1
|
Varlamova EG, Gudkov SV, Blinova EV, Blinov DS, Turovsky EA. Anticancer signal transduction pathways of selenium nanoparticles in mouse colorectal cancer model. Biochem Biophys Res Commun 2025; 769:151962. [PMID: 40347624 DOI: 10.1016/j.bbrc.2025.151962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/23/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025]
Abstract
Despite significant advances in the treatment of colon cancer, this disease is extremely common, often requiring serious surgery followed by long-term drug treatment. Colon and rectal cancer remain dangerous forms of cancer due to the high degree of metastasis. The development and study of the effectiveness of anticancer drugs based on nanoparticles is an urgent task of modern biomedicine. Of particular interest are attempts to move research from the in vitro level to the in vivo level of preclinical studies. In the presented study, mice were subcutaneously implanted with MC-38 cell line, a tumor was grown, and selenium nanoparticles (SeNPs) with a diameter of 100 nm obtained using the laser ablation method were administered intraperitoneally. Using morphometric measurements, it was found that injections of 1 μg/g or 10 μg/g SeNPs inhibited weight loss of mice during cancer development, reduced tumor size by 2-2.5 times, and suppressed metastasis by 1.5-3 times. Analysis of selenium levels in mouse blood, liver and tumor samples by atomic absorption spectrometry after the end of SeNPs treatment showed that the nanoparticles increased selenium levels in the blood and liver of mice without a significant dose-dependence, whereas in tumors a dose-dependent increase in selenium concentration was detected from the concentration of nanoparticles, with 10 μg/g SeNPs causing a more pronounced increase in selenium concentration. Using PCR and Western blot analysis, it was possible to establish that SeNPs injections led to an increase in the expression of genes encoding anti-inflammatory and anti-hypoxic proteins, but reduced the expression of antioxidant selenium-containing proteins and proteins responsible for the proliferation of cancer cells. Both concentrations of SeNPs led to similar effects, but increasing the concentration of nanoselenium to 10 μg/g affected the expression of a larger number of genes and the effects on expression were more "bright". Thus, the complex of presented experiments showed that injections of selenium nanoparticles in concentrations of 1 μg/g or 10 μg/g are capable to transport by the bloodstream and accumulating in the highest concentration in colon adenocarcinoma, compared with liver, which indicates the targeting of SeNPs in relation to tumors even without functionalization by specific molecules. As a result, there was a change in the expression patterns of genes and a number of proteins, and as a result, there was a decrease in tumor volume, normalization of mouse weight and maintenance of positive dynamics throughout the entire observation period.
Collapse
Affiliation(s)
- Elena G Varlamova
- Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", 142290, Pushchino, Russia.
| | - Sergey V Gudkov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilove st., 119991, Moscow, Russia
| | | | - Dmitrii S Blinov
- All-Union Research Center for Biological Active Compounds Safety, 23 Kirova St., 142450, StarajaKupavna, Russia
| | - Egor A Turovsky
- Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", 142290, Pushchino, Russia.
| |
Collapse
|
2
|
Cyr M, Chabaytah N, Babik J, Behmand B, St-Jean G, Enger SA. Establishing a standardized murine orthotopic intra-rectal model for the study of colorectal adenocarcinoma. J Gastrointest Oncol 2024; 15:2578-2587. [PMID: 39816036 PMCID: PMC11732354 DOI: 10.21037/jgo-24-515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 09/24/2024] [Indexed: 01/18/2025] Open
Abstract
Background Orthotopic models offer a more accurate representation of colorectal cancer (CRC) compared to subcutaneous models. Despite promising results from the reported intra-rectal models, establishing a standardized method for CRC research remains challenging due to model variability, hindering comprehensive studies on CRC pathogenesis and treatment modalities, such as brachytherapy. This study aimed to establish a standardized workflow for an orthotopic intra-rectal animal model to induce the growth of colorectal adenocarcinoma in male and female mice. Methods HT-29 colorectal adenocarcinoma cells were injected into the rectal mucosa of female (n=21) and male (n=26) non-obese diabetic severe combined immunodeficiency (NOD SCID) gamma (NSG) mice. Mice were placed on a 45° wedge elevating their pelvis for better visualization of the anus. Tumor growth and localization were monitored using a 7-T magnetic resonance imaging (MRI) scanner with rapid acquisition with relaxation echo (RARE) sequence at weeks 1, 2, and 3 post-cell instillation. Once tumors reached 5-8 mm in diameter, the mice were euthanized. Histopathology and immunohistochemical analyses confirmed the tumors' morphology, including necrosis, vascularity (CD-31) and apoptosis (cleaved caspase-3). Results There was a 92% and 95% tumor growth success rate in male and female mice, respectively. Tumors grew to 5-8 mm in diameter within ~20 days. No significant difference in tumor size was observed between genders. Tumor morphology was consistent across cases. Most tumors exhibited a lack of central blood vessels, accompanied by varying degrees of necrosis and apoptosis, whereas external portions were highly vascularized. Conclusions An orthotopic intra-rectal model was successfully developed. This model will be used in future studies to evaluate the efficacy of CRC treatments.
Collapse
Affiliation(s)
- Mélodie Cyr
- Medical Physics Unit, Department of Oncology, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Naim Chabaytah
- Medical Physics Unit, Department of Oncology, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Joud Babik
- Medical Physics Unit, Department of Oncology, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Behnaz Behmand
- Medical Physics Unit, Department of Oncology, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Guillaume St-Jean
- Department of Pathology and Microbiology, Faculty of Veterinary Medicine, University of Montreal, Montreal, QC, Canada
| | - Shirin A. Enger
- Medical Physics Unit, Department of Oncology, Faculty of Medicine, McGill University, Montreal, QC, Canada
- Research Institute of McGill University Health Center, Montreal, QC, Canada
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada
| |
Collapse
|
3
|
Blanco-Domínguez R, Mensurado S, Barros L, Carreira M, Silva-Santos B. An orthotopic metastatic xenograft model of colorectal cancer. Methods Cell Biol 2024; 190:119-132. [PMID: 39515876 DOI: 10.1016/bs.mcb.2024.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Colorectal cancer (CRC) presents a substantial global health challenge, prompting the necessity for the development and validation of preclinical models to enhance our comprehension and therapeutic interventions. Among the myriad of murine models available for CRC evaluation, orthotopic implantation via intercaecal microinjection stands out as a preferred method for replicating the intricate tumor microenvironment while ensuring uniformity and standardized applicability. In this study, we delineate a methodology addressing the required steps for tumor cell line selection and reporter transduction, animal model preparation, orthotopic tumor implantation, in vivo monitoring of tumor growth and metastasis formation. We comprehensively describe the generation of a xenograft murine model based on the intercaecal implantation of human GFP+/luciferase+ SW620 CRC cells, facilitating the evaluation of responses to pre-clinical human-based therapeutic approaches. The implementation of these standardized protocols promises to augment the reliability and reproducibility of preclinical studies, ultimately advancing our comprehension of CRC pathogenesis and guiding the development of innovative therapeutic strategies.
Collapse
Affiliation(s)
- Rafael Blanco-Domínguez
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.
| | - Sofia Mensurado
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.
| | - Leandro Barros
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Mariana Carreira
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Bruno Silva-Santos
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
4
|
De Vleeschauwer SI, van de Ven M, Oudin A, Debusschere K, Connor K, Byrne AT, Ram D, Rhebergen AM, Raeves YD, Dahlhoff M, Dangles-Marie V, Hermans ER. OBSERVE: guidelines for the refinement of rodent cancer models. Nat Protoc 2024; 19:2571-2596. [PMID: 38992214 DOI: 10.1038/s41596-024-00998-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 02/23/2024] [Indexed: 07/13/2024]
Abstract
Existing guidelines on the preparation (Planning Research and Experimental Procedures on Animals: Recommendations for Excellence (PREPARE)) and reporting (Animal Research: Reporting of In Vivo Experiments (ARRIVE)) of animal experiments do not provide a clear and standardized approach for refinement during in vivo cancer studies, resulting in the publication of generic methodological sections that poorly reflect the attempts made at accurately monitoring different pathologies. Compliance with the 3Rs guidelines has mainly focused on reduction and replacement; however, refinement has been harder to implement. The Oncology Best-practices: Signs, Endpoints and Refinements for in Vivo Experiments (OBSERVE) guidelines are the result of a European initiative supported by EurOPDX and INFRAFRONTIER, and aim to facilitate the refinement of studies using in vivo cancer models by offering robust and practical recommendations on approaches to research scientists and animal care staff. We listed cancer-specific clinical signs as a reference point and from there developed sets of guidelines for a wide variety of rodent models, including genetically engineered models and patient derived xenografts. In this Consensus Statement, we systematically and comprehensively address refinement and monitoring approaches during the design and execution of murine cancer studies. We elaborate on the appropriate preparation of tumor-initiating biologicals and the refinement of tumor-implantation methods. We describe the clinical signs to monitor associated with tumor growth, the appropriate follow-up of animals tailored to varying clinical signs and humane endpoints, and an overview of severity assessment in relation to clinical signs, implantation method and tumor characteristics. The guidelines provide oncology researchers clear and robust guidance for the refinement of in vivo cancer models.
Collapse
Affiliation(s)
| | - Marieke van de Ven
- Laboratory Animal Facility, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Anaïs Oudin
- NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Karlijn Debusschere
- Animal Core Facility VUB, Brussels, Belgium
- Core ARTH Animal Facilities, Medicine and Health Sciences Ghent University, Ghent, Belgium
| | - Kate Connor
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Annette T Byrne
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Doreen Ram
- Laboratory Animal Facility, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | | | | | - Maik Dahlhoff
- Institute of in vivo and in vitro Models, University of Veterinary Medicine Vienna, Vienna, Austria
| | | | - Els R Hermans
- Laboratory Animal Facility, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| |
Collapse
|
5
|
van Baarle L, De Simone V, Schneider L, Santhosh S, Abdurahiman S, Biscu F, Schneider R, Zanoletti L, Siqueira de Mello R, Verbandt S, Hu Z, Stakenborg M, Ke BJ, Stakenborg N, Salvador Laureano R, García-Reyes B, Henn J, Toma M, Vanmechelen M, Boeckxstaens G, De Smet F, Garg AD, Ibiza S, Tejpar S, Wehner S, Matteoli G. IL-1R signaling drives enteric glia-macrophage interactions in colorectal cancer. Nat Commun 2024; 15:6079. [PMID: 39030280 PMCID: PMC11271635 DOI: 10.1038/s41467-024-50438-2] [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: 09/16/2023] [Accepted: 07/11/2024] [Indexed: 07/21/2024] Open
Abstract
Enteric glia have been recently recognized as key components of the colonic tumor microenvironment indicating their potential role in colorectal cancer pathogenesis. Although enteric glia modulate immune responses in other intestinal diseases, their interaction with the colorectal cancer immune cell compartment remains unclear. Through a combination of single-cell and bulk RNA-sequencing, both in murine models and patients, here we find that enteric glia acquire an immunomodulatory phenotype by bi-directional communication with tumor-infiltrating monocytes. The latter direct a reactive enteric glial cell phenotypic and functional switch via glial IL-1R signaling. In turn, tumor glia promote monocyte differentiation towards pro-tumorigenic SPP1+ tumor-associated macrophages by IL-6 release. Enteric glia cell abundancy correlates with worse disease outcomes in preclinical models and colorectal cancer patients. Thereby, our study reveals a neuroimmune interaction between enteric glia and tumor-associated macrophages in the colorectal tumor microenvironment, providing insights into colorectal cancer pathogenesis.
Collapse
Affiliation(s)
- Lies van Baarle
- Laboratory of Mucosal Immunology, Department of Chronic Diseases and Metabolism (CHROMETA), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Veronica De Simone
- Laboratory of Mucosal Immunology, Department of Chronic Diseases and Metabolism (CHROMETA), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Linda Schneider
- Department of Surgery, University Hospital Bonn, Medical Faculty, Bonn, Germany
| | - Sneha Santhosh
- Laboratory of Mucosal Immunology, Department of Chronic Diseases and Metabolism (CHROMETA), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
- Department of Anatomy and Physiology, University of Melbourne, Parkville, VIC, Australia
| | - Saeed Abdurahiman
- Laboratory of Mucosal Immunology, Department of Chronic Diseases and Metabolism (CHROMETA), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Francesca Biscu
- Laboratory of Mucosal Immunology, Department of Chronic Diseases and Metabolism (CHROMETA), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK
| | - Reiner Schneider
- Department of Surgery, University Hospital Bonn, Medical Faculty, Bonn, Germany
| | - Lisa Zanoletti
- Laboratory of Mucosal Immunology, Department of Chronic Diseases and Metabolism (CHROMETA), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| | - Renata Siqueira de Mello
- Laboratory of Mucosal Immunology, Department of Chronic Diseases and Metabolism (CHROMETA), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Sara Verbandt
- Digestive Oncology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Zedong Hu
- Digestive Oncology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Michelle Stakenborg
- Laboratory of Mucosal Immunology, Department of Chronic Diseases and Metabolism (CHROMETA), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Bo-Jun Ke
- Laboratory of Mucosal Immunology, Department of Chronic Diseases and Metabolism (CHROMETA), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Nathalie Stakenborg
- Laboratory for Intestinal Neuro-Immune Interaction, Department of Chronic Diseases and Metabolism (CHROMETA), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Raquel Salvador Laureano
- Cell Stress and Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Balbina García-Reyes
- Department of Surgery, University Hospital Bonn, Medical Faculty, Bonn, Germany
- Mildred Scheel School of Oncology, Aachen Bonn Cologne Düsseldorf (MSSO ABCD), University Hospital Bonn, Medical Faculty, Bonn, Germany
| | - Jonas Henn
- Department of Surgery, University Hospital Bonn, Medical Faculty, Bonn, Germany
| | - Marieta Toma
- Department of Pathology, University Hospital Bonn, Medical Faculty, Bonn, Germany
| | - Maxime Vanmechelen
- Translational Cell and Tissue Research Unit, Department of Imaging & Pathology, Laboratory for Precision Cancer Medicine, KU Leuven, Leuven, Belgium
- Leuven Institute for Single-Cell Omics (LISCO), KU Leuven, Leuven, Belgium
| | - Guy Boeckxstaens
- Laboratory for Intestinal Neuro-Immune Interaction, Department of Chronic Diseases and Metabolism (CHROMETA), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Frederik De Smet
- Translational Cell and Tissue Research Unit, Department of Imaging & Pathology, Laboratory for Precision Cancer Medicine, KU Leuven, Leuven, Belgium
- Leuven Institute for Single-Cell Omics (LISCO), KU Leuven, Leuven, Belgium
| | - Abhishek D Garg
- Cell Stress and Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Sales Ibiza
- Laboratory of Cell Biology & Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | - Sabine Tejpar
- Digestive Oncology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Sven Wehner
- Department of Surgery, University Hospital Bonn, Medical Faculty, Bonn, Germany.
| | - Gianluca Matteoli
- Laboratory of Mucosal Immunology, Department of Chronic Diseases and Metabolism (CHROMETA), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium.
