 Expression Mediated by Direct Contact between Breast Cancer Cells and Adipose-Derived Stromal Cells or Adipocytes. Cancers (Basel) 2023; 15:3501. [PMID: 37444610 DOI: 10.3390/cancers15133501] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/26/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
The tumor microenvironment (TME) in breast cancer is determined by the complex crosstalk of cancer cells with adipose tissue-inherent cells such as adipose-derived stromal cells (ASCs) and adipocytes resulting from the local invasion of tumor cells in the mammary fat pad. This leads to heterotypic cellular contacts between these cell types. To adequately mimic the specific cell-to-cell interaction in an in vivo-like 3D environment, we developed a direct co-culture spheroid model using ASCs or differentiated adipocytes in combination with MDA-MB-231 or MCF-7 breast carcinoma cells. Co-spheroids were generated in a well-defined and reproducible manner in a high-throughput process. We compared the expression of the tumor-promoting chemokine CCL5 and its cognate receptors in these co-spheroids to indirect and direct standard 2D co-cultures. A marked up-regulation of CCL5 and in particular the receptor CCR1 with strict dependence on cell-cell contacts and culture dimensionality was evident. Furthermore, the impact of direct contacts between ASCs and tumor cells and the involvement of CCR1 in promoting tumor cell migration were demonstrated. Overall, these results show the importance of direct 3D co-culture models to better represent the complex tumor-stroma interaction in a tissue-like context. The unveiling of tumor-specific markers that are up-regulated upon direct cell-cell contact with neighboring stromal cells, as demonstrated in the 3D co-culture spheroids, may represent a promising strategy to find new targets for the diagnosis and treatment of invasive breast cancer.
Collapse
Affiliation(s)
- Martin Watzling
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Lorenz Klaus
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Tamara Weidemeier
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Hannes Horder
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Regina Ebert
- Department of Musculoskeletal Tissue Regeneration, Julius-Maximilians-Universität Würzburg, 97074 Würzburg, Germany
| | - Torsten Blunk
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Petra Bauer-Kreisel
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, University Hospital Würzburg, 97080 Würzburg, Germany
| |
Collapse
|
57
|
Irie N, Warita K, Tashiro J, Zhou Y, Ishikawa T, Oltvai ZN, Warita T. Expression of housekeeping genes varies depending on mevalonate pathway inhibition in cancer cells. Heliyon 2023; 9:e18017. [PMID: 37501994 PMCID: PMC10368838 DOI: 10.1016/j.heliyon.2023.e18017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 06/28/2023] [Accepted: 07/05/2023] [Indexed: 07/29/2023] Open
Abstract
Statins have anticancer effects and may be used as anticancer agents via drug repositioning. In reverse transcription-quantitative polymerase chain reaction (RT-qPCR) assays, the internal reference gene must not be affected by any experimental conditions. As statins exert a wide range of effects on cells by inhibiting the mevalonate pathway, it is possible that statin treatment might alter the expression of housekeeping genes used as internal reference genes, thereby misleading the assessment of obtained gene expression data. Here, we evaluated the expression stability of internal reference genes in atorvastatin-treated cancer cell lines. We treated both statin-sensitive and statin-resistant cancer cell lines with atorvastatin at seven different concentrations and performed RT-qPCR on 15 housekeeping genes whose expression stability was then assessed using five different algorithms. In both statin-sensitive and statin-resistant cancer cell lines, atorvastatin affected the expression of certain internal reference genes in a dose-dependent and cancer cell line-dependent manner; therefore, caution should be exercised when comparing target gene expression between cells. Our findings emphasize the importance of the validation of internal reference genes in gene expression analyses in drug treatment-based cancer research.
Collapse
Affiliation(s)
- Nanami Irie
- Graduate School of Science and Technology, Kwansei Gakuin University, 1 Gakuen Uegahara, Sanda, Hyogo 669‐1330, Japan
| | - Katsuhiko Warita
- Department of Veterinary Anatomy, School of Veterinary Medicine, Tottori University, 4-101 Koyama Minami, Tottori, Tottori 680-8553, Japan
| | - Jiro Tashiro
- Department of Veterinary Anatomy, School of Veterinary Medicine, Tottori University, 4-101 Koyama Minami, Tottori, Tottori 680-8553, Japan
| | - Yaxuan Zhou
- Graduate School of Science and Technology, Kwansei Gakuin University, 1 Gakuen Uegahara, Sanda, Hyogo 669‐1330, Japan
| | - Takuro Ishikawa
- Department of Anatomy, School of Medicine, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi 480-1195, Japan
| | - Zoltán N. Oltvai
- Department of Pathology and Laboratory Medicine, University of Rochester, 601 Elmwood Ave., Rochester, NY 14642, USA
| | - Tomoko Warita
- Department of Biomedical Sciences, School of Biological and Environmental Sciences, Kwansei Gakuin University, 1 Gakuen Uegahara, Sanda, Hyogo 669‐1330, Japan
| |
Collapse
|
58
|
Roohani S, Loskutov J, Heufelder J, Ehret F, Wedeken L, Regenbrecht M, Sauer R, Zips D, Denker A, Joussen AM, Regenbrecht CRA, Kaul D. Photon and Proton irradiation in Patient-derived, Three-Dimensional Soft Tissue Sarcoma Models. BMC Cancer 2023; 23:577. [PMID: 37349697 DOI: 10.1186/s12885-023-11013-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 05/25/2023] [Indexed: 06/24/2023] Open
Abstract
BACKGROUND Despite their heterogeneity, the current standard preoperative radiotherapy regimen for localized high-grade soft tissue sarcoma (STS) follows a one fits all approach for all STS subtypes. Sarcoma patient-derived three-dimensional cell culture models represent an innovative tool to overcome challenges in clinical research enabling reproducible subtype-specific research on STS. In this pilot study, we present our methodology and preliminary results using STS patient-derived 3D cell cultures that were exposed to different doses of photon and proton radiation. Our aim was: (i) to establish a reproducible method for irradiation of STS patient-derived 3D cell cultures and (ii) to explore the differences in tumor cell viability of two different STS subtypes exposed to increasing doses of photon and proton radiation at different time points. METHODS Two patient-derived cell cultures of untreated localized high-grade STS (an undifferentiated pleomorphic sarcoma (UPS) and a pleomorphic liposarcoma (PLS)) were exposed to a single fraction of photon or proton irradiation using doses of 0 Gy (sham irradiation), 2 Gy, 4 Gy, 8 Gy and 16 Gy. Cell viability was measured and compared to sham irradiation at two different time points (four and eight days after irradiation). RESULTS The proportion of viable tumor cells four days after photon irradiation for UPS vs. PLS were significantly different with 85% vs. 65% (4 Gy), 80% vs. 50% (8 Gy) and 70% vs. 35% (16 Gy). Proton irradiation led to similar diverging viability curves between UPS vs. PLS four days after irradiation with 90% vs. 75% (4 Gy), 85% vs. 45% (8 Gy) and 80% vs. 35% (16 Gy). Photon and proton radiation displayed only minor differences in cell-killing properties within each cell culture (UPS and PLS). The cell-killing effect of radiation sustained at eight days after irradiation in both cell cultures. CONCLUSIONS Pronounced differences in radiosensitivity are evident among UPS and PLS 3D patient-derived sarcoma cell cultures which may reflect the clinical heterogeneity. Photon and proton radiation showed similar dose-dependent cell-killing effectiveness in both 3D cell cultures. Patient-derived 3D STS cell cultures may represent a valuable tool to enable translational studies towards individualized subtype-specific radiotherapy in patients with STS.
Collapse
Affiliation(s)
- Siyer Roohani
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Radiation Oncology, Augustenburger Platz 1, 13353, Berlin, Germany.
- Charité - Universitätsmedizin Berlin, German Cancer Consortium (DKTK), partner site Berlin, and German Cancer Research Center (DKFZ), 69120, Berlin, Heidelberg, Germany.
| | - Jürgen Loskutov
- CELLphenomics GmbH, Robert-Rössle-Str. 10, 13125, Berlin, Germany
| | - Jens Heufelder
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, BerlinProtonen am Helmholtz-Zentrum Berlin, 14109, Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Ophthalmology, 12200, Berlin, Germany
| | - Felix Ehret
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Radiation Oncology, Augustenburger Platz 1, 13353, Berlin, Germany
- Charité - Universitätsmedizin Berlin, German Cancer Consortium (DKTK), partner site Berlin, and German Cancer Research Center (DKFZ), 69120, Berlin, Heidelberg, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Lena Wedeken
- CELLphenomics GmbH, Robert-Rössle-Str. 10, 13125, Berlin, Germany
| | - Manuela Regenbrecht
- CELLphenomics GmbH, Robert-Rössle-Str. 10, 13125, Berlin, Germany
- Helios Klinikum Berlin-Buch, Schwanebecker Chaussee 50, 13125, Berlin, Germany
- ASC Oncology GmbH, Robert-Rössle-Str. 10, 13125, Berlin, Germany
| | - Rica Sauer
- Institute of Pathology, Helios Klinikum Emil von Behring, Walterhöferstr. 11, 14165, Berlin, Germany
| | - Daniel Zips
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Radiation Oncology, Augustenburger Platz 1, 13353, Berlin, Germany
- Charité - Universitätsmedizin Berlin, German Cancer Consortium (DKTK), partner site Berlin, and German Cancer Research Center (DKFZ), 69120, Berlin, Heidelberg, Germany
| | - Andrea Denker
- Helmholtz-Zentrum Berlin für Materialien und Energie, 14109, Berlin, Germany
| | - Antonia M Joussen
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Ophthalmology, 12200, Berlin, Germany
| | - Christian R A Regenbrecht
- CELLphenomics GmbH, Robert-Rössle-Str. 10, 13125, Berlin, Germany
- ASC Oncology GmbH, Robert-Rössle-Str. 10, 13125, Berlin, Germany
- Institut für Pathologie, Universitätsmedizin Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany
| | - David Kaul
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Radiation Oncology, Augustenburger Platz 1, 13353, Berlin, Germany
- Charité - Universitätsmedizin Berlin, German Cancer Consortium (DKTK), partner site Berlin, and German Cancer Research Center (DKFZ), 69120, Berlin, Heidelberg, Germany
| |
Collapse
|
59
|
Jamil M, Mustafa IS, Sahul Hamid SB, Ahmed NM, Khazaalah TH, Godwin E, Ezra NS, Salah HN. Parameterisation and cellular evaluation of poly(ethylene) oxide-coated erbium oxide in MCF-7 cells as MRI diagnostic nanofibres. Colloids Surf B Biointerfaces 2023; 228:113423. [PMID: 37390675 DOI: 10.1016/j.colsurfb.2023.113423] [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: 03/29/2023] [Revised: 06/14/2023] [Accepted: 06/19/2023] [Indexed: 07/02/2023]
Abstract
The novelty of this work is the conjugation of poly(ethylene) oxide (PEO) with the erbium oxide (Er2O3) nanoparticles using the electrospinning technique. In this work, synthesised PEO-coated Er2O3 nanofibres were characterised and evaluated for their cytotoxicity to assess their potential use as diagnostic nanofibres for magnetic resonance imaging (MRI). PEO has significantly impacted nanoparticle conductivity due to its lower ionic conductivity at room temperature. The findings showed that the surface roughness was improved over the nanofiller loading, implying an improvement in cell attachment. The release profile performed for drug-controlling purposes has demonstrated a stable release after 30 min. Cellular response in MCF-7 cells showed high biocompatibility of the synthesised nanofibres. The cytotoxicity assay results showed that the diagnostic nanofibres had excellent biocompatibility, indicating the feasibility for diagnosis purposes. With excellent contrast performance, the PEO-coated Er2O3 nanofibres developed novel T2 and T1-T2 dual-mode MRI diagnostic nanofibres leading to better cancer diagnosis. In conclusion, this work has demonstrated that the conjugation of PEO-coated Er2O3 nanofibres improved the surface modification of the Er2O3 nanoparticles as a potential diagnostic agent. Using PEO in this study as a carrier or polymer matrix significantly influenced the biocompatibility and internalisation efficiency of the Er2O3 nanoparticles without triggering any morphological changes after treatment. This work has suggested permissible concentrations of PEO-coated Er2O3 nanofibres for diagnostic uses.
Collapse
Affiliation(s)
- Munirah Jamil
- School of Physics, Universiti Sains Malaysia, 11800 Gelugor, Penang, Malaysia.
| | | | - Shahrul Bariyah Sahul Hamid
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam, 13200 Kepala Batas, Penang, Malaysia
| | - Naser Mahmoud Ahmed
- School of Physics, Universiti Sains Malaysia, 11800 Gelugor, Penang, Malaysia; Department of Medical Instrumentation Engineering, Dijlah University College, Baghdad, Iraq
| | | | - Efenji Godwin
- School of Physics, Universiti Sains Malaysia, 11800 Gelugor, Penang, Malaysia; Department of Physics, Federal University Lokoja, P.M.B. 1154, Lokoja, Kogi State, Nigeria
| | - Nabasu Seth Ezra
- School of Physics, Universiti Sains Malaysia, 11800 Gelugor, Penang, Malaysia; Department of Physics, Faculty of Natural Science, Plateau State University, P.O. Box 2012 Bokkos, Jos Plateau State, Nigeria
| | - Hayder Naeem Salah
- School of Physics, Universiti Sains Malaysia, 11800 Gelugor, Penang, Malaysia; Science Department, College of Basic Education, Al-Muthanna University, Iraq
| |
Collapse
|
60
|
Godoy-Gallardo M, Merino-Gómez M, Mateos-Timoneda MA, Eckhard U, Gil FJ, Perez RA. Advanced Binary Guanosine and Guanosine 5'-Monophosphate Cell-Laden Hydrogels for Soft Tissue Reconstruction by 3D Bioprinting. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 37319328 DOI: 10.1021/acsami.2c23277] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Soft tissue defects or pathologies frequently necessitate the use of biomaterials that provide the volume required for subsequent vascularization and tissue formation as autrografts are not always a feasible alternative. Supramolecular hydrogels represent promising candidates because of their 3D structure, which resembles the native extracellular matrix, and their capacity to entrap and sustain living cells. Guanosine-based hydrogels have emerged as prime candidates in recent years since the nucleoside self-assembles into well-ordered structures like G-quadruplexes by coordinating K+ ions and π-π stacking, ultimately forming an extensive nanofibrillar network. However, such compositions were frequently inappropriate for 3D printing due to material spreading and low shape stability over time. Thus, the present work aimed to develop a binary cell-laden hydrogel capable of ensuring cell survival while providing enough stability to ensure scaffold biointegration during soft tissue reconstruction. For that purpose, a binary hydrogel made of guanosine and guanosine 5'-monophosphate was optimized, rat mesenchymal stem cells were entrapped, and the composition was bioprinted. To further increase stability, the printed structure was coated with hyperbranched polyethylenimine. Scanning electron microscopic studies demonstrated an extensive nanofibrillar network, indicating excellent G-quadruplex formation, and rheological analysis confirmed good printing and thixotropic qualities. Additionally, diffusion tests using fluorescein isothiocyanate labeled-dextran (70, 500, and 2000 kDa) showed that nutrients of various molecular weights may diffuse through the hydrogel scaffold. Finally, cells were evenly distributed throughout the printed scaffold, cell survival was 85% after 21 days, and lipid droplet formation was observed after 7 days under adipogenic conditions, indicating successful differentiation and proper cell functioning. To conclude, such hydrogels may enable the 3D bioprinting of customized scaffolds perfectly matching the respective soft tissue defect, thereby potentially improving the outcome of the tissue reconstruction intervention.
