1
|
Coppo R, Kondo J, Onuma K, Inoue M. Tracking the growth fate of single cells and isolating slow-growing cells in human colorectal cancer organoids. STAR Protoc 2023; 4:102395. [PMID: 37384521 PMCID: PMC10511865 DOI: 10.1016/j.xpro.2023.102395] [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: 03/23/2023] [Revised: 05/02/2023] [Accepted: 05/31/2023] [Indexed: 07/01/2023] Open
Abstract
Patient-derived tumor organoids are three-dimensionally cultured cancer cells that enable a suitable platform for studying heterogeneity and plasticity of cancer. We present a protocol for tracking the growth fate of single cells and isolating slow-growing cells in human colorectal cancer organoids. We describe steps for organoid preparation and culturing using the cancer-tissue-originating spheroid method, maintaining cell-cell contact throughout. We then detail a single-cell-derived spheroid-forming and growth assay, confirming single-cell plating, monitoring growth over time, and isolating slow-growing cells. For complete details on the use and execution of this protocol, please refer to Coppo et al.1.
Collapse
Affiliation(s)
- Roberto Coppo
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | - Jumpei Kondo
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kunishige Onuma
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masahiro Inoue
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| |
Collapse
|
2
|
Coppo R, Kondo J, Iida K, Okada M, Onuma K, Tanaka Y, Kamada M, Ohue M, Kawada K, Obama K, Inoue M. Distinct but interchangeable subpopulations of colorectal cancer cells with different growth fates and drug sensitivity. iScience 2023; 26:105962. [PMID: 36718360 PMCID: PMC9883198 DOI: 10.1016/j.isci.2023.105962] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 11/22/2022] [Accepted: 01/09/2023] [Indexed: 01/14/2023] Open
Abstract
Dynamic changes in cell properties lead to intratumor heterogeneity; however, the mechanisms of nongenetic cellular plasticity remain elusive. When the fate of each cell from colorectal cancer organoids was tracked through a clonogenic growth assay, the cells showed a wide range of growth ability even within the clonal organoids, consisting of distinct subpopulations; the cells generating large spheroids and the cells generating small spheroids. The cells from the small spheroids generated only small spheroids (S-pattern), while the cells from the large spheroids generated both small and large spheroids (D-pattern), both of which were tumorigenic. Transition from the S-pattern to the D-pattern occurred by various extrinsic triggers, in which Notch signaling and Musashi-1 played a key role. The S-pattern spheroids were resistant to chemotherapy and transited to the D-pattern upon drug treatment through Notch signaling. As the transition is linked to the drug resistance, it can be a therapeutic target.
Collapse
Affiliation(s)
- Roberto Coppo
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Jumpei Kondo
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Keita Iida
- Institute for Protein Research, Osaka University, Suita, Osaka, Japan
| | - Mariko Okada
- Institute for Protein Research, Osaka University, Suita, Osaka, Japan
| | - Kunishige Onuma
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yoshihisa Tanaka
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan,RIKEN Center for Computational Science, HPC- and AI-driven Drug Development Platform Division, Biomedical Computational Intelligence Unit, Hyogo, Japan
| | - Mayumi Kamada
- Department of Biomedical Data Intelligence, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masayuki Ohue
- Department of Gastroenterological Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Kenji Kawada
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kazutaka Obama
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masahiro Inoue
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan,Corresponding author
| |
Collapse
|
3
|
Regan JL. Protocol for isolation and functional validation of label-retaining quiescent colorectal cancer stem cells from patient-derived organoids for RNA-seq. STAR Protoc 2022; 3:101225. [PMID: 35300001 PMCID: PMC8920924 DOI: 10.1016/j.xpro.2022.101225] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Quiescent cancer stem cells (qCSCs) are a major source of posttreatment relapse, but methods to identify molecular targets for qCSC elimination are limited. Here, we present a protocol using the fluorescent dye PKH26 to isolate label-retaining qCSCs from colorectal cancer (CRC) patient-derived organoids (PDOs). We describe processing of organoids to single cells, followed by PKH26 labeling and FACS-based cell isolation. We then detail steps for functional assays and RNA sequencing. This protocol can also be applied to normal tissue-derived organoids. For complete details on the use and execution of this protocol, please refer to Regan et al. (2021).