- Leuven Institute for Single-Cell Omics (LISCO), KU Leuven, Leuven, Belgium.
| |
Collapse
|
6
|
Pennel K, Dutton L, Melissourgou-Syka L, Roxburgh C, Birch J, Edwards J. Novel radiation and targeted therapy combinations for improving rectal cancer outcomes. Expert Rev Mol Med 2024; 26:e14. [PMID: 38623751 PMCID: PMC11140547 DOI: 10.1017/erm.2024.15] [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: 06/07/2023] [Revised: 01/29/2024] [Accepted: 03/22/2024] [Indexed: 04/17/2024]
Abstract
Neoadjuvant radiotherapy (RT) is commonly used as standard treatment for rectal cancer. However, response rates are variable and survival outcomes remain poor, highlighting the need to develop new therapeutic strategies. Research is focused on identifying novel methods for sensitising rectal tumours to RT to enhance responses and improve patient outcomes. This can be achieved through harnessing tumour promoting effects of radiation or preventing development of radio-resistance in cancer cells. Many of the approaches being investigated involve targeting the recently published new dimensions of cancer hallmarks. This review article will discuss key radiation and targeted therapy combination strategies being investigated in the rectal cancer setting, with a focus on exploitation of mechanisms which target the hallmarks of cancer.
Collapse
Affiliation(s)
- Kathryn Pennel
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, G61 1BD, UK
| | - Louise Dutton
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, G61 1BD, UK
| | - Lydia Melissourgou-Syka
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, G61 1BD, UK
- CRUK Scotland Institute, Glasgow, G611BD, UK
| | - Campbell Roxburgh
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, G61 1BD, UK
- Academic Unit of Surgery, Glasgow Royal Infirmary, University of Glasgow, Glasgow, G4 0SF, UK
| | - Joanna Birch
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, G61 1BD, UK
| | - Joanne Edwards
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, G61 1BD, UK
| |
Collapse
|
7
|
Thoma OM, Naschberger E, Kubánková M, Larafa I, Kramer V, Menchicchi B, Merkel S, Britzen-Laurent N, Jefremow A, Grützmann R, Koop K, Neufert C, Atreya R, Guck J, Stürzl M, Neurath MF, Waldner MJ. p21 Prevents the Exhaustion of CD4 + T Cells Within the Antitumor Immune Response Against Colorectal Cancer. Gastroenterology 2024; 166:284-297.e11. [PMID: 37734420 DOI: 10.1053/j.gastro.2023.09.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 08/30/2023] [Accepted: 09/05/2023] [Indexed: 09/23/2023]
Abstract
BACKGROUND & AIMS T cells are crucial for the antitumor response against colorectal cancer (CRC). T-cell reactivity to CRC is nevertheless limited by T-cell exhaustion. However, molecular mechanisms regulating T-cell exhaustion are only poorly understood. METHODS We investigated the functional role of cyclin-dependent kinase 1a (Cdkn1a or p21) in cluster of differentiation (CD) 4+ T cells using murine CRC models. Furthermore, we evaluated the expression of p21 in patients with stage I to IV CRC. In vitro coculture models were used to understand the effector function of p21-deficient CD4+ T cells. RESULTS We observed that the activation of cell cycle regulator p21 is crucial for CD4+ T-cell cytotoxic function and that p21 deficiency in type 1 helper T cells (Th1) leads to increased tumor growth in murine CRC. Similarly, low p21 expression in CD4+ T cells infiltrated into tumors of CRC patients is associated with reduced cancer-related survival. In mouse models of CRC, p21-deficient Th1 cells show signs of exhaustion, where an accumulation of effector/effector memory T cells and CD27/CD28 loss are predominant. Immune reconstitution of tumor-bearing Rag1-/- mice using ex vivo-treated p21-deficient T cells with palbociclib, an inhibitor of cyclin-dependent kinase 4/6, restored cytotoxic function and prevented exhaustion of p21-deficient CD4+ T cells as a possible concept for future immunotherapy of human disease. CONCLUSIONS Our data reveal the importance of p21 in controlling the cell cycle and preventing exhaustion of Th1 cells. Furthermore, we unveil the therapeutic potential of cyclin-dependent kinase inhibitors such as palbociclib to reduce T-cell exhaustion for future treatment of patients with colorectal cancer.
Collapse
Affiliation(s)
- Oana-Maria Thoma
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; German Center for Immunotherapy, Deutsches Zentrum Immuntherapie (DZI), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany.
| | - Elisabeth Naschberger
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany; Division of Molecular and Experimental Surgery, Department of Surgery, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Markéta Kubánková
- Max Planck Institute for the Science of Light & Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
| | - Imen Larafa
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; German Center for Immunotherapy, Deutsches Zentrum Immuntherapie (DZI), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
| | - Viktoria Kramer
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; German Center for Immunotherapy, Deutsches Zentrum Immuntherapie (DZI), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
| | - Bianca Menchicchi
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; German Center for Immunotherapy, Deutsches Zentrum Immuntherapie (DZI), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
| | - Susanne Merkel
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany; Department of Surgery, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Nathalie Britzen-Laurent
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany; Department of Surgery, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - André Jefremow
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; German Center for Immunotherapy, Deutsches Zentrum Immuntherapie (DZI), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
| | - Robert Grützmann
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany; Department of Surgery, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Kristina Koop
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; German Center for Immunotherapy, Deutsches Zentrum Immuntherapie (DZI), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
| | - Clemens Neufert
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; German Center for Immunotherapy, Deutsches Zentrum Immuntherapie (DZI), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
| | - Raja Atreya
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; German Center for Immunotherapy, Deutsches Zentrum Immuntherapie (DZI), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
| | - Jochen Guck
- Max Planck Institute for the Science of Light & Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
| | - Michael Stürzl
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany; Division of Molecular and Experimental Surgery, Department of Surgery, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Markus F Neurath
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; German Center for Immunotherapy, Deutsches Zentrum Immuntherapie (DZI), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
| | - Maximilian J Waldner
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; German Center for Immunotherapy, Deutsches Zentrum Immuntherapie (DZI), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany; Erlangen Graduate School in Advanced Optical Technologies (SAOT), Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
8
|
Melissourgou-Syka L, Gillespie MA, O'Cathail SM, Sansom OJ, Steele CW, Roxburgh CSD. A Review of Scheduling Strategies for Radiotherapy and Immune Checkpoint Inhibition in Locally Advanced Rectal Cancer. JOURNAL OF IMMUNOTHERAPY AND PRECISION ONCOLOGY 2023; 6:187-197. [PMID: 38143952 PMCID: PMC10734391 DOI: 10.36401/jipo-23-10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 07/07/2023] [Accepted: 07/15/2023] [Indexed: 12/26/2023]
Abstract
Colorectal cancer (CRC) is the third most common malignancy across the globe and, despite advances in treatment strategies, survival rates remain low. Rectal cancer (RC) accounts for most of these cases, and traditional management strategies for advanced disease include total neoadjuvant therapy (TNT) with chemoradiotherapy followed by curative surgery. Unfortunately, approximately 10-15% of patients have no response to treatment or have recurrence at a short interval following radiotherapy. The introduction of immunotherapy in the form of immune checkpoint blockade (ICB) in metastatic colorectal cancer has improved clinical outcomes, yet most patients with RC present with microsatellite stable disease, which lacks the immune-rich microenvironment where ICB is most effective. There is evidence that combining radiotherapy with ICB can unlock the mechanisms that drive resistance in patients; however, the sequencing of these therapies is still debated. This review offers a comprehensive overview of clinical trials and preclinical models that use radiotherapy-immunotherapy combinations in RC in an attempt to extrapolate the ideal sequencing of the two treatment modalities. The results highlight the dearth of evidence to answer the question of whether ICB should be given before, during, or after radiotherapy, yet it is suggested that improving the relevance of our preclinical models will provide a platform with higher translational value and will lead to appropriate clinical trial designs.
Collapse
Affiliation(s)
- Lydia Melissourgou-Syka
- School of Cancer Sciences, University of Glasgow, Glasgow, Scotland
- CRUK Beatson Institute, Glasgow, Scotland
| | | | - Sean M. O'Cathail
- School of Cancer Sciences, University of Glasgow, Glasgow, Scotland
- Beatson West of Scotland Cancer Centre, Glasgow, Scotland
| | - Owen J. Sansom
- School of Cancer Sciences, University of Glasgow, Glasgow, Scotland
- CRUK Beatson Institute, Glasgow, Scotland
| | - Colin W. Steele
- School of Cancer Sciences, University of Glasgow, Glasgow, Scotland
- CRUK Beatson Institute, Glasgow, Scotland
- Academic Unit of Surgery, Glasgow Royal Infirmary, Glasgow, Scotland
| | - Campbell S. D. Roxburgh
- School of Cancer Sciences, University of Glasgow, Glasgow, Scotland
- Academic Unit of Surgery, Glasgow Royal Infirmary, Glasgow, Scotland
| |
Collapse
|
9
|
Shields NJ, Peyroux EM, Ferguson AL, Steain M, Neumann S, Young SL. Late-stage MC38 tumours recapitulate features of human colorectal cancer - implications for appropriate timepoint selection in preclinical studies. Front Immunol 2023; 14:1152035. [PMID: 37153625 PMCID: PMC10160415 DOI: 10.3389/fimmu.2023.1152035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 04/10/2023] [Indexed: 05/10/2023] Open
Abstract
Anti-tumour T cell responses play a crucial role in controlling the progression of colorectal cancer (CRC), making this disease a promising candidate for immunotherapy. However, responses to immune-targeted therapies are currently limited to subpopulations of patients and specific types of cancer. Clinical studies have therefore focussed on identifying biomarkers that predict immunotherapy responses and elucidating the immunological landscapes of different cancers. Meanwhile, our understanding of how preclinical tumour models resemble human disease has fallen behind, despite their crucial role in immune-targeted drug development. A deeper understanding of these models is therefore needed to improve the development of immunotherapies and the translation of findings made in these systems. MC38 colon adenocarcinoma is a widely used preclinical model, yet how it recapitulates human colorectal cancer remains poorly defined. This study investigated the tumour-T cell immune landscape of MC38 tumours using histology, immunohistochemistry, and flow cytometry. We demonstrate that early-stage tumours exhibit a nascent TME, lacking important immune-resistance mechanisms of clinical interest, while late-stage tumours exhibit a mature TME resembling human tumours, with desmoplasia, T cell exhaustion, and T cell exclusion. Consequently, these findings clarify appropriate timepoint selection in the MC38 model when investigating both immunotherapies and mechanisms that contribute to immunotherapy resistance. Overall, this study provides a valuable resource that will enable appropriate application of the MC38 model and expedite the development and clinical translation of new immunotherapies.
Collapse
Affiliation(s)
- Nicholas J. Shields
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Department of Pathology, Otago Medical School, University of Otago, Dunedin, New Zealand
| | - Estelle M. Peyroux
- Department of Pathology, Otago Medical School, University of Otago, Dunedin, New Zealand
| | - Angela L. Ferguson
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Liver Injury and Cancer Program, Centenary Institute, Sydney, NSW, Australia
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Megan Steain
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Silke Neumann
- Department of Pathology, Otago Medical School, University of Otago, Dunedin, New Zealand
| | - Sarah L. Young
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Faculty of Science, University of Canterbury, Christchurch, New Zealand
| |
Collapse
|
10
|
Hotta R, Pan W, Bhave S, Nagy N, Stavely R, Ohkura T, Krishnan K, de Couto G, Myers R, Rodriguez-Borlado L, Burns AJ, Goldstein AM. Isolation, Expansion, and Endoscopic Delivery of Autologous Enteric Neuronal Stem Cells in Swine. Cell Transplant 2023; 32:9636897231215233. [PMID: 38049927 PMCID: PMC10697035 DOI: 10.1177/09636897231215233] [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: 06/24/2023] [Revised: 09/22/2023] [Accepted: 11/01/2023] [Indexed: 12/06/2023] Open
Abstract
The enteric nervous system (ENS) is an extensive network of neurons and glia within the wall of the gastrointestinal (GI) tract that regulates many essential GI functions. Consequently, disorders of the ENS due to developmental defects, inflammation, infection, or age-associated neurodegeneration lead to serious neurointestinal diseases. Despite the prevalence and severity of these diseases, effective treatments are lacking as they fail to directly address the underlying pathology. Neuronal stem cell therapy represents a promising approach to treating diseases of the ENS by replacing the absent or injured neurons, and an autologous source of stem cells would be optimal by obviating the need for immunosuppression. We utilized the swine model to address key questions concerning cell isolation, delivery, engraftment, and fate in a large animal relevant to human therapy. We successfully isolated neural stem cells from a segment of small intestine resected from 1-month-old swine. Enteric neuronal stem cells (ENSCs) were expanded as neurospheres that grew optimally in low-oxygen (5%) culture conditions. Enteric neuronal stem cells were labeled by lentiviral green fluorescent protein (GFP) transduction, then transplanted into the same swine from which they had been harvested. Endoscopic ultrasound was then utilized to deliver the ENSCs (10,000-30,000 neurospheres per animal) into the rectal wall. At 10 and 28 days following injection, autologously derived ENSCs were found to have engrafted within rectal wall, with neuroglial differentiation and no evidence of ectopic spreading. These findings strongly support the feasibility of autologous cell isolation and delivery using a clinically useful and minimally invasive technique, bringing us closer to first-in-human ENSC therapy for neurointestinal diseases.