Collapse
Affiliation(s)
- Maria Godoy-Gallardo
- Bioengineering Institute of Technology (BIT), International University of Catalonia (UIC), Carrer de Josep Trueta, Sant Cugat del Vallès, Barcelona 08195, Spain
| | - Maria Merino-Gómez
- Bioengineering Institute of Technology (BIT), International University of Catalonia (UIC), Carrer de Josep Trueta, Sant Cugat del Vallès, Barcelona 08195, Spain
| | - Miguel A Mateos-Timoneda
- Bioengineering Institute of Technology (BIT), International University of Catalonia (UIC), Carrer de Josep Trueta, Sant Cugat del Vallès, Barcelona 08195, Spain
| | - Ulrich Eckhard
- Department of Structural and Molecular Biology, Molecular Biology Institute of Barcelona (IBMB), Higher Scientific Research Council (CSIC), Barcelona Science Park, Baldiri Reixac 15-21, Barcelona 08028, Spain
| | - F Javier Gil
- Bioengineering Institute of Technology (BIT), International University of Catalonia (UIC), Carrer de Josep Trueta, Sant Cugat del Vallès, Barcelona 08195, Spain
| | - Roman A Perez
- Bioengineering Institute of Technology (BIT), International University of Catalonia (UIC), Carrer de Josep Trueta, Sant Cugat del Vallès, Barcelona 08195, Spain
| |
Collapse
|
61
|
Neagu AN, Whitham D, Bruno P, Morrissiey H, Darie CA, Darie CC. Omics-Based Investigations of Breast Cancer. Molecules 2023; 28:4768. [PMID: 37375323 DOI: 10.3390/molecules28124768] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/08/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
Breast cancer (BC) is characterized by an extensive genotypic and phenotypic heterogeneity. In-depth investigations into the molecular bases of BC phenotypes, carcinogenesis, progression, and metastasis are necessary for accurate diagnoses, prognoses, and therapy assessments in predictive, precision, and personalized oncology. This review discusses both classic as well as several novel omics fields that are involved or should be used in modern BC investigations, which may be integrated as a holistic term, onco-breastomics. Rapid and recent advances in molecular profiling strategies and analytical techniques based on high-throughput sequencing and mass spectrometry (MS) development have generated large-scale multi-omics datasets, mainly emerging from the three "big omics", based on the central dogma of molecular biology: genomics, transcriptomics, and proteomics. Metabolomics-based approaches also reflect the dynamic response of BC cells to genetic modifications. Interactomics promotes a holistic view in BC research by constructing and characterizing protein-protein interaction (PPI) networks that provide a novel hypothesis for the pathophysiological processes involved in BC progression and subtyping. The emergence of new omics- and epiomics-based multidimensional approaches provide opportunities to gain insights into BC heterogeneity and its underlying mechanisms. The three main epiomics fields (epigenomics, epitranscriptomics, and epiproteomics) are focused on the epigenetic DNA changes, RNAs modifications, and posttranslational modifications (PTMs) affecting protein functions for an in-depth understanding of cancer cell proliferation, migration, and invasion. Novel omics fields, such as epichaperomics or epimetabolomics, could investigate the modifications in the interactome induced by stressors and provide PPI changes, as well as in metabolites, as drivers of BC-causing phenotypes. Over the last years, several proteomics-derived omics, such as matrisomics, exosomics, secretomics, kinomics, phosphoproteomics, or immunomics, provided valuable data for a deep understanding of dysregulated pathways in BC cells and their tumor microenvironment (TME) or tumor immune microenvironment (TIMW). Most of these omics datasets are still assessed individually using distinct approches and do not generate the desired and expected global-integrative knowledge with applications in clinical diagnostics. However, several hyphenated omics approaches, such as proteo-genomics, proteo-transcriptomics, and phosphoproteomics-exosomics are useful for the identification of putative BC biomarkers and therapeutic targets. To develop non-invasive diagnostic tests and to discover new biomarkers for BC, classic and novel omics-based strategies allow for significant advances in blood/plasma-based omics. Salivaomics, urinomics, and milkomics appear as integrative omics that may develop a high potential for early and non-invasive diagnoses in BC. Thus, the analysis of the tumor circulome is considered a novel frontier in liquid biopsy. Omics-based investigations have applications in BC modeling, as well as accurate BC classification and subtype characterization. The future in omics-based investigations of BC may be also focused on multi-omics single-cell analyses.
Collapse
Affiliation(s)
- Anca-Narcisa Neagu
- Laboratory of Animal Histology, Faculty of Biology, "Alexandru Ioan Cuza" University of Iasi, Carol I Bvd, No. 20A, 700505 Iasi, Romania
| | - Danielle Whitham
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, 8 Clarkson Avenue, Potsdam, NY 13699, USA
| | - Pathea Bruno
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, 8 Clarkson Avenue, Potsdam, NY 13699, USA
| | - Hailey Morrissiey
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, 8 Clarkson Avenue, Potsdam, NY 13699, USA
| | - Celeste A Darie
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, 8 Clarkson Avenue, Potsdam, NY 13699, USA
| | - Costel C Darie
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, 8 Clarkson Avenue, Potsdam, NY 13699, USA
| |
Collapse
|
62
|
BABAHAN C, ABDI ABGARMI S, SONUGÜR FG, ÖÇAL M, AKBULUT H. The effects of anti-PD-L1 monoclonal antibody on the expression of angiogenesis and invasion-related genes. Turk J Biol 2023; 47:262-275. [PMID: 38152616 PMCID: PMC10751090 DOI: 10.55730/1300-0152.2661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 08/31/2023] [Accepted: 06/07/2023] [Indexed: 12/29/2023] Open
Abstract
Background/aim The role of PD-L1 in regulating the immunosuppressive tumor microenvironment via its binding on PD-1 receptors is extensively studied. The PD-1/PD-L1 axis is a significant way of cancer immune escape, and PD-L1 expression on tumor cells is suggested as a predictive marker for anti-PD-1/PD-L1 monoclonal antibodies (MoAbs). However, the tumor-intrinsic role of PD-L1 is not known well. Therefore, we aimed to investigate the effects of anti-PD-L1 antibodies on the expression of angiogenesis and metastasis-related genes in tumor cells. Materials and methods The experiments were done with prostate cancer and melanoma cells with low PD-L1 expression (<5%) and prostate and breast cancer cells with high PD-L1 expression (>50%). The gene and protein expressions of VEGFA, E-cadherin, TGFβ1, EGFR, and bFGF in tumor cells were assayed at the 3 different doses of the anti-PD-L1 antibody. Results We found that VEGFA, E-cadherin and TGFβ1 expressions increased in PD-L1 high cells but decreased in PD-L1 low cells after anti-PD-L1 treatment. EGFR expression levels were variable in PD-L1 high cells, while decreased in PD-L1 low cells upon treatment. Also, the anti-PD-L1 antibody was found to increase bFGF expression in the prostate cancer cell line with high PD-L1 expression. Conclusion Our results suggest that the binding of PD-L1 on tumor cells by an anti-PD-L1 monoclonal antibody may affect tumor-intrinsic mechanisms. The activation of angiogenesis and metastasis-related pathways by anti-PD-L1 treatment in PD-L1 high tumors might be a tumor-promoting mechanism. The decrease of VEGFA, TGFβ1 and EGFR upon anti-PD-L1 treatment in PD-L1 low tumor cells provides a rationale for the use of those antibodies in PD-L1 low tumors.
Collapse
Affiliation(s)
- Cansu BABAHAN
- Ankara University Cancer Research Institute, Ankara,
Turkiye
| | | | | | - Müge ÖÇAL
- Ankara University Cancer Research Institute, Ankara,
Turkiye
| | - Hakan AKBULUT
- Ankara University Cancer Research Institute, Ankara,
Turkiye
- Department of Medical Oncology, School of Medicine, Ankara University, Ankara,
Turkiye
| |
Collapse
|
63
|
Tebon PJ, Wang B, Markowitz AL, Davarifar A, Tsai BL, Krawczuk P, Gonzalez AE, Sartini S, Murray GF, Nguyen HTL, Tavanaie N, Nguyen TL, Boutros PC, Teitell MA, Soragni A. Drug screening at single-organoid resolution via bioprinting and interferometry. Nat Commun 2023; 14:3168. [PMID: 37280220 PMCID: PMC10244450 DOI: 10.1038/s41467-023-38832-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 05/17/2023] [Indexed: 06/08/2023] Open
Abstract
High throughput drug screening is an established approach to investigate tumor biology and identify therapeutic leads. Traditional platforms use two-dimensional cultures which do not accurately reflect the biology of human tumors. More clinically relevant model systems such as three-dimensional tumor organoids can be difficult to scale and screen. Manually seeded organoids coupled to destructive endpoint assays allow for the characterization of treatment response, but do not capture transitory changes and intra-sample heterogeneity underlying clinically observed resistance to therapy. We present a pipeline to generate bioprinted tumor organoids linked to label-free, time-resolved imaging via high-speed live cell interferometry (HSLCI) and machine learning-based quantitation of individual organoids. Bioprinting cells gives rise to 3D structures with unaltered tumor histology and gene expression profiles. HSLCI imaging in tandem with machine learning-based segmentation and classification tools enables accurate, label-free parallel mass measurements for thousands of organoids. We demonstrate that this strategy identifies organoids transiently or persistently sensitive or resistant to specific therapies, information that could be used to guide rapid therapy selection.
Collapse
Affiliation(s)
- Peyton J Tebon
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA, USA
| | - Bowen Wang
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA
- Department of Pathology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Alexander L Markowitz
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA, USA
- Department of Human Genetics, University of California Los Angeles, Los Angeles, CA, USA
- Institute for Precision Health, University of California Los Angeles, Los Angeles, CA, USA
| | - Ardalan Davarifar
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Department of Human Genetics, University of California Los Angeles, Los Angeles, CA, USA
| | - Brandon L Tsai
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA, USA
- Department of Human Genetics, University of California Los Angeles, Los Angeles, CA, USA
- Institute for Precision Health, University of California Los Angeles, Los Angeles, CA, USA
| | - Patrycja Krawczuk
- Information Sciences Institute, University of Southern California, Marina Del Rey, CA, USA
| | - Alfredo E Gonzalez
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA, USA
- Department of Human Genetics, University of California Los Angeles, Los Angeles, CA, USA
- Institute for Precision Health, University of California Los Angeles, Los Angeles, CA, USA
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA, USA
| | - Sara Sartini
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Graeme F Murray
- Department of Physics, Virginia Commonwealth University, Richmond, VA, USA
| | - Huyen Thi Lam Nguyen
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Nasrin Tavanaie
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Thang L Nguyen
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA
- Department of Pathology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Paul C Boutros
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA, USA
- Department of Human Genetics, University of California Los Angeles, Los Angeles, CA, USA
- Institute for Precision Health, University of California Los Angeles, Los Angeles, CA, USA
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA, USA
- California NanoSystems Institute, University of California Los Angeles, Los Angeles, CA, USA
- Department of Urology, University of California Los Angeles, Los Angeles, CA, USA
| | - Michael A Teitell
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA.
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA, USA.
- Department of Pathology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA, USA.
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA, USA.
- California NanoSystems Institute, University of California Los Angeles, Los Angeles, CA, USA.
- Department of Pediatrics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
| | - Alice Soragni
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA, USA.
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA, USA.
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA, USA.
- California NanoSystems Institute, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
64
|
Madhavan M, Jaiswal D, Karlberg S, Duggan A, Almarshad HA, Claffey KP, Hoshino K. Electron microscopy imaging and mechanical characterization of T47D multicellular tumor spheroids-Older spheroids reduce interstitial space and become stiffer. PLoS One 2023; 18:e0286291. [PMID: 37228139 DOI: 10.1371/journal.pone.0286291] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 05/15/2023] [Indexed: 05/27/2023] Open
Abstract
Multicellular cancer spheroids are an in vitro tissue model that mimics the three-dimensional microenvironment. As spheroids grow, they develop the gradients of oxygen, nutrients, and catabolites, affecting crucial tumor characteristics such as proliferation and treatment responses. The measurement of spheroid stiffness provides a quantitative measure to evaluate such structural changes over time. In this report, we measured the stiffness of size-matched day 5 and day 20 tumor spheroids using a custom-built microscale force sensor and conducted transmission electron microscopy (TEM) imaging to compare the internal structures. We found that older spheroids reduce interstitial spaces in the core region and became significantly stiffer. The measured elastic moduli were 260±100 and 680±150 Pa, for day 5 and day 20 spheroids, respectively. The day 20 spheroids showed an optically dark region in the center. Analyzing the high-resolution TEM images of spheroid middle sections across the diameter showed that the cells in the inner region of the day 20 spheroids are significantly larger and more closely packed than those in the outer regions. On the other hand, the day 5 spheroids did not show a significant difference between the inner and outer regions. The observed reduction of the interstitial space may be one factor that contributes to stiffer older spheroids.
Collapse
Affiliation(s)
- Mathangi Madhavan
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut, United States of America
| | - Devina Jaiswal
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut, United States of America
- Department of Biomedical Engineering, Western New England University, Springfield, Massachusetts, United States of America
| | - Sarah Karlberg
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut, United States of America
| | - Alexis Duggan
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut, United States of America
| | - Hassan A Almarshad
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut, United States of America
- College of Applied Medical Sciences, Al Jouf University, Sakakah, Al Jawf, Saudi Arabia
| | - Kevin P Claffey
- Department of Cell Biology, University of Connecticut Health Center, Farmington, Connecticut, United States of America
| | - Kazunori Hoshino
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut, United States of America
| |
Collapse
|
65
|
Tong Y, Qi Y, Xiong G, Li J, Scott TL, Chen J, He D, Li L, Wang C, Lane AN, Xu R. The PLOD2/succinate axis regulates the epithelial-mesenchymal plasticity and cancer cell stemness. Proc Natl Acad Sci U S A 2023; 120:e2214942120. [PMID: 37155842 PMCID: PMC10194013 DOI: 10.1073/pnas.2214942120] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 03/08/2023] [Indexed: 05/10/2023] Open
Abstract
Aberrant accumulation of succinate has been detected in many cancers. However, the cellular function and regulation of succinate in cancer progression is not completely understood. Using stable isotope-resolved metabolomics analysis, we showed that the epithelial mesenchymal transition (EMT) was associated with profound changes in metabolites, including elevation of cytoplasmic succinate levels. The treatment with cell-permeable succinate induced mesenchymal phenotypes in mammary epithelial cells and enhanced cancer cell stemness. Chromatin immunoprecipitation and sequence analysis showed that elevated cytoplasmic succinate levels were sufficient to reduce global 5-hydroxymethylcytosinene (5hmC) accumulation and induce transcriptional repression of EMT-related genes. We showed that expression of procollagen-lysine,2-oxoglutarate 5-dioxygenase 2 (PLOD2) was associated with elevation of cytoplasmic succinate during the EMT process. Silencing of PLOD2 expression in breast cancer cells reduced succinate levels and inhibited cancer cell mesenchymal phenotypes and stemness, which was accompanied by elevated 5hmC levels in chromatin. Importantly, exogenous succinate rescued cancer cell stemness and 5hmC levels in PLOD2-silenced cells, suggesting that PLOD2 promotes cancer progression at least partially through succinate. These results reveal the previously unidentified function of succinate in enhancing cancer cell plasticity and stemness.