Collapse
Affiliation(s)
- Joseph L. Regan
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin, 10117 Berlin, Germany
| |
Collapse
|
4
|
Disis ML, Cecil DL. Breast cancer vaccines for treatment and prevention. Breast Cancer Res Treat 2021; 191:481-489. [PMID: 34846625 DOI: 10.1007/s10549-021-06459-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 11/15/2021] [Indexed: 12/22/2022]
Abstract
Breast cancer is immunogenic and a variety of vaccines have been designed to boost immunity directed against the disease. The components of a breast cancer vaccine, the antigen, the delivery system, and the adjuvant, can have a significant impact on vaccine immunogenicity. There have been numerous immunogenic proteins identified in all subtypes of breast cancer. The majority of these antigens are weakly immunogenic nonmutated tumor-associated proteins. Mutated proteins and neoantigen epitopes are found only in a small minority of patients and are enriched in the triple negative subtype. Several vaccines have advanced to large randomized Phase II or Phase III clinical trials. None of these trials met their primary endpoint of either progression-free or overall survival. Despite these set-backs investigators have learned important lessons regarding the clinical application of breast cancer vaccines from the type of immune response needed for tumor eradication, Type I T-cell immunity, to the patient populations most likely to benefit from vaccination. Many therapeutic breast cancer vaccines are now being tested in combination with other forms of immune therapy or chemotherapy and radiation. Breast cancer vaccines as single agents are now studied in the context of the prevention of relapse or development of disease. Newer approaches are designing vaccines to prevent breast cancer by intercepting high-risk lesions such as ductal carcinoma in situ to limit the progression of these tumors to invasive cancer. There are also several efforts to develop vaccines for the primary prevention of breast cancer by targeting antigens expressed during breast cancer initiation.
Collapse
Affiliation(s)
- Mary L Disis
- Cancer Vaccine Institute, University of Washington, Seattle, WA, USA.
| | - Denise L Cecil
- Cancer Vaccine Institute, University of Washington, Seattle, WA, USA
| |
Collapse
|
5
|
Mare M, Colarossi L, Veschi V, Turdo A, Giuffrida D, Memeo L, Stassi G, Colarossi C. Cancer Stem Cell Biomarkers Predictive of Radiotherapy Response in Rectal Cancer: A Systematic Review. Genes (Basel) 2021; 12:genes12101502. [PMID: 34680897 PMCID: PMC8535834 DOI: 10.3390/genes12101502] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/17/2021] [Accepted: 09/23/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Rectal cancer (RC) is one of the most commonly diagnosed and particularly challenging tumours to treat due to its location in the pelvis and close proximity to critical genitourinary organs. Radiotherapy (RT) is recognised as a key component of therapeutic strategy to treat RC, promoting the downsizing and downstaging of large RCs in neoadjuvant settings, although its therapeutic effect is limited due to radioresistance. Evidence from experimental and clinical studies indicates that the likelihood of achieving local tumour control by RT depends on the complete eradication of cancer stem cells (CSC), a minority subset of tumour cells with stemness properties. METHODS A systematic literature review was conducted by querying two scientific databases (Pubmed and Scopus). The search was restricted to papers published from 2009 to 2021. RESULTS After assessing the quality and the risk of bias, a total of 11 studies were selected as they mainly focused on biomarkers predictive of RT-response in CSCs isolated from patients affected by RC. Specifically these studies showed that elevated levels of CD133, CD44, ALDH1, Lgr5 and G9a are associated with RT-resistance and poor prognosis. CONCLUSIONS This review aimed to provide an overview of the current scenario of in vitro and in vivo studies evaluating the biomarkers predictive of RT-response in CSCs derived from RC patients.
Collapse
Affiliation(s)
- Marzia Mare
- Medical Oncology Unit, Mediterranean Institute of Oncology, 95029 Viagrande, Italy; (M.M.); (D.G.)