Collapse
Affiliation(s)
- Ryo Hotta
- Department of Pediatric Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Weikang Pan
- Department of Pediatric Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Sukhada Bhave
- Department of Pediatric Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Nandor Nagy
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Rhian Stavely
- Department of Pediatric Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Takahiro Ohkura
- Department of Pediatric Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Kumar Krishnan
- Division of Gastroenterology, Department of Internal Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Geoffrey de Couto
- Gastrointestinal Drug Discovery Unit, Takeda Development Center Americas, Inc., Cambridge, MA, USA
| | - Richard Myers
- Gastrointestinal Drug Discovery Unit, Takeda Development Center Americas, Inc., Cambridge, MA, USA
| | - Luis Rodriguez-Borlado
- Gastrointestinal Drug Discovery Unit, Takeda Development Center Americas, Inc., Cambridge, MA, USA
| | - Alan J. Burns
- Gastrointestinal Drug Discovery Unit, Takeda Development Center Americas, Inc., Cambridge, MA, USA
- Stem Cells and Regenerative Medicine, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Allan M. Goldstein
- Department of Pediatric Surgery, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
11
|
Hu LP, Huang W, Wang X, Xu C, Qin WT, Li D, Tian G, Li Q, Zhou Y, Chen S, Nie HZ, Hao Y, Song J, Zhang XL, Sundquist J, Sundquist K, Li J, Jiang SH, Zhang ZG, Ji J. Terbinafine prevents colorectal cancer growth by inducing dNTP starvation and reducing immune suppression. Mol Ther 2022; 30:3284-3299. [PMID: 35765243 PMCID: PMC9552806 DOI: 10.1016/j.ymthe.2022.06.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 06/07/2022] [Accepted: 06/23/2022] [Indexed: 12/31/2022] Open
Abstract
Existing evidence indicates that gut fungal dysbiosis might play a key role in the pathogenesis of colorectal cancer (CRC). We sought to explore whether reversing the fungal dysbiosis by terbinafine, an approved antifungal drug, might inhibit the development of CRC. A population-based study from Sweden identified a total of 185 patients who received terbinafine after their CRC diagnosis and found that they had a decreased risk of death (hazard ratio = 0.50) and metastasis (hazard ratio = 0.44) compared with patients without terbinafine administration. In multiple mouse models of CRC, administration of terbinafine decreased the fungal load, the fungus-induced myeloid-derived suppressor cell (MDSC) expansion, and the tumor burden. Fecal microbiota transplantation from mice without terbinafine treatment reversed MDSC infiltration and partially restored tumor proliferation. Mechanistically, terbinafine directly impaired tumor cell proliferation by reducing the ratio of nicotinamide adenine dinucleotide phosphate (NADP+) to reduced form of nicotinamide adenine dinucleotide phosphate (NADPH), suppressing the activity of glucose-6-phosphate dehydrogenase (G6PD), resulting in nucleotide synthesis disruption, deoxyribonucleotide (dNTP) starvation, and cell-cycle arrest. Collectively, terbinafine can inhibit CRC by reversing fungal dysbiosis, suppressing tumor cell proliferation, inhibiting fungus-induced MDSC infiltration, and restoring antitumor immune response.
Collapse
Affiliation(s)
- Li-Peng Hu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Wuqing Huang
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, FuZhou 350108, China
| | - Xu Wang
- Department of Radiation Oncology, Institute of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang 212013, China
| | - Chunjie Xu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Wei-Ting Qin
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Dongxue Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Guangang Tian
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Qing Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yaoqi Zhou
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Suyuan Chen
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Hui-Zhen Nie
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yujun Hao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jian Song
- Department of Radiation Oncology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Xue-Li Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jan Sundquist
- Center for Primary Health Care Research, Department of Clinical Sciences, Lund University, Malmö 20502, Sweden; Department of Family Medicine and Community Health, Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kristina Sundquist
- Center for Primary Health Care Research, Department of Clinical Sciences, Lund University, Malmö 20502, Sweden; Department of Family Medicine and Community Health, Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jun Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Shu-Heng Jiang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Zhi-Gang Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Jianguang Ji
- Center for Primary Health Care Research, Department of Clinical Sciences, Lund University, Malmö 20502, Sweden.
| |
Collapse
|
12
|
Modeling Colorectal Cancer Progression Reveals Niche-Dependent Clonal Selection. Cancers (Basel) 2022; 14:cancers14174260. [PMID: 36077793 PMCID: PMC9454531 DOI: 10.3390/cancers14174260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 08/22/2022] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) is among the deadliest cancers worldwide, with metastasis being the main cause of patient mortality. During CRC progression the complex tumor ecosystem changes in its composition at virtually every stage. However, clonal dynamics and associated niche-dependencies at these stages are unknown. Hence, it is of importance to utilize models that faithfully recapitulate human CRC to define its clonal dynamics. We used an optical barcoding approach in mouse-derived organoids (MDOs) that revealed niche-dependent clonal selection. Our findings highlight that clonal selection is controlled by a site-specific niche, which critically contributes to cancer heterogeneity and has implications for therapeutic intervention.
Collapse
|
13
|
Yuan C, Zhao X, Wangmo D, Alshareef D, Gates TJ, Subramanian S. Tumor models to assess immune response and tumor-microbiome interactions in colorectal cancer. Pharmacol Ther 2022; 231:107981. [PMID: 34480964 PMCID: PMC8844062 DOI: 10.1016/j.pharmthera.2021.107981] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 08/20/2021] [Accepted: 08/24/2021] [Indexed: 02/07/2023]
Abstract
Despite significant advances over the past 2 decades in preventive screening and therapy aimed at improving patient survival, colorectal cancer (CRC) remains the second most common cause of cancer death in the United States. The average 5-year survival rate of CRC patients with positive regional lymph nodes is only 40%, while less than 5% of patients with distant metastases survive beyond 5 years. There is a critical need to develop novel therapies that can improve overall survival in patients with poor prognoses, particularly since 60% of them are diagnosed at an advanced stage. Pertinently, immune checkpoint blockade therapy has dramatically changed how we treat CRC patients with microsatellite-instable high tumors. Furthermore, accumulating evidence shows that changes in gut microbiota are associated with the regulation of host antitumor immune response and cancer progression. Appropriate animal models are essential to deciphering the complex mechanisms of host antitumor immune response and tumor-gut microbiome metabolic interactions. Here, we discuss various mouse models of colorectal cancer that are developed to address key questions on tumor immune response and tumor-microbiota interactions. These CRC models will also serve as resourceful tools for effective preclinical studies.
Collapse
Affiliation(s)
- Ce Yuan
- Department of Surgery, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Xianda Zhao
- Department of Surgery, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Dechen Wangmo
- Department of Surgery, University of Minnesota, Minneapolis, MN 55455, United States of America; Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Duha Alshareef
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Travis J Gates
- Department of Surgery, University of Minnesota, Minneapolis, MN 55455, United States of America; Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Subbaya Subramanian
- Department of Surgery, University of Minnesota, Minneapolis, MN 55455, United States of America; Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, United States of America; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, United States of America.
| |
Collapse
|
14
|
Colonoscopy-based intramucosal transplantation of cancer cells for mouse modeling of colon cancer and lung metastasis. Biotechniques 2021; 71:456-464. [PMID: 34392706 PMCID: PMC8867397 DOI: 10.2144/btn-2020-0172] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The conventional orthotopic/xenograft models or genetically engineered murine models of colon cancer (CRC) are limited in their scope for a true understanding of tumor growth, progression and eventual metastasis in its natural microenvironment. In the currently used murine models of CRC metastasis, the metastasis occurs primarily in the liver, though lung metastasis accounts for a significant proportion of CRC metastasis. There is an urgent need for a murine model of CRC, which not only allows tumor progression in the colonic mucosa but also metastasis of the lung. The authors describe a minimally invasive murine model of colon cancer progression that may be ideal for a wide range of applications, including evaluating gene function, microenvironment, cancer metastasis and therapeutic translational research.
Collapse
|
15
|
Inducible mouse models of colon cancer for the analysis of sporadic and inflammation-driven tumor progression and lymph node metastasis. Nat Protoc 2020; 16:61-85. [PMID: 33318692 DOI: 10.1038/s41596-020-00412-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 09/17/2020] [Indexed: 12/30/2022]
Abstract
Despite advances in the detection and therapy of colorectal cancer (CRC) in recent years, CRC has remained a major challenge in clinical practice. Although alternative methods for modeling CRC have been developed, animal models of CRC remain helpful when analyzing molecular aspects of pathogenesis and are often used to perform preclinical in vivo studies of potential therapeutics. This protocol updates our protocol published in 2007, which provided an azoxymethane (AOM)-based setup for investigations into sporadic (Step 5A) and, when combined with dextran sodium sulfate (Step 5B), inflammation-associated tumor growth. This update also extends the applications beyond those of the original protocol by including an option in which AOM is serially applied to mice with p53 deficiency in the intestinal epithelium (Step 5C). In this model, the combination of p53 deficiency and AOM promotes tumor development, including growth of invasive cancers and lymph node metastasis. It also provides details on analysis of colorectal tumor growth and metastasis, including analysis of partial epithelial-to-mesenchymal transition, cell isolation and co-culture studies, high-resolution mini-endoscopy, light-sheet fluorescence microscopy and micro-CT imaging in mice. The target audience for our protocol is researchers who plan in vivo studies to address mechanisms influencing sporadic or inflammation-driven tumor development, including the analysis of local invasiveness and lymph node metastasis. It is suitable for preclinical in vivo testing of novel drugs and other interventional strategies for clinical translation, plus the evaluation of emerging imaging devices/modalities. It can be completed within 24 weeks (using Step 5A/C) or 10 weeks (using Step 5B).
Collapse
|
16
|
Establishment of an Endoscopy-Guided Minimally Invasive Orthotopic Mouse Model of Colorectal Cancer. Cancers (Basel) 2020; 12:cancers12103007. [PMID: 33081354 PMCID: PMC7650778 DOI: 10.3390/cancers12103007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 10/12/2020] [Accepted: 10/15/2020] [Indexed: 02/08/2023] Open
Abstract
Simple Summary Open orthotopic mouse models of colorectal cancer have disadvantages such as the requirement for advanced surgical skills or the trauma caused by laparotomy. To overcome these limitations, this study aimed to evaluate the establishment of an endoscopy-guided minimally invasive model without laparotomy. Different concentrations of the murine CRC cell lines CT26 and MC38 were endoscopically injected into the colorectal wall of BALB/C and C57BL/6J mice, respectively. Consistent tumor growth with the presence of tumor-infiltrating lymphocytes, lympho-vascular invasion, and early spontaneous lymph node, peritoneal, and hepatic metastases were observed. Analysis of the learning curve demonstrated that this model is easy to learn and quick to establish. It enables intra-individual follow-up endoscopies, and features tumors to study mechanisms of metastasis and the interaction with the immune system. The application of specific cell lines and concentrations enables a controlled local tumor growth and metastatic formation within short observation periods. Abstract Open orthotopic mouse models of colorectal cancer have disadvantages such as the requirement for advanced surgical skills or the trauma caused by laparotomy. To overcome these drawbacks, this study aimed to evaluate the establishment of a minimally invasive model using murine colonoscopy. CT26 and MC38 CRC cells of different concentrations were injected into BALB/C and C57BL/6J mice, respectively. Follow-up endoscopies were performed to assign an endoscopic score to tumor growth. Gross autopsy, histologic and immuno-histochemical evaluation, and immune scoring were performed. To describe the learning curve of the procedures, a performance score was given. Local tumor growth with colorectal wall infiltration, luminal ulceration, the presence of tumor-infiltrating lymphocytes, lympho-vascular invasion, and early spontaneous lymph node, peritoneal, and hepatic metastases were observed. The tumors showed cytoplasmic immuno-staining for CK20. Compared to the MC38/C57BL/6J model, tumorigenicity and immunogenicity of the CT26/BALB/C model were higher. Tumor volume correlated with the endoscopic score. This endoscopy-guided orthotopic mouse model is easy to learn and quick to establish. It features early metastasis and enables the study of interactions with the immune system. When specific cell concentrations and cell lines are applied, controlled local tumor growth and metastasis can be achieved within short observation periods.