Collapse
Affiliation(s)
- Yuxin Tong
- Markey Cancer Center, University of Kentucky, Lexington, KY40536
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY40536
| | - Yifei Qi
- Markey Cancer Center, University of Kentucky, Lexington, KY40536
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY40536
| | - Gaofeng Xiong
- Markey Cancer Center, University of Kentucky, Lexington, KY40536
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY40536
| | - Junyan Li
- Markey Cancer Center, University of Kentucky, Lexington, KY40536
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY40536
| | - Timothy L. Scott
- Center for Environmental and Systems Biochemistry, University of Kentucky, Lexington, KY40536
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY40536
| | - Jie Chen
- Markey Cancer Center, University of Kentucky, Lexington, KY40536
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY40536
| | - Daheng He
- Markey Cancer Center, University of Kentucky, Lexington, KY40536
| | - Linzhang Li
- Markey Cancer Center, University of Kentucky, Lexington, KY40536
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY40536
| | - Chi Wang
- Markey Cancer Center, University of Kentucky, Lexington, KY40536
| | - Andrew N. Lane
- Center for Environmental and Systems Biochemistry, University of Kentucky, Lexington, KY40536
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY40536
| | - Ren Xu
- Markey Cancer Center, University of Kentucky, Lexington, KY40536
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY40536
| |
Collapse
|
66
|
Raik S, Sharma P, Kumar S, Rattan V, Das A, Kumar N, Srinivasan R, Bhattacharyya S. Three-dimensional spheroid culture of dental pulp-derived stromal cells enhance their biological and regenerative properties for potential therapeutic applications. Int J Biochem Cell Biol 2023; 160:106422. [PMID: 37172928 DOI: 10.1016/j.biocel.2023.106422] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 04/19/2023] [Accepted: 05/09/2023] [Indexed: 05/15/2023]
Abstract
Mesenchymal stem/stromal cell (MSC) spheroids generated in a three-dimensional (3D) culture system serve as a surrogate model that maintain stem cell characteristics since these mimic the in vivo behavior of cells and tissue more closely. Our study involved a detailed characterization of the spheroids generated in ultra-low attachment flasks. The spheroids were evaluated and compared for their morphology, structural integrity, viability, proliferation, biocomponents, stem cell phenotype and differentiation abilities with monolayer culture derived cells (2D culture). The in-vivo therapeutic efficacy of DPSCs derived from 2D and 3D culture was also assessed by transplanting them in an animal model of the critical-sized calvarial defect. DPSCs formed compact and well-organized multicellular spheroids when cultured in ultra-low attachment condition with superior stemness, differentiation, and regenerative abilities than monolayer cells. They maintained lower proliferative state and showed marked difference in the cellular biocomponents such as lipid, amide and nucleic acid between DPSCs from 2D and 3D cultures. The scaffold-free 3D culture efficiently preserves DPSCs intrinsic properties and functionality by maintaining them in the state close to the native tissues. The scaffold free 3D culture methods allow easy collection of a large number of multicellular spheroids of DPSCs and therefore, this can be adopted as a feasible and efficient method of generating robust spheroids for various in-vitro and in-vivo therapeutic applications.
Collapse
Affiliation(s)
- Shalini Raik
- Department of Biophysics, Post Graduate Institution of Medical Education and Research (PGIMER), Chandigarh, India
| | - Prakshi Sharma
- Department of Biophysics, Post Graduate Institution of Medical Education and Research (PGIMER), Chandigarh, India
| | - Saroj Kumar
- Department of Mechanical Engineering, Indian Institute of Technology Ropar, Rupnagar, Punjab, India
| | - Vidya Rattan
- Unit of oral and maxillofacial surgery, Department of Oral Health Sciences, PGIMER, Chandigarh, India
| | - Ashim Das
- Department of Histopathology, Post Graduate Institution of Medical Education and Research (PGIMER), Chandigarh, India
| | - Navin Kumar
- Department of Mechanical Engineering, Indian Institute of Technology Ropar, Rupnagar, Punjab, India
| | - Radhika Srinivasan
- Department of Cytology and Gynecologic Pathology, Post Graduate Institution of Medical Education and Research (PGIMER), Chandigarh, India
| | - Shalmoli Bhattacharyya
- Department of Biophysics, Post Graduate Institution of Medical Education and Research (PGIMER), Chandigarh, India.
| |
Collapse
|
67
|
Bjørnstad OV, Carrasco M, Finne K, Winge I, Askeland C, Arnes JB, Knutsvik G, Kleftogiannis D, Paulo JA, Akslen LA, Vethe H. Global and single-cell proteomics view of the co-evolution between neural progenitors and breast cancer cells in a co-culture model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.03.539050. [PMID: 37205344 PMCID: PMC10187147 DOI: 10.1101/2023.05.03.539050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Tumor neurogenesis, a process by which new nerves invade tumors, is a growing area of interest in cancer research. Nerve presence has been linked to aggressive features of various solid tumors, including breast and prostate cancer. A recent study suggested that the tumor microenvironment may influence cancer progression through recruitment of neural progenitor cells from the central nervous system. However, the presence of neural progenitors in human breast tumors has not been reported. Here, we investigate the presence of Doublecortin (DCX) and Neurofilament-Light (NFL) co-expressing (DCX+/NFL+) cells in patient breast cancer tissue using Imaging Mass Cytometry. To map the interaction between breast cancer cells and neural progenitor cells further, we created an in vitro model mimicking breast cancer innervation, and characterized using mass spectrometry-based proteomics on the two cell types as they co- evolved in co-culture. Our results indicate stromal presence of DCX+/NFL+ cells in breast tumor tissue from a cohort of 107 patient cases, and that neural interaction contribute to drive a more aggressive breast cancer phenotype in our co-culture models. Our results support that neural involvement plays an active role in breast cancer and warrants further studies on the interaction between nervous system and breast cancer progression.
Collapse
|
68
|
Hu Q, Hu X, Shi Y, Liang L, Zhu J, Zhao S, Wang Y, Wu Z, Wang F, Zhou F, Yang Y. Heterogeneous tissue construction by on-demand bubble-assisted acoustic patterning. LAB ON A CHIP 2023; 23:2206-2216. [PMID: 37006165 DOI: 10.1039/d3lc00122a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Highly heterogeneous structures are closely related to the realization of the tissue functions of living organisms. However, precisely controlling the assembly of heterogeneous structures is still a crucial challenge. This work presents an on-demand bubble-assisted acoustic method for active cell patterning to achieve high-precision heterogeneous structures. Active cell patterning is achieved by the combined effect of acoustic radiation forces and microstreaming around oscillating bubble arrays. On-demand bubble arrays allow flexible construction of cell patterns with a precision of up to 45 μm. As a typical example, the in vitro model of hepatic lobules, composed of patterned endothelial cells and hepatic parenchymal cells, was constructed and cultured for 5 days. The good performance of urea and albumin secretion, enzymatic activity and good proliferation of both cells prove the feasibility of this technique. Overall, this bubble-assisted acoustic approach provides a simple and efficient strategy for on-demand large-area tissue construction, with considerable potential for different tissue model fabrication.
Collapse
Affiliation(s)
- Qinghao Hu
- Department of Clinical Laboratory, Institute of Medicine and Physics, Renmin Hospital, Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics & Technology, Wuhan University, Wuhan 430072, People's Republic of China.
- Shenzhen Research Institute, Wuhan University, Shenzhen 518000, People's Republic of China
| | - Xuejia Hu
- Department of Electronic Engineering, School of Electronic Science and Engineering, Xiamen University, Xiamen 361005, People's Republic of China
| | - Yang Shi
- Institute of Nanophotonics, Jinan University, Guangzhou 510632, China
| | - Li Liang
- Department of Clinical Laboratory, Institute of Medicine and Physics, Renmin Hospital, Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics & Technology, Wuhan University, Wuhan 430072, People's Republic of China.
- Shenzhen Research Institute, Wuhan University, Shenzhen 518000, People's Republic of China
| | - Jiaomeng Zhu
- Department of Clinical Laboratory, Institute of Medicine and Physics, Renmin Hospital, Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics & Technology, Wuhan University, Wuhan 430072, People's Republic of China.
- Shenzhen Research Institute, Wuhan University, Shenzhen 518000, People's Republic of China
| | - Shukun Zhao
- Department of Clinical Laboratory, Institute of Medicine and Physics, Renmin Hospital, Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics & Technology, Wuhan University, Wuhan 430072, People's Republic of China.
- Shenzhen Research Institute, Wuhan University, Shenzhen 518000, People's Republic of China
| | - Yifan Wang
- Department of Clinical Laboratory, Institute of Medicine and Physics, Renmin Hospital, Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics & Technology, Wuhan University, Wuhan 430072, People's Republic of China.
- Shenzhen Research Institute, Wuhan University, Shenzhen 518000, People's Republic of China
| | - Zezheng Wu
- Department of Clinical Laboratory, Institute of Medicine and Physics, Renmin Hospital, Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics & Technology, Wuhan University, Wuhan 430072, People's Republic of China.
| | - Fubing Wang
- Department of Laboratory Medicine, Zhongnan Hospital, Wuhan University, Wuhan 430071, China
| | - Fuling Zhou
- Department of Hematology, Zhongnan Hospital, Wuhan University, Wuhan 430071, China
| | - Yi Yang
- Department of Clinical Laboratory, Institute of Medicine and Physics, Renmin Hospital, Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics & Technology, Wuhan University, Wuhan 430072, People's Republic of China.
- Shenzhen Research Institute, Wuhan University, Shenzhen 518000, People's Republic of China
| |
Collapse
|
69
|
Esser L, Springer R, Dreissen G, Lövenich L, Konrad J, Hampe N, Merkel R, Hoffmann B, Noetzel E. Elastomeric Pillar Cages Modulate Actomyosin Contractility of Epithelial Microtissues by Substrate Stiffness and Topography. Cells 2023; 12:cells12091256. [PMID: 37174659 PMCID: PMC10177551 DOI: 10.3390/cells12091256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 04/19/2023] [Accepted: 04/21/2023] [Indexed: 05/15/2023] Open
Abstract
Cell contractility regulates epithelial tissue geometry development and homeostasis. The underlying mechanobiological regulation circuits are poorly understood and experimentally challenging. We developed an elastomeric pillar cage (EPC) array to quantify cell contractility as a mechanoresponse of epithelial microtissues to substrate stiffness and topography. The spatially confined EPC geometry consisted of 24 circularly arranged slender pillars (1.2 MPa, height: 50 µm; diameter: 10 µm, distance: 5 µm). These high-aspect-ratio pillars were confined at both ends by planar substrates with different stiffness (0.15-1.2 MPa). Analytical modeling and finite elements simulation retrieved cell forces from pillar displacements. For evaluation, highly contractile myofibroblasts and cardiomyocytes were assessed to demonstrate that the EPC device can resolve static and dynamic cellular force modes. Human breast (MCF10A) and skin (HaCaT) cells grew as adherence junction-stabilized 3D microtissues within the EPC geometry. Planar substrate areas triggered the spread of monolayered clusters with substrate stiffness-dependent actin stress fiber (SF)-formation and substantial single-cell actomyosin contractility (150-200 nN). Within the same continuous microtissues, the pillar-ring topography induced the growth of bilayered cell tubes. The low effective pillar stiffness overwrote cellular sensing of the high substrate stiffness and induced SF-lacking roundish cell shapes with extremely low cortical actin tension (11-15 nN). This work introduced a versatile biophysical tool to explore mechanobiological regulation circuits driving low- and high-tensional states during microtissue development and homeostasis. EPC arrays facilitate simultaneously analyzing the impact of planar substrate stiffness and topography on microtissue contractility, hence microtissue geometry and function.
Collapse
Affiliation(s)
- Lisann Esser
- Institute of Biological Information Processing 2 (IBI-2): Mechanobiology, Forschungszentrum Jülich, 52428 Jülich, Germany
| | - Ronald Springer
- Institute of Biological Information Processing 2 (IBI-2): Mechanobiology, Forschungszentrum Jülich, 52428 Jülich, Germany
| | - Georg Dreissen
- Institute of Biological Information Processing 2 (IBI-2): Mechanobiology, Forschungszentrum Jülich, 52428 Jülich, Germany
| | - Lukas Lövenich
- Institute of Biological Information Processing 2 (IBI-2): Mechanobiology, Forschungszentrum Jülich, 52428 Jülich, Germany
| | - Jens Konrad
- Institute of Biological Information Processing 2 (IBI-2): Mechanobiology, Forschungszentrum Jülich, 52428 Jülich, Germany
| | - Nico Hampe
- Institute of Biological Information Processing 2 (IBI-2): Mechanobiology, Forschungszentrum Jülich, 52428 Jülich, Germany
| | - Rudolf Merkel
- Institute of Biological Information Processing 2 (IBI-2): Mechanobiology, Forschungszentrum Jülich, 52428 Jülich, Germany
| | - Bernd Hoffmann
- Institute of Biological Information Processing 2 (IBI-2): Mechanobiology, Forschungszentrum Jülich, 52428 Jülich, Germany
| | - Erik Noetzel
- Institute of Biological Information Processing 2 (IBI-2): Mechanobiology, Forschungszentrum Jülich, 52428 Jülich, Germany
| |
Collapse
|
70
|
Zhou C, Wu Y, Wang Z, Liu Y, Yu J, Wang W, Chen S, Wu W, Wang J, Qian G, He A. Standardization of organoid culture in cancer research. Cancer Med 2023. [PMID: 37081739 DOI: 10.1002/cam4.5943] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 03/24/2023] [Accepted: 04/01/2023] [Indexed: 04/22/2023] Open
Abstract
Establishing a valid in vitro model to represent tumor heterogeneity and biology is critical but challenging. Tumor organoids are self-assembled three-dimensional cell clusters which are of great significance for recapitulating the histopathological, genetic, and phenotypic characteristics of primary tissues. The organoid has emerged as an attractive in vitro platform for tumor biology research and high-throughput drug screening in cancer medicine. Organoids offer unique advantages over cell lines and patient-derived xenograft models, but there are no standardized methods to guide the culture of organoids, leading to confusion in organoid studies that may affect accurate judgments of tumor biology. This review summarizes the shortcomings of current organoid culture methods, presents the latest research findings on organoid standardization, and proposes an outlook for organoid modeling.
Collapse
Affiliation(s)
- Changchun Zhou
- Biobank, Cancer Research Center, Shandong Cancer Hospital, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yuanbo Wu
- Department of Ultrasound, Yangxin County People's Hospital, Huangshi, Hubei, China
| | - Zeyu Wang
- Department of Gastrointestinal Surgery, Renji Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yanli Liu
- Biobank, Cancer Research Center, Shandong Cancer Hospital, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Jiaqi Yu
- Biobank, Cancer Research Center, Shandong Cancer Hospital, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Weiping Wang
- Department of Pharmacology and Pharmacy, Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Hong Kong, China
| | - Sunrui Chen
- Shanghai OneTar Biomedicine, Shanghai, China
| | - Weihua Wu
- Shanghai OneTar Biomedicine, Shanghai, China
| | - Jidong Wang
- Shanghai OneTar Biomedicine, Shanghai, China
| | - Guowei Qian
- Department of Oncology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiaotong University, Shanghai, China
| | - Aina He
- Department of Oncology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
71
|
Nie L, Wang YN, Hsu JM, Hou J, Chu YY, Chan LC, Huo L, Wei Y, Deng R, Tang J, Hsu YH, Ko HW, Lim SO, Huang K, Chen MK, Chiu TJ, Cheng CC, Fang YF, Li CW, Goverdhan A, Wu HJ, Lee CC, Wang WL, Hsu J, Chiao P, Wang SC, Hung MC. Nuclear export signal mutation of epidermal growth factor receptor enhances malignant phenotypes of cancer cells. Am J Cancer Res 2023; 13:1209-1239. [PMID: 37168336 PMCID: PMC10164793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 02/16/2023] [Indexed: 05/13/2023] Open
Abstract
Nuclear epidermal growth factor receptor (EGFR) has been shown to be correlated with drug resistance and a poor prognosis in patients with cancer. Previously, we have identified a tripartite nuclear localization signal (NLS) within EGFR. To comprehensively determine the functions and underlying mechanism of nuclear EGFR and its clinical implications, we aimed to explore the nuclear export signal (NES) sequence of EGFR that is responsible for interacting with the exportins. We combined in silico prediction with site-directed mutagenesis approaches and identified a putative NES motif of EGFR, which is located in amino acid residues 736-749. Mutation at leucine 747 (L747) in the EGFR NES led to increased nuclear accumulation of the protein via a less efficient release of the exportin CRM1. Interestingly, L747 with serine (L747S) and with proline (L747P) mutations were found in both tyrosine kinase inhibitor (TKI)-treated and -naïve patients with lung cancer who had acquired or de novo TKI resistance and a poor outcome. Reconstituted expression of the single NES mutant EGFRL747P or EGFRL747S, but not the dual mutant along with the internalization-defective or NLS mutation, in lung cancer cells promoted malignant phenotypes, including cell migration, invasiveness, TKI resistance, and tumor initiation, supporting an oncogenic role of nuclear EGFR. Intriguingly, cells with germline expression of the NES L747 mutant developed into B cell lymphoma. Mechanistically, nuclear EGFR signaling is required for sustaining nuclear activated STAT3, but not for Erk. These findings suggest that EGFR functions are compartmentalized and that nuclear EGFR signaling plays a crucial role in tumor malignant phenotypes, leading to tumorigenesis in human cancer.