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, 98122 Messina, Italy
| | - Lorenzo Colarossi
- Pathology Unit, Mediterranean Institute of Oncology, 95029 Viagrande, Italy; (L.C.); (L.M.); (C.C.)
| | - Veronica Veschi
- Department of Surgical Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, 90127 Palermo, Italy;
| | - Alice Turdo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy;
| | - Dario Giuffrida
- Medical Oncology Unit, Mediterranean Institute of Oncology, 95029 Viagrande, Italy; (M.M.); (D.G.)
| | - Lorenzo Memeo
- Pathology Unit, Mediterranean Institute of Oncology, 95029 Viagrande, Italy; (L.C.); (L.M.); (C.C.)
| | - Giorgio Stassi
- Department of Surgical Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, 90127 Palermo, Italy;
- Correspondence: ; Tel.: +39-091-2389-0813
| | - Cristina Colarossi
- Pathology Unit, Mediterranean Institute of Oncology, 95029 Viagrande, Italy; (L.C.); (L.M.); (C.C.)
| |
Collapse
|
6
|
Establishment and characterization of a novel cell line, NCC-DDLPS2-C1, derived from a patient with dedifferentiated liposarcoma. Hum Cell 2021; 34:990-997. [PMID: 33555519 DOI: 10.1007/s13577-021-00497-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 01/25/2021] [Indexed: 10/22/2022]
Abstract
Dedifferentiated liposarcoma (DDLPS) is a highly aggressive subtype of liposarcoma that is histologically a transition form between an atypical lipomatous tumor/well-differentiated liposarcoma and a non-lipogenic sarcoma. DDLPS is genetically characterized by a complex karyotype with copy number variations and genomic complexity. DDLPS has a poor prognosis, a high local recurrence rate, and refractory behaviors for chemotherapy and radiation, which indicate a requirement for a novel therapeutic strategy for better clinical outcomes. We report here, a novel DDLPS cell line (NCC-DDLPS2-C1) developed from a tumor tissue. NCC-DDLPS2-C1 cells showed an amplified 12q13-15 region and exhibited constant growth, spheroid formation, and invasion. High-throughput drug screening revealed distinct sensitivity between monolayer- and three-dimensional cells. Romidepsin and trabectedin especially showed high anti-proliferative effects in both culture methods of NCC-DDLPS2-C1. Thus, the NCC-DDLPS2-C1 cell line may serve as a useful resource for DDLPS studies.
Collapse
|
7
|
Endo H, Kondo J, Onuma K, Ohue M, Inoue M. Small subset of Wnt-activated cells is an initiator of regrowth in colorectal cancer organoids after irradiation. Cancer Sci 2020; 111:4429-4441. [PMID: 33043499 PMCID: PMC7734167 DOI: 10.1111/cas.14683] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 10/04/2020] [Accepted: 10/06/2020] [Indexed: 12/22/2022] Open
Abstract
Most colorectal cancers (CRCs) are differentiated adenocarcinomas, which maintain expression of both stemness and differentiation markers. This observation suggests that CRC cells could retain a regeneration system of normal cells upon injury. However, the role of stemness in cancer cell regeneration after irradiation is poorly understood. Here, we examined the effect of radiation on growth, stemness, and differentiation in organoids derived from differentiated adenocarcinomas. Following a sublethal dose of irradiation, proliferation and stemness markers, including Wnt target genes, were drastically reduced, but differentiation markers remained. After a static growth phase after high dose of radiation, regrowth foci appeared; these consisted of highly proliferating cells that expressed stem cell markers. Radiosensitivity and the ability to form foci differed among the cancer tissue‐originated spheroid (CTOS) lines examined and showed good correlation with in vivo radiation sensitivity. Pre‐treating organoids with histone deacetylase inhibitors increased radiation sensitivity; this increase was accompanied by the suppression of Wnt signal‐related gene expression. Accordingly, Wnt inhibitors increased organoid radiosensitivity. These results suggested that only a small subset of, but not all, cancer cells with high Wnt activity at the time of irradiation could give rise to foci formation. In conclusion, we established a radiation sensitivity assay using CRC organoids that could provide a novel platform for evaluating the effects of radiosensitizers on differentiated adenocarcinomas in CRC.
Collapse
Affiliation(s)
- Hiroko Endo
- Department of Biochemistry, Osaka International Cancer Institute, Osaka, Japan
| | - Jumpei Kondo
- Department of Biochemistry, Osaka International Cancer Institute, Osaka, Japan.,Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kunishige Onuma
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masayuki Ohue
- Department of Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Masahiro Inoue
- Department of Biochemistry, Osaka International Cancer Institute, Osaka, Japan.,Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|