Collapse
|
17
|
Shaashua L, Mayer S, Lior C, Lavon H, Novoselsky A, Scherz-Shouval R. Stromal Expression of the Core Clock Gene Period 2 Is Essential for Tumor Initiation and Metastatic Colonization. Front Cell Dev Biol 2020; 8:587697. [PMID: 33123539 PMCID: PMC7573548 DOI: 10.3389/fcell.2020.587697] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 09/03/2020] [Indexed: 12/11/2022] Open
Abstract
The circadian clock regulates diverse physiological processes by maintaining a 24-h gene expression pattern. Genetic and environmental cues that disrupt normal clock rhythms can lead to cancer, yet the extent to which this effect is controlled by the cancer cells versus non-malignant cells in the tumor microenvironment (TME) is not clear. Here we set out to address this question, by selective manipulation of circadian clock genes in the TME. In two different mouse models of cancer we find that expression of the core clock gene Per2 in the TME is crucial for tumor initiation and metastatic colonization, whereas another core gene, Per1, is dispensable. We further show that loss of Per2 in the TME leads to significant transcriptional changes in response to cancer cell introduction. These changes may contribute to a tumor-suppressive microenvironment. Thus, our work unravels an unexpected protumorigenic role for the core clock gene Per2 in the TME, with potential implications for therapeutic dosing strategies and treatment regimens.
Collapse
Affiliation(s)
- Lee Shaashua
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Shimrit Mayer
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Chen Lior
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Hagar Lavon
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Alexander Novoselsky
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Ruth Scherz-Shouval
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
18
|
Machado VF, Parra RS, Leite CA, Minto SB, Cunha TM, Cunha FDQ, Garcia SB, Feitosa MR, da Rocha JJR, Feres O. Experimental Model of Rectal Carcinogenesis Induced by N-Methyl-N-Nitrosoguanidine in Mice with Endoscopic Evaluation. Int J Med Sci 2020; 17:2505-2510. [PMID: 33029093 PMCID: PMC7532479 DOI: 10.7150/ijms.48231] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 08/10/2020] [Indexed: 11/26/2022] Open
Abstract
Background and purpose: The discovery of chemical substances with carcinogenic properties has allowed the development of several experimental models of colorectal cancer (CRC). Classically, experimental models of CRC in mice have been evaluated through clinical or serial euthanasia. The present study aims to investigate the role of low endoscopy in the analysis of carcinogenesis induced by N-methyl-N'-nitro-N-nitrosoguanidine (MNNG). Methods: Thirty C57BL6 mice were divided into two groups: a control group with fifteen animals that underwent rectal instillation of saline solution on day 0 and a carcinogen group with fifteen animals that underwent a 100 mg/kg MNNG rectal instillation on day 0. In both groups, low endoscopies were performed on weeks 4 and 8. We used a validated endoscopic scoring system to evaluate the severity of colitis and colorectal tumor. Euthanasia was carried out at week 12. Results: We observed higher inflammation scores (p <0.001) and a higher number of tumors (p <0.05) in the MNNG group than the control group, both at weeks 4 and 8. A worsening of inflammation scores from the first to the second endoscopy was also noticeable in the MNNG group. There were no bowel perforations related to the procedure, and there was one death in the control group. Conclusion: Low endoscopy in experimental animals allows safe macroscopic evaluation of colorectal carcinogenesis without the need for euthanasia.
Collapse
Affiliation(s)
- Vanessa Foresto Machado
- Department of Surgery and Anatomy, School of Medicine of Ribeirão Preto, University of São Paulo, Brazil
| | - Rogerio Serafim Parra
- Department of Surgery and Anatomy, School of Medicine of Ribeirão Preto, University of São Paulo, Brazil
| | - Caio Abner Leite
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo, Brazil
| | - Stefania Bovo Minto
- Pathology and Legal Medicine Department, School of Medicine of Ribeirão Preto, University of São Paulo, Brazil
| | - Thiago Mattar Cunha
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo, Brazil
| | | | - Sergio Britto Garcia
- Pathology and Legal Medicine Department, School of Medicine of Ribeirão Preto, University of São Paulo, Brazil
| | - Marley Ribeiro Feitosa
- Department of Surgery and Anatomy, School of Medicine of Ribeirão Preto, University of São Paulo, Brazil
| | | | - Omar Feres
- Department of Surgery and Anatomy, School of Medicine of Ribeirão Preto, University of São Paulo, Brazil
| |
Collapse
|
19
|
Heichler C, Scheibe K, Schmied A, Geppert CI, Schmid B, Wirtz S, Thoma OM, Kramer V, Waldner MJ, Büttner C, Farin HF, Pešić M, Knieling F, Merkel S, Grüneboom A, Gunzer M, Grützmann R, Rose-John S, Koralov SB, Kollias G, Vieth M, Hartmann A, Greten FR, Neurath MF, Neufert C. STAT3 activation through IL-6/IL-11 in cancer-associated fibroblasts promotes colorectal tumour development and correlates with poor prognosis. Gut 2020; 69:1269-1282. [PMID: 31685519 DOI: 10.1136/gutjnl-2019-319200] [Citation(s) in RCA: 213] [Impact Index Per Article: 42.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 09/22/2019] [Accepted: 10/08/2019] [Indexed: 01/10/2023]
Abstract
OBJECTIVE Cancer-associated fibroblasts (CAFs) influence the tumour microenvironment and tumour growth. However, the role of CAFs in colorectal cancer (CRC) development is incompletely understood. DESIGN We quantified phosphorylation of STAT3 (pSTAT3) expression in CAFs of human colon cancer tissue using a tissue microarray (TMA) of 375 patients, immunofluorescence staining and digital pathology. To investigate the functional role of CAFs in CRC, we took advantage of two murine models of colorectal neoplasia and advanced imaging technologies. In loss-of-function and gain-of-function experiments, using genetically modified mice with collagen type VI (COLVI)-specific signal transducer and activator of transcription 3 (STAT3) targeting, we evaluated STAT3 signalling in fibroblasts during colorectal tumour development. We performed a comparative gene expression profiling by whole genome RNA-sequencing of fibroblast subpopulations (COLVI+ vs COLVI-) on STAT3 activation (IL-6 vs IL-11). RESULTS The analysis of pSTAT3 expression in CAFs of human TMAs revealed a negative correlation of increased stromal pSTAT3 expression with the survival of colon cancer patients. In the loss-of-function and gain-of-function approach, we found a critical role of STAT3 activation in fibroblasts in driving colorectal tumourigenesis in vivo. With different imaging technologies, we detected an expansion of activated fibroblasts in colorectal neoplasias. Comparative gene expression profiling of fibroblast subpopulations on STAT3 activation revealed the regulation of transcriptional patterns associated with angiogenesis. Finally, the blockade of proangiogenic signalling significantly reduced colorectal tumour growth in mice with constitutive STAT3 activation in COLVI+ fibroblasts. CONCLUSION Altogether our work demonstrates a critical role of STAT3 activation in CAFs in CRC development.
Collapse
Affiliation(s)
- Christina Heichler
- First Department of Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Kristina Scheibe
- First Department of Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Anabel Schmied
- First Department of Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Carol I Geppert
- Department of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Benjamin Schmid
- Optical Imaging Center, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Stefan Wirtz
- First Department of Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Oana-Maria Thoma
- First Department of Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany.,Erlangen Graduate School of Advanced Optical Technologies (SAOT), Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Viktoria Kramer
- First Department of Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Maximilian J Waldner
- First Department of Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Christian Büttner
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Henner F Farin
- German Cancer Consortium (DKTK), Heidelberg, Germany.,Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt am Main, Germany
| | - Marina Pešić
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt am Main, Germany
| | - Ferdinand Knieling
- First Department of Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany.,Department of Pediatrics and Adolescent Medicine, Universitätsklinikum Erlangen Kinder- und Jugendklinik, Erlangen, Germany
| | - Susanne Merkel
- Chirurgische Klinik, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Anika Grüneboom
- Third Department of Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Matthias Gunzer
- Institute of Experimental Immunology and Imaging, University Duisburg-Essen and University Hospital Essen, Essen, Germany
| | - Robert Grützmann
- Chirurgische Klinik, Universitätsklinikum Erlangen, Erlangen, Germany
| | | | - Sergei B Koralov
- Department of Pathology, New York University School of Medicine, New York, New York, USA
| | - George Kollias
- Biomedical Sciences Research Center Alexander Fleming, Vari, Greece
| | - Michael Vieth
- Institute of Pathology, Klinikum Bayreuth, Bayreuth, Germany
| | - Arndt Hartmann
- Department of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Florian R Greten
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt am Main, Germany
| | - Markus F Neurath
- First Department of Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Clemens Neufert
- First Department of Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
20
|
Bürtin F, Mullins CS, Linnebacher M. Mouse models of colorectal cancer: Past, present and future perspectives. World J Gastroenterol 2020; 26:1394-1426. [PMID: 32308343 PMCID: PMC7152519 DOI: 10.3748/wjg.v26.i13.1394] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 03/05/2020] [Accepted: 03/10/2020] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is the third most common diagnosed malignancy among both sexes in the United States as well as in the European Union. While the incidence and mortality rates in western, high developed countries are declining, reflecting the success of screening programs and improved treatment regimen, a rise of the overall global CRC burden can be observed due to lifestyle changes paralleling an increasing human development index. Despite a growing insight into the biology of CRC and many therapeutic improvements in the recent decades, preclinical in vivo models are still indispensable for the development of new treatment approaches. Since the development of carcinogen-induced rodent models for CRC more than 80 years ago, a plethora of animal models has been established to study colon cancer biology. Despite tenuous invasiveness and metastatic behavior, these models are useful for chemoprevention studies and to evaluate colitis-related carcinogenesis. Genetically engineered mouse models (GEMM) mirror the pathogenesis of sporadic as well as inherited CRC depending on the specific molecular pathways activated or inhibited. Although the vast majority of CRC GEMM lack invasiveness, metastasis and tumor heterogeneity, they still have proven useful for examination of the tumor microenvironment as well as systemic immune responses; thus, supporting development of new therapeutic avenues. Induction of metastatic disease by orthotopic injection of CRC cell lines is possible, but the so generated models lack genetic diversity and the number of suited cell lines is very limited. Patient-derived xenografts, in contrast, maintain the pathological and molecular characteristics of the individual patient's CRC after subcutaneous implantation into immunodeficient mice and are therefore most reliable for preclinical drug development - even in comparison to GEMM or cell line-based analyses. However, subcutaneous patient-derived xenograft models are less suitable for studying most aspects of the tumor microenvironment and anti-tumoral immune responses. The authors review the distinct mouse models of CRC with an emphasis on their clinical relevance and shed light on the latest developments in the field of preclinical CRC models.
Collapse
Affiliation(s)
- Florian Bürtin
- Department of General, Visceral, Vascular and Transplantation Surgery, University Medical Center Rostock, University of Rostock, Rostock 18057, Germany
| | - Christina S Mullins
- Department of Thoracic Surgery, University Medical Center Rostock, University of Rostock, Rostock 18057, Germany
| | - Michael Linnebacher
- Molecular Oncology and Immunotherapy, Department of General, Visceral, Vascular and Transplantation Surgery, University Medical Center Rostock, Rostock 18057, Germany
| |
Collapse
|
21
|
Xu Y, Zhang L, Wang Q, Zheng M. Comparison of Different Colorectal Cancer With Liver Metastases Models Using Six Colorectal Cancer Cell Lines. Pathol Oncol Res 2020; 26:2177-2183. [PMID: 32172478 DOI: 10.1007/s12253-020-00805-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Accepted: 03/04/2020] [Indexed: 02/06/2023]
Abstract
At present, modeling methods of colorectal cancer with liver metastases have significant limitations. Here, we established orthotopic and ectopic hepatic metastases models using six colorectal cancer cell lines to choose an ideal animal model for studying colorectal cancer growth and liver metastases. Luciferin-expressing six colorectal cancer cell lines were used to induce animal models of colorectal cancer with liver metastases by intra-splenic injection or implantation of tumor tissue in the caecum. Tumors growth and metastatic events were observed by bioluminescence imaging. In orthotopic transplantation group, six cell lines all had taken rates of 100% for orthotopic tumors but showed variations in rates of growth. HCT-116 cell developed the 50% liver metastases. However, the ectopic transplantation group achieved higher liver metastatic rate, with the highest frequencies for HCT116 cell (90%) and SW620 cell (77.8%). Furthermore, the time to develop liver metastases and survival rates of bearing-tumor mice were shorter than orthotopic transplantation group. Additionally, six colorectal cancer cell lines resulted in more lymph node metastases in orthotopic transplantation group, whereas produced widespread peritoneal seeding in ectopic transplantation group. Bioluminescence imaging and pathological findings confirmed the growth and metastatic characteristics of tumors. Two animal models of colorectal cancer using six cell lines showed highly variations in rates of growth, survival rates of bearing-tumor mice and frequencies of metastases. The study provides useful information for the establishment of clinically relevant colorectal cancer with liver metastases animal models.