Collapse
Affiliation(s)
- Lei Nie
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
| | - Ying-Nai Wang
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
| | - Jung-Mao Hsu
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
- Center for Molecular Medicine, China Medical University HospitalTaichung, Taiwan
| | - Junwei Hou
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
| | - Yu-Yi Chu
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
| | - Li-Chuan Chan
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
- UTHealth Houston Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
| | - Longfei Huo
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
| | - Yongkun Wei
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
| | - Rong Deng
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen UniversityGuangzhou, Guangdong, China
| | - Jun Tang
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
- Department of Breast Oncology, Cancer Center, Sun Yat-Sen UniversityGuangzhou, Guangdong, China
| | - Yi-Hsin Hsu
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
| | - How-Wen Ko
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
- UTHealth Houston Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Chang Gung University College of MedicineTaoyuan, Taiwan
| | - Seung-Oe Lim
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
| | - Kebin Huang
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry & Pharmacy, Guangxi Normal UniversityGuilin, Guangxi, China
| | - Mei-Kuang Chen
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
- UTHealth Houston Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
| | - Tai-Jan Chiu
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
- Department of Hematology-Oncology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of MedicineKaohsiung, Taiwan
| | - Chien-Chia Cheng
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
| | - Yueh-Fu Fang
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Chang Gung University College of MedicineTaoyuan, Taiwan
| | - Chia-Wei Li
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
| | - Aarthi Goverdhan
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
- UTHealth Houston Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
| | - Hsing-Ju Wu
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
- Department of Medical Research, Chang Bing Show Chwan Memorial HospitalChanghua, Taiwan
| | - Cheng-Chung Lee
- Center for Molecular Medicine, China Medical University HospitalTaichung, Taiwan
| | - Wen-Ling Wang
- Center for Molecular Medicine, China Medical University HospitalTaichung, Taiwan
| | - Jennifer Hsu
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
| | - Paul Chiao
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
- UTHealth Houston Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
| | - Shao-Chun Wang
- Center for Molecular Medicine, China Medical University HospitalTaichung, Taiwan
| | - Mien-Chie Hung
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
- Center for Molecular Medicine, China Medical University HospitalTaichung, Taiwan
- Graduate Institute of Biomedical Sciences, Institute of Biochemistry and Molecular Biology, Research Center for Cancer Biology, China Medical UniversityTaichung, Taiwan
| |
Collapse
|
72
|
Brisson BK, Dekky B, Berger AC, Mauldin EA, Loebel C, Yen W, Stewart DC, Gillette D, Assenmacher CA, Cukierman E, Burdick JA, Borges VF, Volk SW. Tumor-restrictive type III collagen in the breast cancer microenvironment: prognostic and therapeutic implications. RESEARCH SQUARE 2023:rs.3.rs-2631314. [PMID: 37090621 PMCID: PMC10120781 DOI: 10.21203/rs.3.rs-2631314/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Collagen plays a critical role in regulating breast cancer progression and therapeutic resistance. An improved understanding of both the features and drivers of tumor-permissive and -restrictive collagen matrices are critical to improve prognostication and develop more effective therapeutic strategies. In this study, using a combination of in vitro, in vivo and in silico experiments, we show that type III collagen (Col3) plays a tumor-restrictive role in human breast cancer. We demonstrate that Col3-deficient, human fibroblasts produce tumor-permissive collagen matrices that drive cell proliferation and suppress apoptosis in noninvasive and invasive breast cancer cell lines. In human TNBC biopsy samples, we demonstrate elevated deposition of Col3 relative to type I collagen (Col1) in noninvasive compared to invasive regions. Similarly, in silico analyses of over 1000 breast cancer patient biopsies from The Cancer Genome Atlas BRCA cohort revealed that patients with higher Col3:Col1 bulk tumor expression had improved overall, disease-free and progression-free survival relative to those with higher Col1:Col3 expression. Using an established 3D culture model, we show that Col3 increases spheroid formation and induces formation of lumen-like structures that resemble non-neoplastic mammary acini. Finally, our in vivo study shows co-injection of murine breast cancer cells (4T1) with rhCol3-supplemented hydrogels limits tumor growth and decreases pulmonary metastatic burden compared to controls. Taken together, these data collectively support a tumor-suppressive role for Col3 in human breast cancer and suggest that strategies that increase Col3 may provide a safe and effective modality to limit recurrence in breast cancer patients.
Collapse
Affiliation(s)
- Becky K. Brisson
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Bassil Dekky
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ashton C. Berger
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Elizabeth A. Mauldin
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Claudia Loebel
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Materials Science & Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - William Yen
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Daniel C. Stewart
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Deborah Gillette
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Charles-Antoine Assenmacher
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Edna Cukierman
- Cancer Signaling and Microenvironment Program, The Martin and Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, USA
| | - Jason A. Burdick
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- BioFrontiers Institute and Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, Colorado, USA
| | - Virginia F. Borges
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- University of Colorado Cancer Center, Aurora, Colorado, USA
- Young Women’s Breast Cancer Translational Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Susan W. Volk
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
73
|
Peng J, Chan C, Zhang S, Sklavounos AA, Olson ME, Scott EY, Hu Y, Rajesh V, Li BB, Chamberlain MD, Zhang S, Peng H, Wheeler AR. All-in-One digital microfluidics pipeline for proteomic sample preparation and analysis. Chem Sci 2023; 14:2887-2900. [PMID: 36937585 PMCID: PMC10016607 DOI: 10.1039/d3sc00560g] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 02/07/2023] [Indexed: 02/24/2023] Open
Abstract
Highly sensitive and reproducible analysis of samples containing low amounts of protein is restricted by sample loss and the introduction of contaminants during processing. Here, we report an All-in-One digital microfluidic (DMF) pipeline for proteomic sample reduction, alkylation, digestion, isotopic labeling and analysis. The system features end-to-end automation, with integrated thermal control for digestion, optimized droplet additives for sample manipulation and analysis, and an automated interface to liquid chromatography with tandem mass spectrometry (HPLC-MS/MS). Dimethyl labeling was integrated into the pipeline to allow for relative quantification of the trace samples at the nanogram level, and the new pipeline was applied to evaluating cancer cell lines and cancer tissue samples. Several known proteins (including HSP90AB1, HSPB1, LDHA, ENO1, PGK1, KRT18, and AKR1C2) and pathways were observed between model breast cancer cell lines related to hormone response, cell metabolism, and cell morphology. Furthermore, differentially quantified proteins (such as PGS2, UGDH, ASPN, LUM, COEA1, and PRELP) were found in comparisons of healthy and cancer breast tissues, suggesting potential utility of the All-in-One pipeline for the emerging application of proteomic cancer sub-typing. In sum, the All-in-One pipeline represents a powerful new tool for automated proteome processing and analysis, with the potential to be useful for evaluating mass-limited samples for a wide range of applications.
Collapse
Affiliation(s)
- Jiaxi Peng
- Department of Chemistry, University of Toronto 80 St. George Street Toronto ON M5S 3H6 Canada +1-416-946-3865 +1-416-946-3866
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto 160 College Street Toronto ON M5S 3E1 Canada
- Institute of Biomedical Engineering, University of Toronto 164 College Street Toronto ON M5S 3G9 Canada
| | - Calvin Chan
- Department of Chemistry, University of Toronto 80 St. George Street Toronto ON M5S 3H6 Canada +1-416-946-3865 +1-416-946-3866
| | - Shuailong Zhang
- Department of Chemistry, University of Toronto 80 St. George Street Toronto ON M5S 3H6 Canada +1-416-946-3865 +1-416-946-3866
- School of Mechatronical Engineering, Beijing Institute of Technology Beijing 100081 China
- Beijing Advanced Innovation Center for Intelligent Robots and Systems, Beijing Institute of Technology Beijing 100081 China
| | - Alexandros A Sklavounos
- Department of Chemistry, University of Toronto 80 St. George Street Toronto ON M5S 3H6 Canada +1-416-946-3865 +1-416-946-3866
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto 160 College Street Toronto ON M5S 3E1 Canada
| | - Maxwell E Olson
- Department of Chemistry, University of Toronto 80 St. George Street Toronto ON M5S 3H6 Canada +1-416-946-3865 +1-416-946-3866
| | - Erica Y Scott
- Department of Chemistry, University of Toronto 80 St. George Street Toronto ON M5S 3H6 Canada +1-416-946-3865 +1-416-946-3866
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto 160 College Street Toronto ON M5S 3E1 Canada
- Institute of Biomedical Engineering, University of Toronto 164 College Street Toronto ON M5S 3G9 Canada
| | - Yechen Hu
- Department of Chemistry, University of Toronto 80 St. George Street Toronto ON M5S 3H6 Canada +1-416-946-3865 +1-416-946-3866
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto 160 College Street Toronto ON M5S 3E1 Canada
- Institute of Biomedical Engineering, University of Toronto 164 College Street Toronto ON M5S 3G9 Canada
| | - Vigneshwar Rajesh
- Department of Chemistry, University of Toronto 80 St. George Street Toronto ON M5S 3H6 Canada +1-416-946-3865 +1-416-946-3866
| | - Bingyu B Li
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto 160 College Street Toronto ON M5S 3E1 Canada
- Institute of Biomedical Engineering, University of Toronto 164 College Street Toronto ON M5S 3G9 Canada
| | - M Dean Chamberlain
- Department of Chemistry, University of Toronto 80 St. George Street Toronto ON M5S 3H6 Canada +1-416-946-3865 +1-416-946-3866
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto 160 College Street Toronto ON M5S 3E1 Canada
- Institute of Biomedical Engineering, University of Toronto 164 College Street Toronto ON M5S 3G9 Canada
- Saskatchewan Cancer Agency, University of Saskatchewan 107 Wiggins Road Saskatoon SK S7N 5E5 Canada
| | - Shen Zhang
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital 600 University Avenue Toronto ON M5G 1X5 Canada
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA Changsha Hunan 410000 China
| | - Hui Peng
- Department of Chemistry, University of Toronto 80 St. George Street Toronto ON M5S 3H6 Canada +1-416-946-3865 +1-416-946-3866
- School of Environment, University of Toronto 33 Willcocks Street Toronto ON M5S 3E8 Canada
| | - Aaron R Wheeler
- Department of Chemistry, University of Toronto 80 St. George Street Toronto ON M5S 3H6 Canada +1-416-946-3865 +1-416-946-3866
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto 160 College Street Toronto ON M5S 3E1 Canada
- Institute of Biomedical Engineering, University of Toronto 164 College Street Toronto ON M5S 3G9 Canada
| |
Collapse
|
74
|
Rosado-Galindo H, Domenech M. Surface roughness modulates EGFR signaling and stemness of triple-negative breast cancer cells. Front Cell Dev Biol 2023; 11:1124250. [PMID: 36968199 PMCID: PMC10030610 DOI: 10.3389/fcell.2023.1124250] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 02/27/2023] [Indexed: 03/29/2023] Open
Abstract
Introduction: Cancer stem cells (CSC), a major culprit of drug-resistant phenotypes and tumor relapse, represent less than 2 % of the bulk of TNBC cells, making them difficult to isolate, study, and thus, limiting our understanding of the pathogenesis of the disease. Current methods for CSC enrichment, such as 3D spheroid culture, genetic modification, and stem cell conditioning, are time consuming, expensive, and unsuitable for high-throughput assays. One way to address these limitations is to use topographical stimuli to enhance CSC populations in planar culture. Physical cues in the breast tumor microenvironment can influence cell behavior through changes in the mechanical properties of the extracellular matrix (ECM). In this study, we used topographical cues on polystyrene films to investigate their effect on the proteome and stemness of standard TNBC cell lines. Methods: The topographical polystyrene-based array was generated using razor printing and polishing methods. Proteome data were analyzed and enriched bioprocesses were identified using R software. Stemness was assessed measuring CD44, CD24 and ALDH markers using flow cytometry, immunofluorescence, detection assays, and further validated with mammosphere assay. EGF/EGFR expression and activity was evaluated using enzyme-linked immunosorbent assay (ELISA), immunofluorescence and antibody membrane array. A dose-response assay was performed to further investigate the effect of surface topography on the sensitivity of cells to the EGFR inhibitor. Results: Surface roughness enriched the CSC population and modulated epidermal growth factor receptor (EGFR) signaling activity in TNBC cells. Enhanced proliferation of MDA-MB-468 cells in roughness correlated with upregulation of the epidermal growth factor (EGF) ligand, which in turn corresponded with a 3-fold increase in the expression of EGFR and a 42% increase in its phosphorylation compared to standard smooth culture surfaces. The results also demonstrated that phenotypic changes associated with topographical (roughness) stimuli significantly decreased the drug sensitivity to the EGFR inhibitor gefitinib. In addition, the proportion of CD44+/CD24-/ALDH+ was enhanced on surface roughness in both MDA-MB-231 and MDA-MB-468 cell lines. We also demonstrated that YAP/TAZ activation decreased in a roughness-dependent manner, confirming the mechanosensing effect of the topographies on the oncogenic activity of the cells. Discussion: Overall, this study demonstrates the potential of surface roughness as a culture strategy to influence oncogenic activity in TNBC cells and enrich CSC populations in planar cultures. Such a culture strategy may benefit high-throughput screening studies seeking to identify compounds with broader tumor efficacy.