Collapse
Affiliation(s)
- Yuting Xu
- Department of Pathology, Xuzhou Medical University, 221004, Xuzhou, Jiangsu, China.
| | - Lin Zhang
- Department of Pathology, Xuzhou Medical University, 221004, Xuzhou, Jiangsu, China
| | - Qingling Wang
- Department of Pathology, Xuzhou Medical University, 221004, Xuzhou, Jiangsu, China
| | - Maojin Zheng
- Department of Pathology, Xuzhou Medical University, 221004, Xuzhou, Jiangsu, China
| |
Collapse
|
22
|
Salmanpour M, Yousefi G, Samani SM, Mohammadi S, Anbardar MH, Tamaddon A. Nanoparticulate delivery of irinotecan active metabolite (SN38) in murine colorectal carcinoma through conjugation to poly (2-ethyl 2-oxazoline)-b-poly (L-glutamic acid) double hydrophilic copolymer. Eur J Pharm Sci 2019; 136:104941. [DOI: 10.1016/j.ejps.2019.05.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 05/22/2019] [Accepted: 05/24/2019] [Indexed: 01/04/2023]
|
23
|
Plummer R, Papageorge M, Ciomek N, Liu T, Yoo J. Myofibroblasts Enhance Tumor Growth in a Novel Mouse Model of Colorectal Cancer. J Surg Res 2019; 244:374-381. [PMID: 31325658 DOI: 10.1016/j.jss.2019.06.051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 05/21/2019] [Accepted: 06/14/2019] [Indexed: 10/26/2022]
Abstract
BACKGROUND Communication between colorectal cancer and stromal cells alters the tumor microenvironment to regulate locoregional disease and cancer progression. However, colon cancer-stromal cell interactions are difficult to study in vivo. Limitations of existing animal models include the use of immunocompromised mice, the inability to genetically modify a cell population in a single organ system, or a lack of anatomic context. Our goal was to develop a novel mouse model of colorectal cancer that is capable of studying tumor-stromal cell interactions in the native colon of immune-competent mice. METHODS Primary mouse myofibroblasts were isolated from the colon of C57BL/6 mice and were grown in cell culture. Genetically defined (ApcΔ/Δ; Kras G12D/+; Trp53Δ/Δ) primary mouse colon cancer cells were suspended in serum-free media (20 μL) at varying concentrations (5 × 103 to 4 × 104 cells) either alone or in combination with syngeneic myofibroblasts (2 × 105 cells). After isoflurane anesthesia, a colonoscopy was performed on immune-competent 8- to 10-week-old C57BL/6 mice with endoscopic microinjection of the cell suspension into the submucosal space of the colon wall utilizing a small animal colonoscope. Surveillance endoscopy was used to assess for tumor growth, along with histologic analysis. Tumor size is presented on a grading system based on tumor diameter relative to colon circumference. RESULTS A total of 33 mice were injected with a survival rate of 88% (29/33). Endoscopic microinjection of colorectal cancer cells resulted in dose-dependent tumor growth in the distal mouse colon that could be assessed endoscopically without animal sacrifice. Growth curves varied depending on the concentration of injected colorectal cancer cells, with no growth at the lowest concentration of injected cells (5 × 103 cells), progressive growth over 4 wk using 1-2 × 104 cells, while the highest colorectal cancer cell concentration (4 × 104 cells) led to larger tumors at week 1 followed by a steady decline in tumor growth over the 4-wk time period. Combined microinjection of 2 × 104 colorectal cancer cells with 2 × 105 myofibroblasts resulted in much larger tumors that persisted over the 4-wk time period and which were composed primarily of colorectal cancer cells. Immunofluorescence microscopy after coinjection of colorectal cancer cells with green fluorescent protein positive myofibroblasts confirmed that the injected myofibroblasts are present and remain viable over the 4-wk time period. CONCLUSIONS Endoscopic submucosal microinjection of primary mouse colorectal cancer cells is feasible and leads to reliable and reproducible short-term growth of colon tumors in immune-competent mice. Coinjection of primary mouse colorectal cancer cells with syngeneic myofibroblasts leads to enhanced tumor growth. Coimplantation of colorectal cancer cells with syngeneic myofibroblasts provides a novel platform to study tumor-stromal interactions in the native colon of immune-competent mice.
Collapse
Affiliation(s)
- Robert Plummer
- Department of Surgery, Tufts University School of Medicine, Tufts Medical Center, Boston, Massachusetts
| | - Marianna Papageorge
- Department of Surgery, Yale University School of Medicine, Yale New Haven Hospital, New Haven, Connecticut
| | - Natalie Ciomek
- Department of Pathology, Tufts University School of Medicine, Tufts Medical Center, Boston, Massachusetts
| | - Tiegang Liu
- Department of Surgery, Tufts University School of Medicine, Tufts Medical Center, Boston, Massachusetts
| | - James Yoo
- Department of Surgery, Tufts University School of Medicine, Tufts Medical Center, Boston, Massachusetts.
| |
Collapse
|
24
|
Size and lipid modification determine liposomal Indocyanine green performance for tumor imaging in a model of rectal cancer. Sci Rep 2019; 9:8566. [PMID: 31189986 PMCID: PMC6561977 DOI: 10.1038/s41598-019-45038-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 05/29/2019] [Indexed: 01/16/2023] Open
Abstract
Localization of rectal tumors is a challenge in minimally invasive surgery due to the lack of tactile sensation. We had developed liposomal indocyanine green (Lip-ICG) for localization of rectal tumor. In this study we evaluated the effects of liposome size and lipid PEGylation on imaging. We used an endoscopically-guided orthotopic experimental rectal cancer model in which tumor fluorescence was determined at different time points after intravenous (i.v.) administration of Lip-ICG and PEGylated liposomes (PEG-Lip-ICG). Signal intensity was measured by tumor-to-background ratio (TBR), or normalized TBR (compared to TBR of free ICG). Fluorescence microscopy of tumor tissue was performed to determine fluorescence localization within the tissue and blood vessels. Liposomes of 60 nm showed an increased TBR compared with free ICG at 12 hours after i.v. injection: normalized TBR (nTBR) = 3.11 vs. 1, respectively (p = 0.006). Larger liposomes (100 nm and 140 nm) had comparable signal to free ICG (nTBR = 0.98 ± 0.02 and 0.78 ± 0.08, respectively), even when additional time points were examined (0.5, 3 and 24 hours). PEG-Lip- ICG were more efficient than Lip-ICG (TBR = 4.2 ± 0.18 vs. 2.5 ± 0.12, p < 0.01) presumably because of reduced uptake by the reticulo-endothelial system. ICG was found outside the capillaries in tumor margins. We conclude that size and lipid modification impact imaging intensity.
Collapse
|
25
|
Paulson B, Lee S, Kim Y, Moon Y, Kim JK. Miniaturized omnidirectional flexible side-view endoscope for rapid monitoring of thin tubular biostructures. BIOMEDICAL OPTICS EXPRESS 2019; 10:2264-2274. [PMID: 31149372 PMCID: PMC6524575 DOI: 10.1364/boe.10.002264] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 03/27/2019] [Accepted: 03/27/2019] [Indexed: 06/09/2023]
Abstract
Endoscopic imaging allows longitudinal observation of epithelial pathologies in tubular organs throughout the body. However, the imaging and optical diagnosis of tubular biostructures such as small animal models and small pediatric organs require appropriately miniaturized devices. A miniaturized catadioptric flexible side-view endoscope is proposed with omnidirectional field of view (FOV) in the transverse direction and sub-mm-scale feature resolution. The FOV in the longitudinal direction is 50°. Images are unwrapped and stitched together to form composite images of the target by two different algorithms, revealing a composite FOV of more than 3.5 cm × 360°. The endoscope is well suited for minimally invasive rapid monitoring of thin tubular organs in pediatric patients, as well as for imaging of small animal disease models at near-cellular resolution.
Collapse
Affiliation(s)
- Bjorn Paulson
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, 88, Olympic-ro 43-gil, Seoul, 05505, South Korea
- Department of Convergence Medicine, University of Ulsan, College of Medicine, 88, Olympic-ro 43-gil, Seoul, 05505, South Korea
| | - SangHwa Lee
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, 88, Olympic-ro 43-gil, Seoul, 05505, South Korea
- Department of Convergence Medicine, University of Ulsan, College of Medicine, 88, Olympic-ro 43-gil, Seoul, 05505, South Korea
| | - Youngkyu Kim
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, 88, Olympic-ro 43-gil, Seoul, 05505, South Korea
- Department of Convergence Medicine, University of Ulsan, College of Medicine, 88, Olympic-ro 43-gil, Seoul, 05505, South Korea
| | - Youngjin Moon
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, 88, Olympic-ro 43-gil, Seoul, 05505, South Korea
- Department of Convergence Medicine, University of Ulsan, College of Medicine, 88, Olympic-ro 43-gil, Seoul, 05505, South Korea
| | - Jun Ki Kim
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, 88, Olympic-ro 43-gil, Seoul, 05505, South Korea
- Department of Convergence Medicine, University of Ulsan, College of Medicine, 88, Olympic-ro 43-gil, Seoul, 05505, South Korea
| |
Collapse
|
26
|
Lannagan TRM, Lee YK, Wang T, Roper J, Bettington ML, Fennell L, Vrbanac L, Jonavicius L, Somashekar R, Gieniec K, Yang M, Ng JQ, Suzuki N, Ichinose M, Wright JA, Kobayashi H, Putoczki TL, Hayakawa Y, Leedham S, Abud HE, Yilmaz ÖH, Marker J, Klebe S, Wirapati P, Mukherjee S, Tejpar S, Leggett BA, Whitehall VLJ, Worthley DL, Woods SL. Genetic editing of colonic organoids provides a molecularly distinct and orthotopic preclinical model of serrated carcinogenesis. Gut 2019; 68:684-692. [PMID: 29666172 PMCID: PMC6192855 DOI: 10.1136/gutjnl-2017-315920] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 03/14/2018] [Accepted: 03/27/2018] [Indexed: 12/25/2022]
Abstract
OBJECTIVE Serrated colorectal cancer (CRC) accounts for approximately 25% of cases and includes tumours that are among the most treatment resistant and with worst outcomes. This CRC subtype is associated with activating mutations in the mitogen-activated kinase pathway gene, BRAF, and epigenetic modifications termed the CpG Island Methylator Phenotype, leading to epigenetic silencing of key tumour suppressor genes. It is still not clear which (epi-)genetic changes are most important in neoplastic progression and we begin to address this knowledge gap herein. DESIGN We use organoid culture combined with CRISPR/Cas9 genome engineering to sequentially introduce genetic alterations associated with serrated CRC and which regulate the stem cell niche, senescence and DNA mismatch repair. RESULTS Targeted biallelic gene alterations were verified by DNA sequencing. Organoid growth in the absence of niche factors was assessed, as well as analysis of downstream molecular pathway activity. Orthotopic engraftment of complex organoid lines, but not BrafV600E alone, quickly generated adenocarcinoma in vivo with serrated features consistent with human disease. Loss of the essential DNA mismatch repair enzyme, Mlh1, led to microsatellite instability. Sphingolipid metabolism genes are differentially regulated in both our mouse models of serrated CRC and human CRC, with key members of this pathway having prognostic significance in the human setting. CONCLUSION We generate rapid, complex models of serrated CRC to determine the contribution of specific genetic alterations to carcinogenesis. Analysis of our models alongside patient data has led to the identification of a potential susceptibility for this tumour type.
Collapse
Affiliation(s)
- Tamsin RM Lannagan
- School of Medicine, University of Adelaide and South Australian Health and Medical Research Institute, Adelaide, SA Australia
| | - Young K Lee
- School of Medicine, University of Adelaide and South Australian Health and Medical Research Institute, Adelaide, SA Australia
| | - Tongtong Wang
- School of Medicine, University of Adelaide and South Australian Health and Medical Research Institute, Adelaide, SA Australia
| | - Jatin Roper
- The David H. Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA
- Division of Gastroenterology, Tufts Medical Center, Boston, MA, United States
| | - Mark L Bettington
- Envoi Specialist Pathologists, Brisbane, QLD Australia
- QIMR Berghofer Medical Research Institute, Brisbane, QLD Australia
| | - Lochlan Fennell
- QIMR Berghofer Medical Research Institute, Brisbane, QLD Australia
| | - Laura Vrbanac
- School of Medicine, University of Adelaide and South Australian Health and Medical Research Institute, Adelaide, SA Australia
| | - Lisa Jonavicius
- Department of Anatomical Pathology, Flinders Medical Centre, Bedford Park, SA Australia
| | - Roshini Somashekar
- School of Medicine, University of Adelaide and South Australian Health and Medical Research Institute, Adelaide, SA Australia
| | - Krystyna Gieniec
- School of Medicine, University of Adelaide and South Australian Health and Medical Research Institute, Adelaide, SA Australia
| | - Miao Yang
- School of Medicine, University of Adelaide and South Australian Health and Medical Research Institute, Adelaide, SA Australia
| | - Jia Q Ng
- School of Medicine, University of Adelaide and South Australian Health and Medical Research Institute, Adelaide, SA Australia
| | - Nobumi Suzuki
- School of Medicine, University of Adelaide and South Australian Health and Medical Research Institute, Adelaide, SA Australia
| | - Mari Ichinose
- School of Medicine, University of Adelaide and South Australian Health and Medical Research Institute, Adelaide, SA Australia
| | - Josephine A Wright
- School of Medicine, University of Adelaide and South Australian Health and Medical Research Institute, Adelaide, SA Australia
| | - Hiroki Kobayashi
- School of Medicine, University of Adelaide and South Australian Health and Medical Research Institute, Adelaide, SA Australia
| | - Tracy L Putoczki
- Department of Medical Biology, University of Melbourne and the Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC Australia
| | - Yoku Hayakawa
- Dept of Gastroenterology, University of Tokyo, Japan
| | - Simon Leedham
- Gastrointestinal Stem Cell Biology Laboratory, Wellcome Trust Centre for Human Genetics University of Oxford, Oxford, & Translational Gastroenterology Unit, Experimental Medicine Division, Nuffield Department of Clinical Medicine, John Radcliffe Hospital, Oxford, Headington, UK
| | - Helen E Abud
- Cancer Program, Monash Biomedicine Discovery Institute and the Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC Australia
| | - Ömer H. Yilmaz
- The David H. Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA
- Department of Pathology, Massachusetts General Hospital, Boston, MA United States
| | | | - Sonja Klebe
- Department of Anatomical Pathology, Flinders Medical Centre, Bedford Park, SA Australia
| | - Pratyaksha Wirapati
- Swiss Institute of Bioinformatics, Bioinformatics Core Facility, Lausanne, Switzerland
| | | | - Sabine Tejpar
- Digestive Oncology Unit, Department of Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Barbara A Leggett
- QIMR Berghofer Medical Research Institute, Brisbane, QLD Australia
- School of Medicine, University of Queensland, QLD Australia
- Royal Brisbane and Womens Hospital, Brisbane, QLD Australia
| | - Vicki LJ Whitehall
- QIMR Berghofer Medical Research Institute, Brisbane, QLD Australia
- School of Medicine, University of Queensland, QLD Australia
- Pathology Queensland, Brisbane, QLD
| | - Daniel L Worthley
- School of Medicine, University of Adelaide and South Australian Health and Medical Research Institute, Adelaide, SA Australia
| | - Susan L Woods
- School of Medicine, University of Adelaide and South Australian Health and Medical Research Institute, Adelaide, SA Australia
| |
Collapse
|
27
|
Lee LYW, Woolley C, Starkey T, Biswas S, Mirshahi T, Bardella C, Segditsas S, Irshad S, Tomlinson I. Serum- and Glucocorticoid-induced Kinase Sgk1 Directly Promotes the Differentiation of Colorectal Cancer Cells and Restrains Metastasis. Clin Cancer Res 2019; 25:629-640. [PMID: 30322876 PMCID: PMC6339518 DOI: 10.1158/1078-0432.ccr-18-1033] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 08/22/2018] [Accepted: 10/10/2018] [Indexed: 12/26/2022]
Abstract
PURPOSE The molecular events that determine intestinal cell differentiation are poorly understood and it is unclear whether it is primarily a passive event or an active process. It is clinically important to gain a greater understanding of the process, because in colorectal cancer, the degree of differentiation of a tumor is associated with patient survival. SGK1 has previously been identified as a gene that is principally expressed in differentiated intestinal cells. In colorectal cancer, there is marked downregulation of SGK1 compared with normal tissue.Experimental Design: An inducible SGK1 viral overexpression system was utilized to induce reexpression of SGK1 in colorectal cancer cell lines. Transcriptomic and phenotypic analyses of these colorectal cancer lines was performed and validation in mouse and human cohorts was performed. RESULTS We demonstrate that SGK1 is upregulated in response to, and an important controller of, intestinal cell differentiation. Reexpression of SGK1 in colorectal cancer cell lines results in features of differentiation, decreased migration rates, and inhibition of metastasis in an orthotopic xenograft model. These effects may be mediated, in part, by SGK1-induced PKP3 expression and increased degradation of MYC. CONCLUSIONS Our results suggest that SGK1 is an important mediator of differentiation of colorectal cells and may inhibit colorectal cancer metastasis.
Collapse
Affiliation(s)
- Lennard Y W Lee
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom.
| | - Connor Woolley
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Thomas Starkey
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Sujata Biswas
- Cancer Cell Biology Group, Oxford Centre for Cancer Gene Research, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Tia Mirshahi
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Chiara Bardella
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Stefania Segditsas
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Shazia Irshad
- Molecular Mechanisms of Colorectal Cancer Group, Nuffield Department of Medicine, Oxford, United Kingdom
| | - Ian Tomlinson
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
28
|
Paulson B, Kim IH, Namgoong JM, Kim YG, Lee S, Moon Y, Shin DM, Choo MS, Kim JK. Longitudinal micro-endoscopic monitoring of high-success intramucosal xenografts for mouse models of colorectal cancer. Int J Med Sci 2019; 16:1453-1460. [PMID: 31673236 PMCID: PMC6818213 DOI: 10.7150/ijms.35666] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 08/02/2019] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most frequently lethal forms of cancer. Intramucosal injection allows development of better mouse models of CRC, as orthotopic xenografts allow development of adenocarcinoma in the submucosa of the mouse colon wall. In this paper, a method of orthotopic injection is monitored longitudinally using cellular-resolution real-time in vivo fluorescence microendoscopy, following the injection of three different cell lines: 3T3-GFP to confirm immunosuppression and HCT116-RFP cells to model CRC. Adenoma formation is first observable after 7 to 10 days, and by use of 33 G needles a tumor induction rate of greater than 85% is documented. An additional experiment on the injection of rapamycin reveals drug efficacy and localization between 24 and 48 hours, and suggests the promise of real-time cellular-resolution fluorescence micro-endoscopy for developing longitudinal therapy regimes in mural models of CRC.
Collapse
Affiliation(s)
- Bjorn Paulson
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, 88, Olympic-ro 43-gil, Songpa-gu, Seoul 05055, Republic of Korea
| | - Ick Hee Kim
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, 391 Technology Way, Winston-Salem, NC 27101, USA
| | - Jung-Man Namgoong
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul 05055, Republic of Korea
| | - Young Gyu Kim
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, 88, Olympic-ro 43-gil, Songpa-gu, Seoul 05055, Republic of Korea
| | - Sanghwa Lee
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, 88, Olympic-ro 43-gil, Songpa-gu, Seoul 05055, Republic of Korea
| | - Youngjin Moon
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, 88, Olympic-ro 43-gil, Songpa-gu, Seoul 05055, Republic of Korea.,Department of Convergence Medicine, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul 05055, Republic of Korea
| | - Dong-Myung Shin
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul 05055, Republic of Korea.,Department of Physiology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul 05055, Republic of Korea
| | - Myung-Soo Choo
- Department of Urology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul 05055, Republic of Korea
| | - Jun Ki Kim
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, 88, Olympic-ro 43-gil, Songpa-gu, Seoul 05055, Republic of Korea.,Department of Convergence Medicine, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul 05055, Republic of Korea
| |
Collapse
|
29
|
Klotho suppresses colorectal cancer through modulation of the unfolded protein response. Oncogene 2018; 38:794-807. [PMID: 30232408 DOI: 10.1038/s41388-018-0489-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 08/07/2018] [Accepted: 08/10/2018] [Indexed: 12/20/2022]
Abstract
Klotho is an anti-aging transmembrane protein, which can be shed and function as a hormone. Accumulating data indicate klotho as a tumor suppressor in a wide array of malignancies and indicate the subdomain KL1 as the active region of the protein. We aimed to study the role of klotho as a tumor suppressor in colorectal cancer. Bioinformatics analyses of TCGA datasets indicated reduced klotho mRNA levels in human colorectal cancer, along with negative regulation of klotho expression by hypermethylation of the promoter and 1st exon, and hypomethylation of an area within the gene. Overexpression or treatment with klotho or KL1 inhibited proliferation of colorectal cancer cells in vitro. The in vivo activity of klotho and KL1 was examined using two models recapitulating development of tumors in the normal colonic environment of immune-competent mice. Treatment with klotho inhibited formation of colon polyps induced by the carcinogen azoxymethane, and KL1 treatment slowed growth of orthotopically-implanted colorectal tumors. Gene expression array revealed that klotho and KL1 expression enhanced the unfolded protein response (UPR) and this was further established by increased levels of spliced XBP1, GRP78 and phosphorylated-eIF2α. Furthermore, attenuation of the UPR partially abrogated klotho tumor suppressor activity. In conclusion, this study indicates klotho as a tumor suppressor in colorectal cancer and identifies, for the first time, the UPR as a pathway mediating klotho activities in cancer. These data suggest that administration of exogenous klotho or KL1 may serve as a novel strategy for prevention and treatment of colorectal cancer.
Collapse
|
30
|
Ou-Yang GQ, Pan GD, Wu YR, Xu HL. Orthotopic mouse models of colorectal cancer liver metastases. Shijie Huaren Xiaohua Zazhi 2018; 26:512-517. [DOI: 10.11569/wcjd.v26.i8.512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is a malignancy with high incidence and mortality, and its high mortality rate is mainly attributed to metastases, with liver metastasis being the main cause of death. Appropriate animal models can provide a basis for studying the metastatic mechanism of colorectal cancer and assessing pre-clinical therapeutic effects. Orthotopic transplantation models that simulate colorectal cancer with liver metastases can better reflect the characteristic of liver metastasis in colorectal cancer. In this article, we review orthotopic transplantation models of liver metastases of colorectal cancer.
Collapse
|
31
|
Caetano-Oliveira R, Gomes MA, Abrantes AM, Tavares-Silva E, Oliveira MC, Laranjo M, Queirós DB, Casalta-Lopes J, Pires S, Carvalho L, Gouveia R, Santos PR, Priolli DG, Tralhão JG, Botelho MF. Revisiting colorectal cancer animal model - An improved metastatic model for distal rectosigmoid colon carcinoma. ACTA ACUST UNITED AC 2018; 25:89-99. [PMID: 29628185 DOI: 10.1016/j.pathophys.2018.02.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 12/26/2017] [Accepted: 02/04/2018] [Indexed: 01/24/2023]
Abstract
Colorectal cancer (CRC) is the second most frequent and fatal cancer in Western countries. Understanding its biology with different incidence along the colon and rectum, genetic profile and how these factors contribute to local/distant progression, has been hampered by the lack of a suitable CRC model. We report a reproducible model, using human CRC cell lines (CL) (WiDr, LS1034, C2BBe1) injected (1 × 107 cells/animal) in RNU rats (n = 55) which underwent cecostomy and descending colostomy with mucosal-cutaneous fistula of the sigmoid colon. CL were characterized by immunohistochemistry: CK20, CDX2, P53, vimentin, Ki67, CD44, CD133, E-cadherin, β-catenin and CEA; cancer stem cells-immune system interaction was studied and tumor progression was assessed with nuclear medicine imaging (99mTc-MIBI). Animals developed locally invasive tumors and with WiDr neural invasion was registered. Cancer stem cells were detected in WiDr (CD44 positive). All the cell lines stimulated the immune system, being WiDr the most aggressive. Imaging studies demonstrated tumor uptake. With this CRC model we can study the microenvironment role and tumor-stroma interactions. All CL developed primary disease, but only the WiDR established neural invasion which may represent a metastatic pathway. This model can help unveiling the underlying metastatic mechanisms, and ultimately test better therapeutic approaches for CRC.
Collapse
Affiliation(s)
- Rui Caetano-Oliveira
- Biophysics Unit, Faculty of Medicine, University of Coimbra, Coimbra, Portugal; Pathology Department, University Hospital (CHUC), Coimbra, Portugal
| | | | - Ana Margarida Abrantes
- Biophysics Unit, Faculty of Medicine, University of Coimbra, Coimbra, Portugal; Centre of Investigation on Environment, Genetics and Oncobiology (CIMAGO), Coimbra, Portugal
| | - Edgar Tavares-Silva
- Surgery A Department, University Hospital (CHUC), Faculty of Medicine, Coimbra, Portugal
| | - Marco Carvalho Oliveira
- Immunology and Oncology Laboratory, Center for Neurosciences and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal; Immunology Institute, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Mafalda Laranjo
- Biophysics Unit, Faculty of Medicine, University of Coimbra, Coimbra, Portugal; Centre of Investigation on Environment, Genetics and Oncobiology (CIMAGO), Coimbra, Portugal
| | - Débora Basílio Queirós
- Immunology and Oncology Laboratory, Center for Neurosciences and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal; Immunology Institute, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - João Casalta-Lopes
- Biophysics Unit, Faculty of Medicine, University of Coimbra, Coimbra, Portugal; Radiotherapy Department, University Hospital (CHUC), Faculty of Medicine, Coimbra, Portugal
| | - Salomé Pires
- Biophysics Unit, Faculty of Medicine, University of Coimbra, Coimbra, Portugal; Centre of Investigation on Environment, Genetics and Oncobiology (CIMAGO), Coimbra, Portugal
| | - Lina Carvalho
- Institute of Anatomic Pathology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Rosa Gouveia
- Thanatology Service of the National Institute of Legal Medicine (Center Delegation), Coimbra, Portugal
| | - Paulo Rodrigues Santos
- Centre of Investigation on Environment, Genetics and Oncobiology (CIMAGO), Coimbra, Portugal; Immunology and Oncology Laboratory, Center for Neurosciences and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal; Immunology Institute, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Denise Gonçalves Priolli
- Postgraduate Program Strictu Senso in Health Science, Sao Francisco University Medical School, Brazil
| | - José Guilherme Tralhão
- Biophysics Unit, Faculty of Medicine, University of Coimbra, Coimbra, Portugal; Centre of Investigation on Environment, Genetics and Oncobiology (CIMAGO), Coimbra, Portugal; Surgery A Department, University Hospital (CHUC), Faculty of Medicine, Coimbra, Portugal
| | - Maria Filomena Botelho
- Biophysics Unit, Faculty of Medicine, University of Coimbra, Coimbra, Portugal; Centre of Investigation on Environment, Genetics and Oncobiology (CIMAGO), Coimbra, Portugal.
| |
Collapse
|
32
|
Roper J, Tammela T, Akkad A, Almeqdadi M, Santos SB, Jacks T, Yilmaz ÖH. Colonoscopy-based colorectal cancer modeling in mice with CRISPR-Cas9 genome editing and organoid transplantation. Nat Protoc 2018; 13:217-234. [PMID: 29300388 PMCID: PMC6145089 DOI: 10.1038/nprot.2017.136] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Most genetically engineered mouse models (GEMMs) of colorectal cancer are limited by tumor formation in the small intestine, a high tumor burden that limits metastasis, and the need to generate and cross mutant mice. Cell line or organoid transplantation models generally produce tumors in ectopic locations-such as the subcutaneous space, kidney capsule, or cecal wall-that do not reflect the native stromal environment of the colon mucosa. Here, we describe detailed protocols to rapidly and efficiently induce site-directed tumors in the distal colon of mice that are based on colonoscopy-guided mucosal injection. These techniques can be adapted to deliver viral vectors carrying Cre recombinase, CRISPR-Cas9 components, CRISPR-engineered mouse tumor organoids, or human cancer organoids to mice to model the adenoma-carcinoma-metastasis sequence of tumor progression. The colonoscopy injection procedure takes ∼15 min, including preparation. In our experience, anyone with reasonable hand-eye coordination can become proficient with mouse colonoscopy and mucosal injection with a few hours of practice. These approaches are ideal for a wide range of applications, including assessment of gene function in tumorigenesis, examination of tumor-stroma interactions, studies of cancer metastasis, and translational research with patient-derived cancers.