Collapse
Affiliation(s)
| | - Maribella Domenech
- Bioengineering Program, University of Puerto Rico-Mayagüez, Mayagüez, Puerto Rico
- Department of Chemical Engineering, University of Puerto Rico-Mayagüez, Mayagüez, Puerto Rico
| |
Collapse
|
75
|
Targeting mTOR to overcome resistance to hormone and CDK4/6 inhibitors in ER-positive breast cancer models. Sci Rep 2023; 13:2710. [PMID: 36792625 PMCID: PMC9932145 DOI: 10.1038/s41598-023-29425-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 02/03/2023] [Indexed: 02/17/2023] Open
Abstract
Resistance to therapy remains a major obstacle in cancer management. Although treatment with hormone and CDK4/6 inhibitors is successful in luminal breast cancer, resistance to these treatments is frequent, highlighting the need for novel therapeutic strategies to delay disease progression and improve patient survival. Here, we assessed the mechanisms of acquired resistance using T47D and MCF-7 tamoxifen- and palbociclib-resistant cell-line variants in culture and as xenografts, and patient-derived cells (PDCs) obtained from sensitive or resistant patient-derived xenografts (PDXs). In these models, we analyzed the effect of specific kinase inhibitors on survival, signaling and cellular aggressiveness. Our results revealed that mTOR inhibition is more effective than PI3K inhibition in overcoming resistance, irrespective of PIK3CA mutation status, by decreasing cell proliferation and tumor growth, as well as reducing cell migration and stemness. Moreover, a combination of mTOR and CDK4/6 inhibitors may prevent pathway reactivation downstream of PI3K, interfering with the survival of resistant cells and consequent tumor escape. In conclusion, we highlight the benefits of incorporating mTOR inhibitors into the current therapy in ER + breast cancer. This alternative therapeutic strategy not only enhances the antitumor response but may also delay the emergence of resistance and tumor recurrence.
Collapse
|
76
|
Tahara S, de Faria FCC, Sarchet P, Calore F, Sharick J, Leight JL, Casadei L, Pollock RE. Three dimensional models of dedifferentiated liposarcoma cell lines: scaffold-based and scaffold-free approaches. Hum Cell 2023; 36:1081-1089. [PMID: 36763259 DOI: 10.1007/s13577-023-00865-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 01/17/2023] [Indexed: 02/11/2023]
Abstract
Sarcomas are rare malignancies, the number of reports is limited, and this rarity makes further research difficult even though liposarcoma is one of major sarcomas. 2D cell culture remains an important role in establishing basic tumor biology research, but its various shortcomings and limitations are still of concern, and it is now well-accepted that the behavior of 3D-cultured cells is more reflective of in vivo cellular responses compared to 2D models. This study aimed to establish 3D cell culture of liposarcomas using two different methods: scaffold-based (Matrigel extracellular matrix [ECM] scaffold method) and scaffold-free (Ultra-low attachment [ULA] plate). Lipo246, Lipo224 and Lipo863 cell lines were cultured, and distinctive differences in structures were observed in Matrigel 3D model: Lipo224 and Lipo863 formed spheroids, whereas Lipo246 grew radially without forming spheres. In ULA plate approaches, all cell lines formed spheroids, but Lipo224 and Lipo863 spheroids showed bigger size and looser aggregation than Lipo246. Formalin fixed, paraffin embedded (FFPE) blocks were obtained from all 3D models, confirming the spheroid structures. The expression of MDM2, Ki-67 positivity and MDM2 amplification were confirmed by IHC and DNAscope™, respectively. Protein and DNA were extracted from all samples and MDM2 upregulation was confirmed by western blot and qPCR analysis. After treatment with MDM2 inhibitor SAR405838, DDLPS spheroids demonstrated different sensitivity patterns from 2D models. Taken together, we believed that 3D models would have a possibility to provide us a new predictability of efficacy and toxicity, and considered as one important process in in vitro pre-clinical phase prior to moving forward to clinical trials.
Collapse
Affiliation(s)
- Sayumi Tahara
- Comprehensive Cancer Center, The Ohio State University, 460 West 10th Avenue. Suite D920E, Columbus, OH, 43210, USA
| | - Fernanda Costas C de Faria
- Comprehensive Cancer Center, The Ohio State University, 460 West 10th Avenue. Suite D920E, Columbus, OH, 43210, USA
| | - Patricia Sarchet
- Comprehensive Cancer Center, The Ohio State University, 460 West 10th Avenue. Suite D920E, Columbus, OH, 43210, USA
| | - Federica Calore
- Comprehensive Cancer Center, The Ohio State University, 460 West 10th Avenue. Suite D920E, Columbus, OH, 43210, USA
| | - Joe Sharick
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH, USA
| | - Jennifer L Leight
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH, USA
| | - Lucia Casadei
- Comprehensive Cancer Center, The Ohio State University, 460 West 10th Avenue. Suite D920E, Columbus, OH, 43210, USA
| | - Raphael E Pollock
- Comprehensive Cancer Center, The Ohio State University, 460 West 10th Avenue. Suite D920E, Columbus, OH, 43210, USA.
| |
Collapse
|
77
|
Dougherty J, Harvey K, Liou A, Labella K, Moran D, Brosius S, De Raedt T. Identification of therapeutic sensitivities in a spheroid drug combination screen of Neurofibromatosis Type I associated High Grade Gliomas. PLoS One 2023; 18:e0277305. [PMID: 36730269 PMCID: PMC9894422 DOI: 10.1371/journal.pone.0277305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 10/22/2022] [Indexed: 02/03/2023] Open
Abstract
Neurofibromatosis Type 1 (NF1) patients develop an array of benign and malignant tumors, of which Malignant Peripheral Nerve Sheath Tumors (MPNST) and High Grade Gliomas (HGG) have a dismal prognosis. About 15-20% of individuals with NF1 develop brain tumors and one third of these occur outside of the optic pathway. These non-optic pathway gliomas are more likely to progress to malignancy, especially in adults. Despite their low frequency, high grade gliomas have a disproportional effect on the morbidity of NF1 patients. In vitro drug combination screens have not been performed on NF1-associated HGG, hindering our ability to develop informed clinical trials. Here we present the first in vitro drug combination screen (21 compounds alone or in combination with MEK or PI3K inhibitors) on the only human NF1 patient derived HGG cell line available and on three mouse glioma cell lines derived from the NF1-P53 genetically engineered mouse model, which sporadically develop HGG. These mouse glioma cell lines were never exposed to serum, grow as spheres and express markers that are consistent with an Oligodendrocyte Precursor Cell (OPC) lineage origin. Importantly, even though the true cell of origin for HGG remains elusive, they are thought to arise from the OPC lineage. We evaluated drug sensitivities of the three murine glioma cell lines in a 3D spheroid growth assay, which more accurately reflects drug sensitivities in vivo. Excitingly, we identified six compounds targeting HDACs, BRD4, CHEK1, BMI-1, CDK1/2/5/9, and the proteasome that potently induced cell death in our NF1-associated HGG. Moreover, several of these inhibitors work synergistically with either MEK or PI3K inhibitors. This study forms the basis for further pre-clinical evaluation of promising targets, with an eventual hope to translate these to the clinic.
Collapse
Affiliation(s)
- Jacquelyn Dougherty
- Department of Pediatrics, Children’s Hospital Philadelphia, Philadelphia, Pennsylvania, United States of America
- School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Kyra Harvey
- Department of Pediatrics, Children’s Hospital Philadelphia, Philadelphia, Pennsylvania, United States of America
- School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Angela Liou
- Department of Pediatrics, Children’s Hospital Philadelphia, Philadelphia, Pennsylvania, United States of America
- School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Katherine Labella
- Department of Pediatrics, Children’s Hospital Philadelphia, Philadelphia, Pennsylvania, United States of America
- School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Deborah Moran
- Department of Pediatrics, Children’s Hospital Philadelphia, Philadelphia, Pennsylvania, United States of America
- School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Stephanie Brosius
- Department of Pediatrics, Children’s Hospital Philadelphia, Philadelphia, Pennsylvania, United States of America
- School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department or Neurology, Children’s Hospital Philadelphia, Philadelphia, Pennsylvania, United States of America
| | - Thomas De Raedt
- Department of Pediatrics, Children’s Hospital Philadelphia, Philadelphia, Pennsylvania, United States of America
- School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
78
|
Development of 3D-Bioprinted Colitis-Mimicking Model to Assess Epithelial Barrier Function Using Albumin Nano-Encapsulated Anti-Inflammatory Drugs. Biomimetics (Basel) 2023; 8:biomimetics8010041. [PMID: 36810372 PMCID: PMC9944493 DOI: 10.3390/biomimetics8010041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/10/2023] [Accepted: 01/13/2023] [Indexed: 01/19/2023] Open
Abstract
Physiological barrier function is very difficult to replicate in vitro. This situation leads to poor prediction of candidate drugs in the drug development process due to the lack of preclinical modelling for intestinal function. By using 3D bioprinting, we generated a colitis-like condition model that can evaluate the barrier function of albumin nanoencapsulated anti-inflammatory drugs. Histological characterization demonstrated the manifestation of the disease in 3D-bioprinted Caco-2 and HT-29 constructs. A comparison of proliferation rates in 2D monolayer and 3D-bioprinted models was also carried out. This model is compatible with currently available preclinical assays and can be implemented as an effective tool for efficacy and toxicity prediction in drug development.
Collapse
|
79
|
Pajic-Lijakovic I, Eftimie R, Milivojevic M, Bordas SPA. The dynamics along the biointerface between the epithelial and cancer mesenchymal cells: Modeling consideration. Semin Cell Dev Biol 2023; 147:47-57. [PMID: 36631334 DOI: 10.1016/j.semcdb.2022.12.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 12/28/2022] [Indexed: 01/11/2023]
Abstract
Epithelial cancer is the one of most lethal cancer type worldwide. Targeting the early stage of disease would allow dramatic improvements in the survival of cancer patients. The early stage of the disease is related to cancer cell spreading across surrounding healthy epithelium. Consequently, deeper insight into cell dynamics along the biointerface between epithelial and cancer (mesenchymal) cells is necessary in order to control the disease as soon as possible. Cell dynamics along this epithelial-cancer biointerface is the result of the interplay between various biological and physical mechanisms. Despite extensive research devoted to study cancer cell spreading across the epithelium, we still do not understand the physical mechanisms which influences the dynamics along the biointerface. These physical mechanisms are related to the interplay between physical parameters such as: (1) interfacial tension between cancer and epithelial subpopulations, (2) established interfacial tension gradients, (3) the bending rigidity of the biointerface and its impact on the interfacial tension, (4) surface tension of the subpopulations, (5) viscoelasticity caused by collective cell migration, and (6) cell residual stress accumulation. The main goal of this study is to review some of these physical parameters in the context of the epithelial/cancer biointerface elaborated on the model system such as the biointerface between breast epithelial MCF-10A cells and cancer MDA-MB-231 cells and then to incorporate these parameters into a new biophysical model that could describe the dynamics of the biointerface. We conclude by discussing three biophysical scenarios for cell dynamics along the biointerface, which can occur depending on the magnitude of the generated shear stress: a smooth biointerface, a slightly-perturbed biointerface and an intensively-perturbed biointerface in the context of the Kelvin-Helmholtz instability. These scenarios are related to the probability of cancer invasion.
Collapse
Affiliation(s)
- Ivana Pajic-Lijakovic
- University of Belgrade, Faculty of Technology and Metallurgy, Department of Chemical Engineering, Serbia.
| | - Raluca Eftimie
- Laboratoire Mathematiques de Besançon, UMR-CNRS 6623, Université de Bourgogne Franche-Comte, 16 Route de Gray, Besançon 25000, France
| | - Milan Milivojevic
- University of Belgrade, Faculty of Technology and Metallurgy, Department of Chemical Engineering, Serbia
| | - Stéphane P A Bordas
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan; Institute for Computational Engineering, Faculty of Science, Technology and Communication, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| |
Collapse
|
80
|
Whitford MKM, McCaffrey L. Polarity in breast development and cancer. Curr Top Dev Biol 2023; 154:245-283. [PMID: 37100520 DOI: 10.1016/bs.ctdb.2023.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2023]
Abstract
Mammary gland development and breast cancer progression are associated with extensive remodeling of epithelial tissue architecture. Apical-basal polarity is a key feature of epithelial cells that coordinates key elements of epithelial morphogenesis including cell organization, proliferation, survival, and migration. In this review we discuss advances in our understanding of how apical-basal polarity programs are used in breast development and cancer. We describe cell lines, organoids, and in vivo models commonly used for studying apical-basal polarity in breast development and disease and discuss advantages and limitations of each. We also provide examples of how core polarity proteins regulate branching morphogenesis and lactation during development. We describe alterations to core polarity genes in breast cancer and their associations with patient outcomes. The impact of up- or down-regulation of key polarity proteins in breast cancer initiation, growth, invasion, metastasis, and therapeutic resistance are discussed. We also introduce studies demonstrating that polarity programs are involved in regulating the stroma, either through epithelial-stroma crosstalk, or through signaling of polarity proteins in non-epithelial cell types. Overall, a key concept is that the function of individual polarity proteins is highly contextual, depending on developmental or cancer stage and cancer subtype.
Collapse
Affiliation(s)
- Mara K M Whitford
- Goodman Cancer Institute, McGill University, Montreal, Quebec, Canada; Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | - Luke McCaffrey
- Goodman Cancer Institute, McGill University, Montreal, Quebec, Canada; Department of Biochemistry, McGill University, Montreal, Quebec, Canada; Gerald Bronfman Department of Oncology, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
81
|
Abstract
Spheroids enable the study of tumors and tumor hypoxia using a more representative model of the physiological environment compared to 2D cell culture. Spheroids can be grown in a cell suspension or when adhered to a solid scaffold. The spheroid formation method used is dependent on cell type. Here we describe the most common spheroid formation methods, including hanging drop, low adhesion plates, hydrogel, micropatterned plates, and microfluidics. After spheroids are formed, they can be used for drug treatment trials and analyzed using Western Blots, qPCR, and microscopy. Microscopy can then be used to measure the invasiveness of cells when a basement membrane is added to spheroids and for monitoring changes in the proliferation, quiescent, and necrotic zones of spheroids.
Collapse
Affiliation(s)
- Sarah M Kirsh
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| | - Sydney A Pascetta
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| | - James Uniacke
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada.
| |
Collapse
|
82
|
Matsubara D, Yoshimoto T, Akolekar N, Totsuka T, Amano Y, Kihara A, Miura T, Isagawa Y, Sakuma Y, Ishikawa S, Ushiku T, Fukayama M, Niki T. Genetic and phenotypic determinants of morphologies in 3D cultures and xenografts of lung tumor cell lines. Cancer Sci 2022; 114:1757-1770. [PMID: 36533957 PMCID: PMC10067422 DOI: 10.1111/cas.15702] [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: 07/28/2022] [Revised: 12/01/2022] [Accepted: 12/11/2022] [Indexed: 12/23/2022] Open
Abstract
We previously proposed the classification of lung adenocarcinoma into two groups: the bronchial epithelial phenotype (BE phenotype) with high-level expressions of bronchial epithelial markers and actionable genetic abnormalities of tyrosine kinase receptors and the non-BE phenotype with low-level expressions of bronchial Bronchial epithelial (BE) epithelial markers and no actionable genetic abnormalities of tyrosine kinase receptors. Here, we performed a comprehensive analysis of tumor morphologies in 3D cultures and xenografts across a panel of lung cancer cell lines. First, we demonstrated that 40 lung cancer cell lines (23 BE and 17 non-BE) can be classified into three groups based on morphologies in 3D cultures on Matrigel: round (n = 31), stellate (n = 5), and grape-like (n = 4). The latter two morphologies were significantly frequent in the non-BE phenotype (1/23 BE, 8/17 non-BE, p = 0.0014), and the stellate morphology was only found in the non-BE phenotype. SMARCA4 mutations were significantly frequent in stellate-shaped cells (4/4 stellate, 4/34 non-stellate, p = 0.0001). Next, from the 40 cell lines, we successfully established 28 xenograft tumors (18 BE and 10 non-BE) in NOD/SCID mice and classified histological patterns of the xenograft tumors into three groups: solid (n = 20), small nests in desmoplasia (n = 4), and acinar/papillary (n = 4). The latter two patterns were characteristically found in the BE phenotype. The non-BE phenotype exhibited a solid pattern with significantly less content of alpha-SMA-positive fibroblasts (p = 0.0004) and collagen (p = 0.0006) than the BE phenotype. Thus, the morphology of the tumors in 3D cultures and xenografts, including stroma genesis, reflects the intrinsic properties of the cancer cell lines. Furthermore, this study serves as an excellent resource for lung adenocarcinoma cell lines, with clinically relevant information on molecular and morphological characteristics and drug sensitivity.