Collapse
Affiliation(s)
- Jatin Roper
- The David H. Koch Institute for Integrative Cancer Research at MIT, Cambridge, Massachusetts, USA
- Division of Gastroenterology, Tufts Medical Center, Boston, Massachusetts, USA
- Molecular Oncology Research Institute, Tufts Medical Center, Boston, Massachusetts, USA
| | - Tuomas Tammela
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Adam Akkad
- The David H. Koch Institute for Integrative Cancer Research at MIT, Cambridge, Massachusetts, USA
| | - Mohammad Almeqdadi
- The David H. Koch Institute for Integrative Cancer Research at MIT, Cambridge, Massachusetts, USA
| | - Sebastian B Santos
- The David H. Koch Institute for Integrative Cancer Research at MIT, Cambridge, Massachusetts, USA
| | - Tyler Jacks
- The David H. Koch Institute for Integrative Cancer Research at MIT, Cambridge, Massachusetts, USA
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Ömer H Yilmaz
- The David H. Koch Institute for Integrative Cancer Research at MIT, Cambridge, Massachusetts, USA
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
33
|
Endoscopic non-ablative fractional laser therapy in an orthotopic colon tumour model. Sci Rep 2018; 8:1673. [PMID: 29374265 PMCID: PMC5785993 DOI: 10.1038/s41598-018-19792-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 01/02/2018] [Indexed: 12/28/2022] Open
Abstract
Colorectal cancer is one of the leading causes of cancer-related deaths. Although several therapeutic management strategies are available at the early colon cancer stages, such as endoscopic mucosal or submucosal dissection, associated complications often include bleeding or bowel perforations. As an alternative approach, we investigated endoscopic non-ablative fractional laser (eNAFL) irradiation as a minimally invasive therapeutic modality for the treatment of early-stage colorectal cancer. By implanting SL4-DsRed colon cancer cells into the colons of the C57BL/6 mice, we developed an orthotopic colon tumour mouse model and demonstrated the early-stage tumour growth delay following the eNAFL irradiation. Additionally, we evaluated the temperature changes in the eNAFL-irradiated area using numerical simulations, and induced inflammation using histological analysis. Our results indicate a minimal thermal damage confined to the irradiated spot, sparing the adjacent tissue and alteration in the tumour microenvironment. eNAFL irradiation may be clinically useful as a minimally invasive therapeutic intervention at the early stage of tumourigenesis. In future, an optimal eNAFL therapeutic dose should be determined, in order to increase the efficacy of this approach.
Collapse
|
34
|
Fumagalli A, Suijkerbuijk SJE, Begthel H, Beerling E, Oost KC, Snippert HJ, van Rheenen J, Drost J. A surgical orthotopic organoid transplantation approach in mice to visualize and study colorectal cancer progression. Nat Protoc 2018; 13:235-247. [DOI: 10.1038/nprot.2017.137] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
35
|
Mion F, Vetrano S, Tonon S, Valeri V, Piontini A, Burocchi A, Petti L, Frossi B, Gulino A, Tripodo C, Colombo MP, Pucillo CE. Reciprocal influence of B cells and tumor macro and microenvironments in the ApcMin/+ model of colorectal cancer. Oncoimmunology 2017; 6:e1336593. [PMID: 28919998 DOI: 10.1080/2162402x.2017.1336593] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 05/25/2017] [Accepted: 05/26/2017] [Indexed: 12/22/2022] Open
Abstract
One of the most fascinating aspects of the immune system is its dynamism, meant as the ability to change and readapt according to the organism needs. Following an insult, we assist to the spontaneous organization of different immune cells which cooperate, locally and at distance, to build up an appropriate response. Throughout tumor progression, adaptations within the systemic tumor environment, or macroenvironment, result in the promotion of tumor growth, tumor invasion and metastasis to distal organs, but also to dramatic changes in the activity and composition of the immune system. In this work, we show the changes of the B-cell arm of the immune system following tumor progression in the ApcMin/+ model of colorectal cancer. Tumor macroenvironment leads to an increased proportion of total and IL-10-competent B cells in draining LNs while activates a differentiation route that leads to the expansion of IgA+ lymphocytes in the spleen and peritoneum. Importantly, serum IgA levels were significantly higher in ApcMin/+ than Wt mice. The peculiar involvement of IgA response in the adenomatous transformation had correlates in the gut-mucosal compartment where IgA-positive elements increased from normal mucosa to areas of low grade dysplasia while decreasing upon overt carcinomatous transformation. Altogether, our findings provide a snapshot of the tumor education of B lymphocytes in the ApcMin/+ model of colorectal cancer. Understanding how tumor macroenvironment affects the differentiation, function and distribution of B lymphocytes is pivotal to the generation of specific therapies, targeted to switching B cells to an anti-, rather than pro-, tumoral phenotype.
Collapse
Affiliation(s)
- Francesca Mion
- Department of Medicine, University of Udine, Udine, Italy
| | - Stefania Vetrano
- Inflammatory Bowel Disease Center, Humanitas Research Hospital, Rozzano, Italy.,Department of Biomedical Sciences, Humanitas University, Milano, Italy
| | - Silvia Tonon
- Department of Medicine, University of Udine, Udine, Italy
| | - Viviana Valeri
- Department of Medicine, University of Udine, Udine, Italy
| | - Andrea Piontini
- Department of Biomedical Sciences, Humanitas University, Milano, Italy
| | - Alessia Burocchi
- Department of Experimental Oncology and Molecular Medicine, Molecular Immunology Unit, Fondazione IRCCS Istituto Nazionale Tumori, Milano, Italy
| | - Luciana Petti
- Department of Biotechnology and Translational Medicine, University of Milan, Milano, Italy
| | - Barbara Frossi
- Department of Medicine, University of Udine, Udine, Italy
| | - Alessandro Gulino
- Department of Health Science, Tumor Immunology Unit, Human Pathology Section, Palermo University School of Medicine, Palermo, Italy
| | - Claudio Tripodo
- Department of Health Science, Tumor Immunology Unit, Human Pathology Section, Palermo University School of Medicine, Palermo, Italy
| | - Mario P Colombo
- Department of Experimental Oncology and Molecular Medicine, Molecular Immunology Unit, Fondazione IRCCS Istituto Nazionale Tumori, Milano, Italy
| | | |
Collapse
|
36
|
Roper J, Tammela T, Cetinbas NM, Akkad A, Roghanian A, Rickelt S, Almeqdadi M, Wu K, Oberli MA, Sánchez-Rivera FJ, Park YK, Liang X, Eng G, Taylor MS, Azimi R, Kedrin D, Neupane R, Beyaz S, Sicinska ET, Suarez Y, Yoo J, Chen L, Zukerberg L, Katajisto P, Deshpande V, Bass AJ, Tsichlis PN, Lees J, Langer R, Hynes RO, Chen J, Bhutkar A, Jacks T, Yilmaz ÖH. In vivo genome editing and organoid transplantation models of colorectal cancer and metastasis. Nat Biotechnol 2017; 35:569-576. [PMID: 28459449 PMCID: PMC5462879 DOI: 10.1038/nbt.3836] [Citation(s) in RCA: 250] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 03/01/2017] [Indexed: 02/07/2023]
Abstract
In vivo interrogation of the function of genes implicated in tumorigenesis is limited by the need to generate and cross germline mutant mice. Here we describe approaches to model colorectal cancer (CRC) and metastasis, which rely on in situ gene editing and orthotopic organoid transplantation in mice without cancer-predisposing mutations. Autochthonous tumor formation is induced by CRISPR-Cas9-based editing of the Apc and Trp53 tumor suppressor genes in colon epithelial cells and by orthotopic transplantation of Apc-edited colon organoids. ApcΔ/Δ;KrasG12D/+;Trp53Δ/Δ (AKP) mouse colon organoids and human CRC organoids engraft in the distal colon and metastasize to the liver. Finally, we apply the orthotopic transplantation model to characterize the clonal dynamics of Lgr5+ stem cells and demonstrate sequential activation of an oncogene in established colon adenomas. These experimental systems enable rapid in vivo characterization of cancer-associated genes and reproduce the entire spectrum of tumor progression and metastasis.
Collapse
Affiliation(s)
- Jatin Roper
- The David H. Koch Institute for Integrative Cancer Research at MIT, Cambridge, Massachusetts, USA
- Division of Gastroenterology, Tufts Medical Center, Boston, Massachusetts, USA
- Molecular Oncology Research Institute, Tufts Medical Center, Boston, Massachusetts, USA
| | - Tuomas Tammela
- The David H. Koch Institute for Integrative Cancer Research at MIT, Cambridge, Massachusetts, USA
| | - Naniye Malli Cetinbas
- The David H. Koch Institute for Integrative Cancer Research at MIT, Cambridge, Massachusetts, USA
| | - Adam Akkad
- The David H. Koch Institute for Integrative Cancer Research at MIT, Cambridge, Massachusetts, USA
| | - Ali Roghanian
- The David H. Koch Institute for Integrative Cancer Research at MIT, Cambridge, Massachusetts, USA
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, United Kingdom
| | - Steffen Rickelt
- The David H. Koch Institute for Integrative Cancer Research at MIT, Cambridge, Massachusetts, USA
| | - Mohammad Almeqdadi
- The David H. Koch Institute for Integrative Cancer Research at MIT, Cambridge, Massachusetts, USA
| | - Katherine Wu
- The David H. Koch Institute for Integrative Cancer Research at MIT, Cambridge, Massachusetts, USA
| | - Matthias A Oberli
- The David H. Koch Institute for Integrative Cancer Research at MIT, Cambridge, Massachusetts, USA
| | | | - Yoona K Park
- The David H. Koch Institute for Integrative Cancer Research at MIT, Cambridge, Massachusetts, USA
| | - Xu Liang
- The David H. Koch Institute for Integrative Cancer Research at MIT, Cambridge, Massachusetts, USA
| | - George Eng
- The David H. Koch Institute for Integrative Cancer Research at MIT, Cambridge, Massachusetts, USA
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Martin S Taylor
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Roxana Azimi
- The David H. Koch Institute for Integrative Cancer Research at MIT, Cambridge, Massachusetts, USA
| | - Dmitriy Kedrin
- The David H. Koch Institute for Integrative Cancer Research at MIT, Cambridge, Massachusetts, USA
| | - Rachit Neupane
- The David H. Koch Institute for Integrative Cancer Research at MIT, Cambridge, Massachusetts, USA
| | - Semir Beyaz
- The David H. Koch Institute for Integrative Cancer Research at MIT, Cambridge, Massachusetts, USA
| | - Ewa T Sicinska
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Yvelisse Suarez
- Department of Pathology, Tufts Medical Center, Boston, Massachusetts, USA
| | - James Yoo
- Molecular Oncology Research Institute, Tufts Medical Center, Boston, Massachusetts, USA
- Department of Surgery, Tufts Medical Center, Boston, Massachusetts, USA
| | - Lillian Chen
- Department of Surgery, Tufts Medical Center, Boston, Massachusetts, USA
| | - Lawrence Zukerberg
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Pekka Katajisto
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - Vikram Deshpande
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Adam J Bass
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Philip N Tsichlis
- Molecular Oncology Research Institute, Tufts Medical Center, Boston, Massachusetts, USA
| | - Jacqueline Lees
- The David H. Koch Institute for Integrative Cancer Research at MIT, Cambridge, Massachusetts, USA
| | - Robert Langer
- The David H. Koch Institute for Integrative Cancer Research at MIT, Cambridge, Massachusetts, USA
| | - Richard O Hynes
- The David H. Koch Institute for Integrative Cancer Research at MIT, Cambridge, Massachusetts, USA
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Jianzhu Chen
- The David H. Koch Institute for Integrative Cancer Research at MIT, Cambridge, Massachusetts, USA
| | - Arjun Bhutkar
- The David H. Koch Institute for Integrative Cancer Research at MIT, Cambridge, Massachusetts, USA
| | - Tyler Jacks
- The David H. Koch Institute for Integrative Cancer Research at MIT, Cambridge, Massachusetts, USA
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Ömer H Yilmaz
- The David H. Koch Institute for Integrative Cancer Research at MIT, Cambridge, Massachusetts, USA
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
37
|
Magdassi S, Bar-David S, Friedman-Levi Y, Zigmond E, Varol C, Lahat G, Klausner J, Eyal S, Nizri E. Intraoperative Localization of Rectal Tumors Using Liposomal Indocyanine Green. Surg Innov 2017; 24:139-144. [PMID: 28152672 DOI: 10.1177/1553350617690310] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Tumor localization may pose a significant challenge during minimally invasive rectal resection. Near-infrared (NIR) imaging can penetrate biological tissue and afford tumor localization from the external surface of the rectum. Our aim was to develop an NIR-based tool for rectal tumor imaging that can be administered intravenously. METHODS We prepared indocyanine-green (ICG)-loaded liposomes by sonication. Liposomes were evaluated for their size and morphology. We then used an endoscopically induced rectal cancer in mice as a model for rectal cancer. After intravenous administration, tumors were evaluated for their fluorescence intensity. Tumor intensity was expressed in relation to the background signal, that is, tumor to background ratio (TBR). RESULTS Liposomes in various sizes could be prepared by adjusting sonication time. We selected 100-nm-sized liposomes for further experiments. Transmission electron microscopy showed spherical particles and confirmed the size measurements. The liposomes could be lyophilized and then rehydrated again before use without compromising their structure or signal. Fluorescence intensity was kept for 24 hours after solubilization. Testing the optimal time course for rectal tumor imaging revealed that early time course (up to 3 hours) yielded nonspecific imaging, whereas after long time course (24 hours), a very weak signal remained in the tissue. The optimal time window for imaging was after 12 hours from injection, with TBR = 8.1 ± 3.6 ( P = .002). Free ICG could not achieve similar results. CONCLUSIONS The liposomal ICG can be reproducibly prepared and kept in lyophilized form. Liposomal ICG could serve as a tool for intraoperative tumor localization.