Collapse
Affiliation(s)
- Daisuke Matsubara
- Department of Integrative Pathology, Jichi Medical University, Tochigi, Japan.,Department of Pathology, University of Tsukuba, Ibaraki, Japan
| | - Taichiro Yoshimoto
- Department of Integrative Pathology, Jichi Medical University, Tochigi, Japan
| | | | | | - Yusuke Amano
- Department of Integrative Pathology, Jichi Medical University, Tochigi, Japan
| | - Atsushi Kihara
- Department of Integrative Pathology, Jichi Medical University, Tochigi, Japan
| | - Tamaki Miura
- Department of Integrative Pathology, Jichi Medical University, Tochigi, Japan
| | - Yuriko Isagawa
- Department of Integrative Pathology, Jichi Medical University, Tochigi, Japan
| | - Yuji Sakuma
- Department of Integrative Pathology, Jichi Medical University, Tochigi, Japan
| | - Shumpei Ishikawa
- Department of Preventive Medicine, University of Tokyo, Tokyo, Japan
| | - Tetsuo Ushiku
- Human Pathology Department, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Masashi Fukayama
- Human Pathology Department, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Toshiro Niki
- Department of Integrative Pathology, Jichi Medical University, Tochigi, Japan
| |
Collapse
|
83
|
Semi-Synthetic Click-Gelatin Hydrogels as Tunable Platforms for 3D Cancer Cell Culture. Gels 2022; 8:gels8120821. [PMID: 36547345 PMCID: PMC9778549 DOI: 10.3390/gels8120821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 11/25/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
Basement membrane extracts (BME) derived from Engelbreth-Holm-Swarm (EHS) mouse sarcomas such as Matrigel® remain the gold standard extracellular matrix (ECM) for three-dimensional (3D) cell culture in cancer research. Yet, BMEs suffer from substantial batch-to-batch variation, ill-defined composition, and lack the ability for physichochemical manipulation. Here, we developed a novel 3D cell culture system based on thiolated gelatin (Gel-SH), an inexpensive and highly controlled raw material capable of forming hydrogels with a high level of biophysical control and cell-instructive bioactivity. We demonstrate the successful thiolation of gelatin raw materials to enable rapid covalent crosslinking upon mixing with a synthetic poly(ethylene glycol) (PEG)-based crosslinker. The mechanical properties of the resulting gelatin-based hydrogels were readily tuned by varying precursor material concentrations, with Young's moduli ranging from ~2.5 to 5.8 kPa. All hydrogels of varying stiffnesses supported the viability and proliferation of MDA-MB-231 and MCF-7 breast cancer cell lines for 14 and 21 days of cell culture, respectively. Additionally, the gelatin-based hydrogels supported the growth, viability, and osteogenic differentiation of patient-derived preosteoblasts over 28 days of culture. Collectively, our data demonstrate that gelatin-based biomaterials provide an inexpensive and tunable 3D cell culture platform that may overcome the limitations of traditional BMEs.
Collapse
|
84
|
Ellison ST, Duraivel S, Subramaniam V, Hugosson F, Yu B, Lebowitz JJ, Khoshbouei H, Lele TP, Martindale MQ, Angelini TE. Cellular micromasonry: biofabrication with single cell precision. SOFT MATTER 2022; 18:8554-8560. [PMID: 36350122 DOI: 10.1039/d2sm01013e] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
In many tissues, cell type varies over single-cell length-scales, creating detailed heterogeneities fundamental to physiological function. To gain understanding of the relationship between tissue function and detailed structure, and eventually to engineer structurally and physiologically accurate tissues, we need the ability to assemble 3D cellular structures having the level of detail found in living tissue. Here we introduce a method of 3D cell assembly having a level of precision finer than the single-cell scale. With this method we create detailed cellular patterns, demonstrating that cell type can be varied over the single-cell scale and showing function after their assembly.
Collapse
Affiliation(s)
- S Tori Ellison
- Department of Material Sciences and Engineering, University of Florida, Gainesville, Florida 32611, USA.
| | - Senthilkumar Duraivel
- Department of Material Sciences and Engineering, University of Florida, Gainesville, Florida 32611, USA.
| | - Vignesh Subramaniam
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, Florida 32611, USA
| | - Fredrik Hugosson
- The Whitney Laboratory for Marine Bioscience, St. Augustine, Florida 32080, USA
| | - Bo Yu
- Department of Chemical Engineering, University of Florida, Gainesville, Florida 32611, USA
| | - Joseph J Lebowitz
- Department of Neuroscience, University of Florida, Gainesville, Florida 32611, USA
| | - Habibeh Khoshbouei
- Department of Neuroscience, University of Florida, Gainesville, Florida 32611, USA
| | - Tanmay P Lele
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas 77843, USA
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, Texas 77843, USA
- Department of Translational Medical Sciences, Texas A&M University, Houston, Texas 77843, USA
| | - Mark Q Martindale
- The Whitney Laboratory for Marine Bioscience, St. Augustine, Florida 32080, USA
| | - Thomas E Angelini
- Department of Material Sciences and Engineering, University of Florida, Gainesville, Florida 32611, USA.
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, Florida 32611, USA
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida 32611, USA
| |
Collapse
|
85
|
Quicke P, Sun Y, Arias-Garcia M, Beykou M, Acker CD, Djamgoz MBA, Bakal C, Foust AJ. Voltage imaging reveals the dynamic electrical signatures of human breast cancer cells. Commun Biol 2022; 5:1178. [PMID: 36369329 PMCID: PMC9652252 DOI: 10.1038/s42003-022-04077-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 10/05/2022] [Indexed: 11/13/2022] Open
Abstract
Cancer cells feature a resting membrane potential (Vm) that is depolarized compared to normal cells, and express active ionic conductances, which factor directly in their pathophysiological behavior. Despite similarities to 'excitable' tissues, relatively little is known about cancer cell Vm dynamics. Here high-throughput, cellular-resolution Vm imaging reveals that Vm fluctuates dynamically in several breast cancer cell lines compared to non-cancerous MCF-10A cells. We characterize Vm fluctuations of hundreds of human triple-negative breast cancer MDA-MB-231 cells. By quantifying their Dynamic Electrical Signatures (DESs) through an unsupervised machine-learning protocol, we identify four classes ranging from "noisy" to "blinking/waving". The Vm of MDA-MB-231 cells exhibits spontaneous, transient hyperpolarizations inhibited by the voltage-gated sodium channel blocker tetrodotoxin, and by calcium-activated potassium channel inhibitors apamin and iberiotoxin. The Vm of MCF-10A cells is comparatively static, but fluctuations increase following treatment with transforming growth factor-β1, a canonical inducer of the epithelial-to-mesenchymal transition. These data suggest that the ability to generate Vm fluctuations may be a property of hybrid epithelial-mesenchymal cells or those originated from luminal progenitors.
Collapse
Affiliation(s)
- Peter Quicke
- Department of Bioengineering, Imperial College London, London, SW7 2AL, UK
- The Francis Crick Institute, London, NW1 1AT, UK
| | - Yilin Sun
- Department of Bioengineering, Imperial College London, London, SW7 2AL, UK
| | - Mar Arias-Garcia
- Institute of Cancer Research, Cancer Biology, London, SW3 6JB, UK
| | - Melina Beykou
- Institute of Cancer Research, Cancer Biology, London, SW3 6JB, UK
- Department of Electrical and Electronic Engineering, Imperial College London, London, SW7 2AZ, UK
| | - Corey D Acker
- University of Connecticut School of Medicine, R. D. Berlin Center for Cell Analysis and Modeling, Farmington, CT, USA
| | - Mustafa B A Djamgoz
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK
- Biotechnology Research Centre, Cyprus International University, Haspolat, Nicosia, TRNC, Mersin 10, Turkey
| | - Chris Bakal
- Institute of Cancer Research, Cancer Biology, London, SW3 6JB, UK.
| | - Amanda J Foust
- Department of Bioengineering, Imperial College London, London, SW7 2AL, UK.
| |
Collapse
|
86
|
Nuñez-Olvera SI, Aguilar-Arnal L, Cisneros-Villanueva M, Hidalgo-Miranda A, Marchat LA, Salinas-Vera YM, Ramos-Payán R, Pérez-Plasencia C, Carlos-Reyes Á, Puente-Rivera J, López-Camarillo C. Breast Cancer Cells Reprogram the Oncogenic lncRNAs/mRNAs Coexpression Networks in Three-Dimensional Microenvironment. Cells 2022; 11:3458. [PMID: 36359853 PMCID: PMC9656377 DOI: 10.3390/cells11213458] [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: 09/30/2022] [Accepted: 10/21/2022] [Indexed: 11/06/2022] Open
Abstract
Organotypic three-dimensional (3D) cell cultures more accurately mimic the characteristics of solid tumors in vivo in comparison with traditional two-dimensional (2D) monolayer cell models. Currently, studies on the regulation of long non-coding RNAs (lncRNAs) have not been explored in breast cancer cells cultured in 3D microenvironments. In the present research, we studied the expression and potential roles of lncRNAs in estrogen receptor-positive luminal B subtype BT-474 breast cancer cells grown over extracellular matrix proteins-enriched 3D cultures. Global expression profiling using DNA microarrays identifies 290 upregulated and 183 downregulated lncRNAs in 3D cultures relative to 2D condition. Using a co-expression analysis approach of lncRNAs and mRNAs pairs expressed in the same experimental conditions, we identify hundreds of regulatory axes modulating genes involved in cancer hallmarks, such as responses to estrogens, cell proliferation, hypoxia, apical junctions, and resistance to endocrine therapy. In addition, we identified 102 lncRNAs/mRNA correlations in 3D cultures, which were similar to those reported in TCGA datasets obtained from luminal B breast cancer patients. Interestingly, we also found a set of mRNAs transcripts co-expressed with LINC00847 and CTD-2566J3.1 lncRNAs, which were predictors of pathologic complete response and overall survival. Finally, both LINC00847 and CTD -2566J3.1 were co-expressed with essential genes for cancer genetic dependencies, such as FOXA1 y GINS2. Our experimental and predictive findings show that co-expressed lncRNAs/mRNAs pairs exhibit a high degree of similarity with those found in luminal B breast cancer patients, suggesting that they could be adequate pre-clinical tools to identify not only biomarkers related to endocrine therapy response and PCR, but to understand the biological behavior of cancer cells in 3D microenvironments.
Collapse
Affiliation(s)
- Stephanie I. Nuñez-Olvera
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Lorena Aguilar-Arnal
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | | | - Alfredo Hidalgo-Miranda
- Laboratorio de Genómica del Cáncer, Instituto Nacional de Medicina Genómica, Mexico City 14610, Mexico
| | - Laurence A. Marchat
- Programa en Biomedicina Molecular y Red de Biotecnología, ENMyH-Instituto Politécnico Nacional, Mexico City 07320, Mexico
| | - Yarely M. Salinas-Vera
- Departamento de Bioquímica, Centro de Investigación y Estudios Avanzados, Mexico City 07360, Mexico
| | - Rosalio Ramos-Payán
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Sinaloa, Culiacán 80040, Mexico
| | - Carlos Pérez-Plasencia
- Laboratorio de Genómica, Instituto Nacional de Cancerología (INCan). Av. San Fernando 22, Col. Sección XVI. Tlalpan, Mexico City 14080, Mexico
- Laboratorio de Genómica, Unidad de Biomedicina, FES-Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Mexico
| | - Ángeles Carlos-Reyes
- Laboratorio de Onco-Inmunobiología, Departamento de Enfermedades Crónico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias (INER) Ismael Cosio Villegas, Mexico City 14080, Mexico
| | - Jonathan Puente-Rivera
- División de Ciencias de la Salud, Biológicas y Ambientales, Universidad Abierta y a Distancia, Mexico City 03330, Mexico
| | - Cesar López-Camarillo
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, Mexico City 03100, Mexico
| |
Collapse
|
87
|
Nikolić M, Scarcelli G, Tanner K. Multimodal microscale mechanical mapping of cancer cells in complex microenvironments. Biophys J 2022; 121:3586-3599. [PMID: 36059196 PMCID: PMC9617162 DOI: 10.1016/j.bpj.2022.09.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 08/05/2022] [Accepted: 09/02/2022] [Indexed: 02/07/2023] Open
Abstract
The mechanical phenotype of the cell is critical for survival following deformations due to confinement and fluid flow. One idea is that cancer cells are plastic and adopt different mechanical phenotypes under different geometries that aid in their survival. Thus, an attractive goal is to disrupt cancer cells' ability to adopt multiple mechanical states. To begin to address this question, we aimed to quantify the diversity of these mechanical states using in vitro biomimetics to mimic in vivo two-dimensional (2D) and 3D extracellular matrix environments. Here, we used two modalities Brillouin microscopy (∼GHz) and broadband frequency (7-15 kHz) optical tweezer microrheology to measure microscale cell mechanics. We measured the response of intracellular mechanics of cancer cells cultured in 2D and 3D environments where we modified substrate stiffness, dimensionality (2D versus 3D), and presence of fibrillar topography. We determined that there was good agreement between two modalities despite the difference in timescale of the two measurements. These findings on cell mechanical phenotype in different environments confirm a correlation between modalities that employ different mechanisms at different temporal scales (Hz-kHz versus GHz). We also determined that observed heterogeneity in cell shape is more closely linked to the cells' mechanical state. Moreover, individual cells in multicellular spheroids exhibit a lower degree of mechanical heterogeneity when compared with single cells cultured in monodisperse 3D cultures. The observed decreased heterogeneity among cells in spheroids suggested that there is mechanical cooperativity between cells that make up a single spheroid.
Collapse
Affiliation(s)
- Miloš Nikolić
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland; Maryland Biophysics Program, IPST, University of Maryland, College Park, Maryland
| | - Giuliano Scarcelli
- Maryland Biophysics Program, IPST, University of Maryland, College Park, Maryland; Fischell Department of Bioengineering, University of Maryland, College Park, Maryland
| | - Kandice Tanner
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
88
|
Sonju JJ, Dahal A, Prasasty VD, Shrestha P, Liu YY, Jois SD. Assessment of Antitumor and Antiproliferative Efficacy and Detection of Protein-Protein Interactions in Cancer Cells from 3D Tumor Spheroids. Curr Protoc 2022; 2:e569. [PMID: 36286844 PMCID: PMC9886098 DOI: 10.1002/cpz1.569] [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] [Indexed: 11/06/2022]
Abstract
When compared to two-dimensional (2D) cell cultures, 3D spheroids have been considered suitable in vitro models for drug discovery research and other studies of drug activity. Based on different 3D cell culture procedures, we describe procedures we have used to obtain 3D tumor spheroids by both the hanging-drop and ultra-low-attachment plate methods and to analyze the antiproliferative and antitumor efficacy of different chemotherapeutic agents, including a peptidomimetic. We have applied this method to breast and lung cancer cell lines such as BT-474, MCF-7, A549, and Calu-3. We also describe a proximity ligation assay of the cells from the spheroid model to detect protein-protein interactions of EGFR and HER2. © 2022 Wiley Periodicals LLC. Basic Protocol 1: Growth of 3D spheroids using the hanging-drop method Basic Protocol 2: Growth of spheroids using ultra-low-attachment plates Support Protocol 1: Cell viability assay of tumor spheroids Support Protocol 2: Antiproliferative and antitumor study in 3D tumor spheroids Support Protocol 3: Proximity ligation assay on cells derived from 3D spheroids.
Collapse
Affiliation(s)
- Jafrin Jobayer Sonju
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana
- These authors contributed equally to this work
| | - Achyut Dahal
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana
- These authors contributed equally to this work
| | - Vivitri Dewi Prasasty
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana
| | - Prajesh Shrestha
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana
| | - Yong-Yu Liu
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana
| | - Seetharama D. Jois
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana
| |
Collapse
|
89
|
Freckmann EC, Sandilands E, Cumming E, Neilson M, Román-Fernández A, Nikolatou K, Nacke M, Lannagan TRM, Hedley A, Strachan D, Salji M, Morton JP, McGarry L, Leung HY, Sansom OJ, Miller CJ, Bryant DM. Traject3d allows label-free identification of distinct co-occurring phenotypes within 3D culture by live imaging. Nat Commun 2022; 13:5317. [PMID: 36085324 PMCID: PMC9463449 DOI: 10.1038/s41467-022-32958-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 08/25/2022] [Indexed: 11/09/2022] Open
Abstract
Single cell profiling by genetic, proteomic and imaging methods has expanded the ability to identify programmes regulating distinct cell states. The 3-dimensional (3D) culture of cells or tissue fragments provides a system to study how such states contribute to multicellular morphogenesis. Whether cells plated into 3D cultures give rise to a singular phenotype or whether multiple biologically distinct phenotypes arise in parallel is largely unknown due to a lack of tools to detect such heterogeneity. Here we develop Traject3d (Trajectory identification in 3D), a method for identifying heterogeneous states in 3D culture and how these give rise to distinct phenotypes over time, from label-free multi-day time-lapse imaging. We use this to characterise the temporal landscape of morphological states of cancer cell lines, varying in metastatic potential and drug resistance, and use this information to identify drug combinations that inhibit such heterogeneity. Traject3d is therefore an important companion to other single-cell technologies by facilitating real-time identification via live imaging of how distinct states can lead to alternate phenotypes that occur in parallel in 3D culture.
Collapse
Affiliation(s)
- Eva C Freckmann
- Institute of Cancer Sciences, University of Glasgow, Glasgow, G61 1HQ, United Kingdom
- The CRUK Beatson Institute, Glasgow, G61 1BD, United Kingdom
| | - Emma Sandilands
- Institute of Cancer Sciences, University of Glasgow, Glasgow, G61 1HQ, United Kingdom
- The CRUK Beatson Institute, Glasgow, G61 1BD, United Kingdom
| | - Erin Cumming
- Institute of Cancer Sciences, University of Glasgow, Glasgow, G61 1HQ, United Kingdom
- The CRUK Beatson Institute, Glasgow, G61 1BD, United Kingdom
| | - Matthew Neilson
- The CRUK Beatson Institute, Glasgow, G61 1BD, United Kingdom
| | - Alvaro Román-Fernández
- Institute of Cancer Sciences, University of Glasgow, Glasgow, G61 1HQ, United Kingdom
- The CRUK Beatson Institute, Glasgow, G61 1BD, United Kingdom
| | - Konstantina Nikolatou
- Institute of Cancer Sciences, University of Glasgow, Glasgow, G61 1HQ, United Kingdom
- The CRUK Beatson Institute, Glasgow, G61 1BD, United Kingdom
| | - Marisa Nacke
- Institute of Cancer Sciences, University of Glasgow, Glasgow, G61 1HQ, United Kingdom
- The CRUK Beatson Institute, Glasgow, G61 1BD, United Kingdom
| | | | - Ann Hedley
- The CRUK Beatson Institute, Glasgow, G61 1BD, United Kingdom
| | - David Strachan
- The CRUK Beatson Institute, Glasgow, G61 1BD, United Kingdom
| | - Mark Salji
- The CRUK Beatson Institute, Glasgow, G61 1BD, United Kingdom
| | - Jennifer P Morton
- Institute of Cancer Sciences, University of Glasgow, Glasgow, G61 1HQ, United Kingdom
- The CRUK Beatson Institute, Glasgow, G61 1BD, United Kingdom
| | - Lynn McGarry
- The CRUK Beatson Institute, Glasgow, G61 1BD, United Kingdom
| | - Hing Y Leung
- Institute of Cancer Sciences, University of Glasgow, Glasgow, G61 1HQ, United Kingdom
- The CRUK Beatson Institute, Glasgow, G61 1BD, United Kingdom
| | - Owen J Sansom
- Institute of Cancer Sciences, University of Glasgow, Glasgow, G61 1HQ, United Kingdom
- The CRUK Beatson Institute, Glasgow, G61 1BD, United Kingdom
| | - Crispin J Miller
- Institute of Cancer Sciences, University of Glasgow, Glasgow, G61 1HQ, United Kingdom
- The CRUK Beatson Institute, Glasgow, G61 1BD, United Kingdom
| | - David M Bryant
- Institute of Cancer Sciences, University of Glasgow, Glasgow, G61 1HQ, United Kingdom.
- The CRUK Beatson Institute, Glasgow, G61 1BD, United Kingdom.
| |
Collapse
|
90
|
Elangovan A, Hooda J, Savariau L, Puthanmadhomnarayanan S, Yates ME, Chen J, Brown DD, McAuliffe PF, Oesterreich S, Atkinson JM, Lee AV. Loss of E-cadherin Induces IGF1R Activation and Reveals a Targetable Pathway in Invasive Lobular Breast Carcinoma. Mol Cancer Res 2022; 20:1405-1419. [PMID: 35665642 PMCID: PMC9444924 DOI: 10.1158/1541-7786.mcr-22-0090] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 04/23/2022] [Accepted: 06/02/2022] [Indexed: 01/30/2023]
Abstract
No special-type breast cancer [NST; commonly known as invasive ductal carcinoma (IDC)] and invasive lobular carcinoma (ILC) are the two major histological subtypes of breast cancer with significant differences in clinicopathological and molecular characteristics. The defining pathognomonic feature of ILC is loss of cellular adhesion protein, E-cadherin (CDH1). We have previously shown that E-cadherin functions as a negative regulator of the IGF1R and propose that E-cadherin loss in ILC sensitizes cells to growth factor signaling that thus alters their sensitivity to growth factor-signaling inhibitors and their downstream activators. To investigate this potential therapeutic vulnerability, we generated CRISPR-mediated CDH1 knockout (CDH1 KO) IDC cell lines (MCF7, T47D, and ZR75.1) to uncover the mechanism by which loss of E-cadherin results in IGF pathway activation. CDH1 KO cells demonstrated enhanced invasion and migration that was further elevated in response to IGF1, serum and collagen I. CDH1 KO cells exhibited increased sensitivity to IGF resulting in elevated downstream signaling. Despite minimal differences in membranous IGF1R levels between wild-type (WT) and CDH1 KO cells, significantly higher ligand-receptor interaction was observed in the CDH1 KO cells, potentially conferring enhanced downstream signaling activation. Critically, increased sensitivity to IGF1R, PI3K, Akt, and MEK inhibitors was observed in CDH1 KO cells and ILC patient-derived organoids. IMPLICATIONS Overall, this suggests that these targets require further exploration in ILC treatment and that CDH1 loss may be exploited as a biomarker of response for patient stratification.
Collapse
Affiliation(s)
- Ashuvinee Elangovan
- Molecular Genetics and Developmental Biology Graduate Program, University of Pittsburgh School of Medicine, Pittsburgh PA.,Women’s Cancer Research Center, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center (HCC), Magee-Womens Research Institute, Pittsburgh, PA
| | - Jagmohan Hooda
- Women’s Cancer Research Center, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center (HCC), Magee-Womens Research Institute, Pittsburgh, PA
| | - Laura Savariau
- Women’s Cancer Research Center, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center (HCC), Magee-Womens Research Institute, Pittsburgh, PA.,Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA
| | - Susrutha Puthanmadhomnarayanan
- Women’s Cancer Research Center, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center (HCC), Magee-Womens Research Institute, Pittsburgh, PA
| | - Megan E. Yates
- Women’s Cancer Research Center, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center (HCC), Magee-Womens Research Institute, Pittsburgh, PA.,Medical Scientist Training Program, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Jian Chen
- Women’s Cancer Research Center, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center (HCC), Magee-Womens Research Institute, Pittsburgh, PA
| | | | - Priscilla F. McAuliffe
- Women’s Cancer Research Center, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center (HCC), Magee-Womens Research Institute, Pittsburgh, PA.,Department of Surgery, Division of Surgical Oncology, Section of Breast Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Steffi Oesterreich
- Women’s Cancer Research Center, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center (HCC), Magee-Womens Research Institute, Pittsburgh, PA.,Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA
| | - Jennifer M. Atkinson
- Women’s Cancer Research Center, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center (HCC), Magee-Womens Research Institute, Pittsburgh, PA.,Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA.,Corresponding Authors: Adrian V. Lee, PhD, , Phone: 4126417724, Fax: 4126416456, Women’s Cancer Research Center, UPMC Hillman Cancer Center, 204 Craft Avenue, Pittsburgh, PA 15213, USA, Jennifer M. Atkinson, PhD, , Phone: 4126417724, Fax: 4126416456, Women’s Cancer Research Center, UPMC Hillman Cancer Center, 204 Craft Avenue, Pittsburgh, PA 15213, USA
| | - Adrian V. Lee
- Women’s Cancer Research Center, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center (HCC), Magee-Womens Research Institute, Pittsburgh, PA.,Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA.,Corresponding Authors: Adrian V. Lee, PhD, , Phone: 4126417724, Fax: 4126416456, Women’s Cancer Research Center, UPMC Hillman Cancer Center, 204 Craft Avenue, Pittsburgh, PA 15213, USA, Jennifer M. Atkinson, PhD, , Phone: 4126417724, Fax: 4126416456, Women’s Cancer Research Center, UPMC Hillman Cancer Center, 204 Craft Avenue, Pittsburgh, PA 15213, USA
| |
Collapse
|
91
|
Liu H, Heller-Trulli D, Moore CL. Targeting the mRNA endonuclease CPSF73 inhibits breast cancer cell migration, invasion, and self-renewal. iScience 2022; 25:104804. [PMID: 35992060 PMCID: PMC9385686 DOI: 10.1016/j.isci.2022.104804] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 05/26/2022] [Accepted: 07/15/2022] [Indexed: 12/02/2022] Open
Abstract
Cleavage by the endonuclease CPSF73 and polyadenylation of nascent RNA is an essential step in co-transcriptional mRNA maturation. Recent work has surprisingly identified CPSF73 as a promising drug target for inhibiting the growth of specific cancers, triggering further studies on understanding CPSF73 regulation and functions in cells. Here, we report that a HECT-like E3 ligase, UBE3D, participates in stabilizing CPFS73 protein by preventing its ubiquitin-mediated degradation by the proteasome. Depletion of UBE3D leads to CPSF73 downregulation, a pre-mRNA cleavage defect, and dysregulated gene expression in cells. UBE3D dysfunction or chemical inactivation of CPSF73 inhibited migration and invasion as well as stem cell renewal phenotypes in vitro in triple-negative breast cancer cells. In addition, genetic overexpression of CPSF73 promoted breast cancer stemness and knocking down CPSF73 inhibited stem cell renewal properties. Together, our findings indicate that targeting the pre-mRNA processing nuclease CPSF73 has potential for breast cancer therapy.
Collapse
Affiliation(s)
- Huiyun Liu
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Daniel Heller-Trulli
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Claire L. Moore
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA 02111, USA
| |
Collapse
|
92
|
A thermo-sensitive hydrogel composed of methylcellulose/hyaluronic acid/silk fibrin as a biomimetic extracellular matrix to simulate breast cancer malignancy. Eur Polym J 2022. [DOI: 10.1016/j.eurpolymj.2022.111421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
93
|
Cerqueira OLD, Botelho MCS, Fiore APZP, Osório CABDT, Tomasin R, Morais MCC, López RVM, Cardoso EC, Vilella-Arias SA, Reis EM, Bruni-Cardoso A. Prognostic value of integrin αV expression and localization pattern in invasive breast carcinomas. Neoplasia 2022; 30:100803. [PMID: 35526305 PMCID: PMC9092997 DOI: 10.1016/j.neo.2022.100803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 04/18/2022] [Indexed: 10/25/2022]
|
94
|
Tomar A, Uysal-Onganer P, Basnett P, Pati U, Roy I. 3D Disease Modelling of Hard and Soft Cancer Using PHA-Based Scaffolds. Cancers (Basel) 2022; 14:3549. [PMID: 35884609 PMCID: PMC9321847 DOI: 10.3390/cancers14143549] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 07/18/2022] [Indexed: 02/04/2023] Open
Abstract
Tumour cells are shown to change shape and lose polarity when they are cultured in 3D, a feature typically associated with tumour progression in vivo, thus making it significant to study cancer cells in an environment that mimics the in vivo milieu. In this study we established hard (MCF7 and MDA-MB-231, breast cancer) and soft (HCT116, colon cancer) 3D cancer tumour models utilizing a blend of P(3HO-co-3HD) and P(3HB). P(3HO-co-3HD) and P(3HB) belong to a group of natural biodegradable polyesters, PHAs, that are synthesised by microorganisms. The 3D PHA scaffolds produced, with a pore size of 30 to 300 µm, allow for nutrients to diffuse within the scaffold and provide the cells with the flexibility to distribute evenly within the scaffold and grow within the pores. Interestingly, by Day 5, MDA-MB-231 showed dispersed growth in clusters, and MCF7 cells formed an evenly dispersed dense layer, while HCT116 formed large colonies within the pockets of the 3D PHA scaffolds. Our results show Epithelial Mesenchymal Transition (EMT) marker gene expression profiles in the hard tumour cancer models. In the 3D-based PHA scaffolds, MDA-MB-231 cells expressed higher levels of Wnt-11 and mesenchymal markers, such as Snail and its downstream gene Vim mRNAs, while MCF7 cells exhibited no change in their expression. On the other hand, MCF7 cells exhibited a significantly increased E-Cadherin expression as compared to MDA-MB-231 cells. The expression levels of EMT markers were comparative to their expression reported in the tumour samples, making them good representative of cancer models. In future these models will be helpful in mimicking hypoxic tumours, in studying gene expression, cellular signalling, angiogenesis and drug response more accurately than 2D and perhaps other 3D models.
Collapse
Affiliation(s)
- Akanksha Tomar
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India;
| | - Pinar Uysal-Onganer
- Cancer Research Group, School of Life Sciences, College of Liberal Arts and Sciences, University of Westminster, London W1W 6UW, UK;
| | - Pooja Basnett
- School of Life Sciences, College of Liberal Arts and Sciences, University of Westminster, London W1W 6XH, UK;
| | - Uttam Pati
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India;
| | - Ipsita Roy
- Department of Materials Science and Engineering, Faculty of Engineering, University of Sheffield, Sheffield S10 2TN, UK
| |
Collapse
|
95
|
Tong XY, Yang XZ, Gao SQ, Wang XJ, Wen GB, Lin YW. Regulating Effect of Cytochrome b5 Overexpression on Human Breast Cancer Cells. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27144556. [PMID: 35889429 PMCID: PMC9320672 DOI: 10.3390/molecules27144556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 07/11/2022] [Accepted: 07/15/2022] [Indexed: 12/24/2022]
Abstract
Imbalance in the cellular redox system is thought to be associated with the induction and progression of breast cancers, and heme proteins may regulate the redox balance. Cytochrome b5 (Cyt b5) is a small mitochondrial heme protein. Its function and regulating mechanism in breast cancer remain unknown. In this study, we elucidated the level of endogenous oxidative stress in breast cancer cells, MCF-7 cells (hormone receptor-positive cells) and MDA-MB-231 cells (triple-negative cells), and investigated the difference in Cyt b5 content. Based on the low content of Cyt b5 in MDA-MB-231 cells, the overexpression of Cyt b5 was found to regulate the oxidative stress and apoptosis cascades, including ERK1/2 and Akt signaling pathways. The overexpressed Cyt b5 MDA-MB-231 cells were shown to exhibit decreased oxidative stress, less phosphorylation of ERK1/2 and Akt, and less cleavage of caspases 3 and 9 upon treatment with H2O2, as compared to those of normal MDA-MB-231 cells. Moreover, the overexpressed Cyt b5 most likely functioned by interacting with its protein partner, Cyt c, as suggested by co-immunoprecipitation studies. These results indicated that Cyt b5 has different effects on breast cancer cells of different phenotypes, which provides useful information for understanding the multiple roles of Cyt b5 and provides clues for clinical treatment.
Collapse
Affiliation(s)
- Xin-Yi Tong
- School of Chemistry and Chemical Engineering, University of South China, Hengyang 421001, China; (X.-Y.T.); (X.-J.W.)
| | - Xin-Zhi Yang
- Hengyang Medical College, University of South China, Hengyang 421001, China; (X.-Z.Y.); (S.-Q.G.); (G.-B.W.)
| | - Shu-Qin Gao
- Hengyang Medical College, University of South China, Hengyang 421001, China; (X.-Z.Y.); (S.-Q.G.); (G.-B.W.)
- Key Lab of Protein Structure and Function of Universities in Hunan Province, University of South China, Hengyang 421001, China
| | - Xiao-Juan Wang
- School of Chemistry and Chemical Engineering, University of South China, Hengyang 421001, China; (X.-Y.T.); (X.-J.W.)
| | - Ge-Bo Wen
- Hengyang Medical College, University of South China, Hengyang 421001, China; (X.-Z.Y.); (S.-Q.G.); (G.-B.W.)
- Key Lab of Protein Structure and Function of Universities in Hunan Province, University of South China, Hengyang 421001, China
| | - Ying-Wu Lin
- School of Chemistry and Chemical Engineering, University of South China, Hengyang 421001, China; (X.-Y.T.); (X.-J.W.)
- Hengyang Medical College, University of South China, Hengyang 421001, China; (X.-Z.Y.); (S.-Q.G.); (G.-B.W.)
- Key Lab of Protein Structure and Function of Universities in Hunan Province, University of South China, Hengyang 421001, China
- Correspondence: ; Tel.: +86-734-8282375
| |
Collapse
|
96
|
Nette F, Guerra de Souza AC, Laskay T, Ohms M, Dömer D, Drömann D, Rapoport DH. Method for simultaneous tracking of thousands of unlabeled cells within a transparent 3D matrix. PLoS One 2022; 17:e0270456. [PMID: 35749549 PMCID: PMC9232129 DOI: 10.1371/journal.pone.0270456] [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: 03/25/2022] [Accepted: 06/09/2022] [Indexed: 11/30/2022] Open
Abstract
Three-dimensional tracking of cells is one of the most powerful methods to investigate multicellular phenomena, such as ontogenesis, tumor formation or wound healing. However, 3D tracking in a biological environment usually requires fluorescent labeling of the cells and elaborate equipment, such as automated light sheet or confocal microscopy. Here we present a simple method for 3D tracking large numbers of unlabeled cells in a collagen matrix. Using a small lensless imaging setup, consisting of an LED and a photo sensor only, we were able to simultaneously track ~3000 human neutrophil granulocytes in a collagen droplet within an unusually large field of view (>50 mm2) at a time resolution of 4 seconds and a spatial resolution of ~1.5 μm in xy- and ~30 μm in z-direction. The setup, which is small enough to fit into any conventional incubator, was used to investigate chemotaxis towards interleukin-8 (IL-8 or CXCL8) and N-formylmethionyl-leucyl-phenylalanine (fMLP). The influence of varying stiffness and pore size of the embedding collagen matrix could also be quantified. Furthermore, we demonstrate our setup to be capable of telling apart healthy neutrophils from those where a condition of inflammation was (I) induced by exposure to lipopolysaccharide (LPS) and (II) caused by a pre-existing asthma condition. Over the course of our experiments we have tracked more than 420.000 cells. The large cell numbers increase statistical relevance to not only quantify cellular behavior in research, but to make it suitable for future diagnostic applications, too.
Collapse
Affiliation(s)
- Falk Nette
- Fraunhofer Research and Development Center for Marine and Cellular Biotechnology, Lübeck, Germany
| | | | - Tamás Laskay
- Department of Infectious Diseases and Microbiology, University of Lübeck, Lübeck, Germany
| | - Mareike Ohms
- Research Department Virus Immunology, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Daniel Dömer
- Department of Infectious Diseases and Microbiology, University of Lübeck, Lübeck, Germany
| | - Daniel Drömann
- Medical Clinic III Pneumology, University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Daniel Hans Rapoport
- Institute for Medical and Marine Biotechnology, University of Lübeck, Lübeck, Germany
- * E-mail:
| |
Collapse
|
97
|
Souto EP, Dobrolecki LE, Villanueva H, Sikora AG, Lewis MT. In Vivo Modeling of Human Breast Cancer Using Cell Line and Patient-Derived Xenografts. J Mammary Gland Biol Neoplasia 2022; 27:211-230. [PMID: 35697909 PMCID: PMC9433358 DOI: 10.1007/s10911-022-09520-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 05/19/2022] [Indexed: 11/24/2022] Open
Abstract
Historically, human breast cancer has been modeled largely in vitro using long-established cell lines primarily in two-dimensional culture, but also in three-dimensional cultures of varying cellular and molecular complexities. A subset of cell line models has also been used in vivo as cell line-derived xenografts (CDX). While outstanding for conducting detailed molecular analysis of regulatory mechanisms that may function in vivo, results of drug response studies using long-established cell lines have largely failed to translate clinically. In an attempt to address this shortcoming, many laboratories have succeeded in developing clinically annotated patient-derived xenograft (PDX) models of human cancers, including breast, in a variety of host systems. While immunocompromised mice are the predominant host, the immunocompromised rat and pig, zebrafish, as well as the chicken egg chorioallantoic membrane (CAM) have also emerged as potential host platforms to help address perceived shortcomings of immunocompromised mice. With any modeling platform, the two main issues to be resolved are criteria for "credentialing" the models as valid models to represent human cancer, and utility with respect to the ability to generate clinically relevant translational research data. Such data are beginning to emerge, particularly with the activities of PDX consortia such as the NCI PDXNet Program, EuroPDX, and the International Breast Cancer Consortium, as well as a host of pharmaceutical companies and contract research organizations (CRO). This review focuses primarily on these important aspects of PDX-related research, with a focus on breast cancer.
Collapse
Affiliation(s)
- Eric P Souto
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Lacey E Dobrolecki
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Hugo Villanueva
- Otolaryngology-Head and Neck Surgery, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Andrew G Sikora
- Department of Head and Neck Surgery, Division of Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Michael T Lewis
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, 77030, USA.
- Departments of Molecular and Cellular Biology and Radiology, Baylor College of Medicine, Houston, TX, 77030, USA.
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA.
- Baylor College of Medicine, One Baylor Plaza, BCM-600; Room N1210, Houston, TX, 77030, USA.
| |
Collapse
|
98
|
Wang ML, Xu NY, Tang RZ, Liu XQ. A 3D-printed scaffold-based osteosarcoma model allows to investigate tumor phenotypes and pathogenesis in an in vitro bone-mimicking niche. Mater Today Bio 2022; 15:100295. [PMID: 35665234 PMCID: PMC9161108 DOI: 10.1016/j.mtbio.2022.100295] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 05/11/2022] [Accepted: 05/12/2022] [Indexed: 11/23/2022] Open
Abstract
Image 1.
Collapse
Affiliation(s)
- Mei-Ling Wang
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Nian-Yuan Xu
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Rui-Zhi Tang
- Department of Medical Laboratory, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
- Key Laboratory for Molecular Diagnosis of Hubei Province, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Xi-Qiu Liu
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| |
Collapse
|
99
|
Salinas-Vera YM, Valdés J, Pérez-Navarro Y, Mandujano-Lazaro G, Marchat LA, Ramos-Payán R, Nuñez-Olvera SI, Pérez-Plascencia C, López-Camarillo C. Three-Dimensional 3D Culture Models in Gynecological and Breast Cancer Research. Front Oncol 2022; 12:826113. [PMID: 35692756 PMCID: PMC9177953 DOI: 10.3389/fonc.2022.826113] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 04/20/2022] [Indexed: 12/12/2022] Open
Abstract
Traditional two-dimensional (2D) monolayer cell cultures have long been the gold standard for cancer biology research. However, their ability to accurately reflect the molecular mechanisms of tumors occurring in vivo is limited. Recent development of three-dimensional (3D) cell culture models facilitate the possibility to better recapitulate several of the biological and molecular characteristics of tumors in vivo, such as cancer cells heterogeneity, cell-extracellular matrix interactions, development of a hypoxic microenvironment, signaling pathway activities depending on contacts with extracellular matrix, differential growth kinetics, more accurate drugs response, and specific gene expression and epigenetic patterns. In this review, we discuss the utilization of different types of 3D culture models including spheroids, organotypic models and patient-derived organoids in gynecologic cancers research, as well as its potential applications in oncological research mainly for screening drugs with major physiological and clinical relevance. Moreover, microRNAs regulation of cancer hallmarks in 3D cell cultures from different types of cancers is discussed.
Collapse
Affiliation(s)
- Yarely M. Salinas-Vera
- Departamento de Bioquímica, Centro de Investigación de Estudios Avanzados (CINVESTAV-IPN), Ciudad de Mexico, Mexico
| | - Jesús Valdés
- Departamento de Bioquímica, Centro de Investigación de Estudios Avanzados (CINVESTAV-IPN), Ciudad de Mexico, Mexico
| | - Yussel Pérez-Navarro
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de Mexico, Ciudad de Mexico, Mexico
| | - Gilberto Mandujano-Lazaro
- Programa en Biomedicina Molecular y Red de Biotecnología, Instituto Politécnico Nacional, Ciudad de Mexico, Mexico
| | - Laurence A. Marchat
- Programa en Biomedicina Molecular y Red de Biotecnología, Instituto Politécnico Nacional, Ciudad de Mexico, Mexico
| | - Rosalio Ramos-Payán
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Sinaloa, Culiacán Sinaloa, Mexico
| | - Stephanie I. Nuñez-Olvera
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | | | - César López-Camarillo
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de Mexico, Ciudad de Mexico, Mexico
| |
Collapse
|
100
|
Salinas-Vera YM, Valdés J, Hidalgo-Miranda A, Cisneros-Villanueva M, Marchat LA, Nuñez-Olvera SI, Ramos-Payán R, Pérez-Plasencia C, Arriaga-Pizano LA, Prieto-Chávez JL, López-Camarillo C. Three-Dimensional Organotypic Cultures Reshape the microRNAs Transcriptional Program in Breast Cancer Cells. Cancers (Basel) 2022; 14:2490. [PMID: 35626094 PMCID: PMC9139376 DOI: 10.3390/cancers14102490] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 05/03/2022] [Accepted: 05/10/2022] [Indexed: 02/04/2023] Open
Abstract
The 3D organotypic cultures, which depend on the growth of cells over the extracellular matrix (ECM) used as a scaffold, can better mimic several characteristics of solid cancers that influence tumor biology and the response to drug therapies. Most of our current knowledge on cancer is derived from studies in 2D cultures, which lack the ECM-mediated microenvironment. Moreover, the role of miRNAs that is critical for fine-tuning of gene expression is poorly understood in 3D cultures. The aim of this study was to compare the miRNA expression profiles of breast cancer cells grown in 2D and 3D conditions. On an on-top 3D cell culture model using a basement membrane matrix enriched with laminin, collagen IV, entactin, and heparin-sulfate proteoglycans, the basal B (Hs578T) and luminal (T47D) breast cancer cells formed 3D spheroid-like stellate and rounded mass structures, respectively. Morphological changes in 3D cultures were observed as cell stretching, cell-cell, and cell-ECM interactions associated with a loss of polarity and reorganization on bulk structures. Interestingly, we found prolongations of the cytoplasmic membrane of Hs578T cells similar to tunneled nanotubes contacting between neighboring cells, suggesting the existence of cellular intercommunication processes and the possibility of fusion between spheroids. Expression profiling data revealed that 354 miRNAs were differentially expressed in 3D relative to 2D cultures in Hs578T cells. Downregulated miRNAs may contribute to a positive regulation of genes involved in hypoxia, catabolic processes, and focal adhesion, whereas overexpressed miRNAs modulate genes involved in negative regulation of the cell cycle. Target genes of the top ten modulated miRNAs were selected to construct miRNA/mRNA coregulation networks. Around 502 interactions were identified for downregulated miRNAs, including miR-935/HIF1A and miR-5189-3p/AKT that could contribute to cell migration and the response to hypoxia. Furthermore, the expression levels of miR-935 and its target HIF1A correlated with the expression found in clinical tumors and predicted poor outcomes. On the other hand, 416 interactions were identified for overexpressed miRNAs, including miR-6780b-5p/ANKRD45 and miR-7641/CDK4 that may result in cell proliferation inhibition and cell cycle arrest in quiescent layers of 3D cultures. In conclusion, 3D cultures could represent a suitable model that better resembles the miRNA transcriptional programs operating in tumors, with implications not only in the understanding of basic cancer biology in 3D microenvironments, but also in the identification of novel biomarkers of disease and potential targets for personalized therapies in cancer.
Collapse
Affiliation(s)
- Yarely M. Salinas-Vera
- Departamento de Bioquímica, CINVESTAV-IPN, Ciudad de México 07360, Mexico; (Y.M.S.-V.); (J.V.)
| | - Jesús Valdés
- Departamento de Bioquímica, CINVESTAV-IPN, Ciudad de México 07360, Mexico; (Y.M.S.-V.); (J.V.)
| | - Alfredo Hidalgo-Miranda
- Laboratorio de Genómica del Cáncer, Instituto Nacional de Medicina Genómica, Ciudad de México 14610, Mexico; (A.H.-M.); (M.C.-V.)
| | - Mireya Cisneros-Villanueva
- Laboratorio de Genómica del Cáncer, Instituto Nacional de Medicina Genómica, Ciudad de México 14610, Mexico; (A.H.-M.); (M.C.-V.)
| | - Laurence A. Marchat
- Programa en Biomedicina Molecular y Red de Biotecnología, Instituto Politécnico Nacional, Ciudad de México 07320, Mexico;
| | - Stephanie I. Nuñez-Olvera
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico;
| | - Rosalio Ramos-Payán
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Sinaloa, Culiacán 80030, Mexico;
| | - Carlos Pérez-Plasencia
- Laboratorio de Genómica, Instituto Nacional de Cancerología, Ciudad de México 14080, Mexico;
| | - Lourdes A. Arriaga-Pizano
- Unidad de Investigación Médica en Inmunoquímica, Hospital de Especialidades del Centro Medico Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México 06720, Mexico;
| | - Jessica L. Prieto-Chávez
- Laboratorio de Citometria de Flujo, Centro de Instrumentos, Coordinación de Investigación en Salud, Hospital de Especialidades del Centro Médico Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México 06720, Mexico;
| | - César López-Camarillo
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, Ciudad de México 03100, Mexico
| |
Collapse
|