Collapse
Affiliation(s)
- Shlomo Magdassi
- 1 Casali Center for Applied Chemistry, Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Shoshi Bar-David
- 2 Laboratory of Surgical Oncology, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Yael Friedman-Levi
- 3 Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ehud Zigmond
- 4 Research Center for Digestive Tract and Liver Diseases, Tel-Aviv Sourasky Medical Center and Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Chen Varol
- 4 Research Center for Digestive Tract and Liver Diseases, Tel-Aviv Sourasky Medical Center and Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Guy Lahat
- 2 Laboratory of Surgical Oncology, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel.,5 Department of Surgery A, Division of surgery, Tel-Aviv Sourasky Medical Center and Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Joseph Klausner
- 2 Laboratory of Surgical Oncology, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel.,5 Department of Surgery A, Division of surgery, Tel-Aviv Sourasky Medical Center and Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Sara Eyal
- 3 Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Eran Nizri
- 2 Laboratory of Surgical Oncology, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel.,5 Department of Surgery A, Division of surgery, Tel-Aviv Sourasky Medical Center and Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| |
Collapse
|
38
|
Highton AJ, Girardin A, Bell GM, Hook SM, Kemp RA. Chitosan gel vaccine protects against tumour growth in an intracaecal mouse model of cancer by modulating systemic immune responses. BMC Immunol 2016; 17:39. [PMID: 27756214 PMCID: PMC5069793 DOI: 10.1186/s12865-016-0178-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Accepted: 10/13/2016] [Indexed: 02/08/2023] Open
Abstract
Background Vaccination generating a robust memory population of CD8+ T cells may provide protection against cancer. However, immune therapies for cancer are influenced by the local tumour immune microenvironment. An infiltrate of T cells into tumours of people with colorectal cancer has proven to be a significant indicator of good prognosis. Methods We used an intracaecal mouse model of cancer to determine whether a protective immune response against a mucosal gut tumour could be generated using a systemic intervention. We investigated the generation of murine memory CD8+ T cells using a sustained antigen release vaccine vehicle (chitosan gel; Gel + OVA) containing the model antigen ovalbumin, chitosan gel alone (Gel) or conventional dendritic cell vaccination (DC + OVA) using the same protein antigen. Results Following vaccination with Gel + OVA, CD8+ T cell memory populations specific for ovalbumin protein were detected. Only vaccination with Gel + OVA gave decreased tumour burden compared to unvaccinated or DC + OVA-vaccinated mice in the intracaecal cancer challenge model. Conclusion These results indicate that subcutaneous vaccination with Gel + OVA generates a population of functional CD8+ memory T cells in lymphoid tissue able to protect against intracaecal tumour challenge. Vaccination with chitosan gel may be valuable in anti-cancer treatment at both peripheral and mucosal sites. Electronic supplementary material The online version of this article (doi:10.1186/s12865-016-0178-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Andrew J Highton
- Department of Microbiology and Immunology, Otago School of Medical Sciences, University of Otago, PO Box 56, Dunedin, 9054, New Zealand
| | - Adam Girardin
- Department of Microbiology and Immunology, Otago School of Medical Sciences, University of Otago, PO Box 56, Dunedin, 9054, New Zealand
| | - Georgia M Bell
- Department of Microbiology and Immunology, Otago School of Medical Sciences, University of Otago, PO Box 56, Dunedin, 9054, New Zealand
| | - Sarah M Hook
- School of Pharmacy, University of Otago, Dunedin, New Zealand
| | - Roslyn A Kemp
- Department of Microbiology and Immunology, Otago School of Medical Sciences, University of Otago, PO Box 56, Dunedin, 9054, New Zealand.
| |
Collapse
|
39
|
Afik R, Zigmond E, Vugman M, Klepfish M, Shimshoni E, Pasmanik-Chor M, Shenoy A, Bassat E, Halpern Z, Geiger T, Sagi I, Varol C. Tumor macrophages are pivotal constructors of tumor collagenous matrix. J Exp Med 2016; 213:2315-2331. [PMID: 27697834 PMCID: PMC5068227 DOI: 10.1084/jem.20151193] [Citation(s) in RCA: 259] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 08/29/2016] [Indexed: 12/21/2022] Open
Abstract
Tumor-associated macrophages are pivotal constructors of the tumoral ECM structure and molecular composition. In particular, they orchestrate the buildup of the tumorigenic collagenous ECM niche. Tumor-associated macrophages (TAMs) promote tumor development, invasion, and dissemination by various mechanisms. In this study, using an orthotopic colorectal cancer (CRC) model, we found that monocyte-derived TAMs advance tumor development by the remodeling of its extracellular matrix (ECM) composition and structure. Unbiased transcriptomic and proteomic analyses of (a) TAM-abundant and -deficient tumor tissues and (b) sorted tumor-associated and -resident colonic macrophage subpopulations defined a distinct TAM-induced ECM molecular signature composed of an ensemble of matricellular proteins and remodeling enzymes they provide to the tumor microenvironment. Remarkably, many of these ECM proteins are specifically increased in human CRC versus healthy colon. Specifically, we demonstrate that although differentiating into TAMs, monocytes up-regulate matrix-remodeling programs associated with the synthesis and assembly of collagenous ECM, specifically collagen types I, VI, and XIV. This finding was further established by advanced imaging showing that TAMs instruct the deposition, cross-linking, and linearization of collagen fibers during tumor development, especially at areas of tumor invasiveness. Finally, we show that cancer-associated fibroblasts are significantly outnumbered by TAMs in this model and that their expression of collagen XIV and I is reduced by TAM deficiency. Here, we outline a novel TAM protumoral function associated with building of the collagenous ECM niche.
Collapse
Affiliation(s)
- Ran Afik
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Ehud Zigmond
- Research Center for Digestive Tract and Liver Diseases, Tel Aviv Sourasky Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Milena Vugman
- Research Center for Digestive Tract and Liver Diseases, Tel Aviv Sourasky Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Mordehay Klepfish
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Elee Shimshoni
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Metsada Pasmanik-Chor
- Bioinformatics Unit, George S. Wise Faculty of Life Science, Tel Aviv University, Tel Aviv 69978, Israel
| | - Anjana Shenoy
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Elad Bassat
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Zamir Halpern
- Research Center for Digestive Tract and Liver Diseases, Tel Aviv Sourasky Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Tamar Geiger
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Irit Sagi
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Chen Varol
- Research Center for Digestive Tract and Liver Diseases, Tel Aviv Sourasky Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
40
|
Endoscopy-guided orthotopic implantation of colorectal cancer cells results in metastatic colorectal cancer in mice. Clin Exp Metastasis 2016; 33:551-62. [PMID: 27146063 DOI: 10.1007/s10585-016-9797-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 04/26/2016] [Indexed: 01/07/2023]
Abstract
Advanced stage colorectal cancer (CRC) is still associated with limited prognosis. For preclinical evaluation of novel therapeutic approaches, murine models with orthotopic tumor growth and distant metastases are required. However, these models usually require surgical procedures possibly influencing tumor immunogenicity and development. The aim of this study was to establish a minimal-invasive endoscopy-based murine orthotopic model of metastatic CRC. During colonoscopy of CD-1 nude and non-obese diabetic/severe combined immunodeficiency (NOD/SCID) mice, implantation of Caco-2 and HT-29 CRC cells was performed subcutaneously (s.c.) or orthotopic into the colonic submucosa. White light endoscopy (WLE) and fluorescence endoscopy (FE) were applied for tumor detection in vivo. Ex vivo, resected tumors were examined by fluorescence reflectance imaging (FRI), histology, gelatin zymography and immunohistochemistry. In CD-1 nude mice, marked tumor growth was observed within 14 days after subcutaneous implantation while submucosal implantation failed to induce CRC after 17 weeks. In contrast, in NOD/SCID mice submucosal injection of HT-29 cells resulted in pronounced tumor growth 12 days post injectionem. Subsequently, rapid tumor expansion occurred, occupying the entire colonic circumference. Importantly, post mortem histological analyses confirmed liver metastases in 28.6 % and peritoneal metastases in 14.3 % of all mice. FRI and gelatin zymography did not detect a significantly increased matrix metalloproteinases (MMPs) expression in s.c. implanted tumors while MMP-tracer uptake was significantly enhanced in orthotopic implanted tumors. Neither s.c. nor orthotopic Caco-2 cell implantation resulted in tumor development. We successfully established an endoscopy-based model of metastatic CRC in immunodeficient mice.
Collapse
|
41
|
Mittal VK, Bhullar JS, Jayant K. Animal models of human colorectal cancer: Current status, uses and limitations. World J Gastroenterol 2015; 21:11854-11861. [PMID: 26557009 PMCID: PMC4631983 DOI: 10.3748/wjg.v21.i41.11854] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 06/09/2015] [Accepted: 08/31/2015] [Indexed: 02/06/2023] Open
Abstract
AIM To make orthotopic colon cancer murine models a more clearly understood subject. The orthotopic tumor models have been found to be more relevant in replicating the human disease process as compared to heterotopic models, many techniques for making orthotopic colorectal murine models have been reported. METHODS We evaluated the current literature for various reported orthotopic colon cancer models to understand their techniques, advantages and limitations. An extensive literature review was performed by searching the National Library of Medicine Database (PubMed) using MeSH terms animal model; colon cancer; orthotopic model; murine model. Twenty studies related to colon cancer orthotopic xenograft model were evaluated in detail and discussed here. RESULTS The detailed analysis of all relevant reports on orthotopic model showed tumor take rate between 42%-100%. While models using the enema technique and minimally invasive technique have reported development of tumor from mucosa with tumor take rate between 87%-100% with metastasis in 76%-90%. CONCLUSION Over the years, the increased understanding of the murine models of human colon cancer has resulted in the development of various models. Each reported model has some limitations. These latest models have opened up new doors for continuing cancer research for not only understanding the colon cancer pathogenesis but also aid in the development of newer chemotherapeutic drugs as they mimic the human disease closely.
Collapse
|
42
|
Cheng LS, Hotta R, Graham HK, Nagy N, Goldstein AM, Belkind-Gerson J. Endoscopic delivery of enteric neural stem cells to treat Hirschsprung disease. Neurogastroenterol Motil 2015; 27:1509-14. [PMID: 26190543 PMCID: PMC4600089 DOI: 10.1111/nmo.12635] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 06/18/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND Transplantation of enteric neural stem cells (ENSC) holds promise as a potential therapy for enteric neuropathies, including Hirschsprung disease. Delivery of transplantable cells via laparotomy has been described, but we propose a novel, minimally invasive endoscopic method of cell delivery. METHODS Enteric neural stem cells for transplantation were cultured from dissociated gut of postnatal donor mice. Twelve recipient mice, including Ednrb(-/-) mice with distal colonic aganglionosis, underwent colonoscopic injection of ENSC under direct vision using a 30-gauge Hamilton needle passed through a rigid cystoureteroscope. Cell engraftment, survival, and neuroglial differentiation were studied 1-4 weeks after the procedure. KEY RESULTS All recipient mice tolerated the procedure without complications and survived to sacrifice. Transplanted cells were found within the colonic wall in 9 of 12 recipient mice with differentiation into enteric neurons and glia. CONCLUSIONS & INFERENCES Endoscopic injection of ENSC is a safe and reliable method for cell delivery, and can be used to deliver a large number of cells to a specific area of disease. This minimally invasive endoscopic approach may prove beneficial to future human applications of cell therapy for neurointestinal disease.
Collapse
Affiliation(s)
- Lily S. Cheng
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA,Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Ryo Hotta
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Hannah K. Graham
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Nandor Nagy
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA,Semmelweis University, Budapest, Hungary
| | - Allan M. Goldstein
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA,Pediatric Neurogastroenterology Program, MassGeneral Hospital for Children, Boston, MA, USA
| | - Jaime Belkind-Gerson
- Pediatric Neurogastroenterology Program, MassGeneral Hospital for Children, Boston, MA, USA,Department of Pediatric Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
43
|
Golovko D, Kedrin D, Yilmaz ÖH, Roper J. Colorectal cancer models for novel drug discovery. Expert Opin Drug Discov 2015; 10:1217-29. [PMID: 26295972 DOI: 10.1517/17460441.2015.1079618] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Despite increased screening rates and advances in targeted therapy, colorectal cancer (CRC) remains the third leading cause of cancer-related mortality. CRC models that recapitulate key features of human disease are essential to the development of novel and effective therapeutics. Classic methods of modeling CRC such as human cell lines and xenograft mice, while useful for many applications, carry significant limitations. Recently developed in vitro and in vivo models overcome some of these deficiencies and thus can be utilized to better model CRC for mechanistic and translational research. AREAS COVERED The authors review established models of in vitro cell culture and describe advances in organoid culture for studying normal and malignant intestine. They also discuss key features of classic xenograft models and describe other approaches for in vivo CRC research, including patient-derived xenograft, carcinogen-induced, orthotopic transplantation and transgenic mouse models. We also describe mouse models of metastatic CRC. EXPERT OPINION No single model is optimal for drug discovery in CRC. Genetically engineered models overcome many limitations of xenograft models. Three-dimensional organoids can be efficiently derived from both normal and malignant tissue for large-scale in vitro and in vivo (transplantation) studies and are thus a significant advance in CRC drug discovery.
Collapse
Affiliation(s)
- Daniel Golovko
- a 1 Tufts Medical Center, Division of Gastroenterology and Molecular Oncology Research Institute , Boston, MA 02111, USA
| | - Dmitriy Kedrin
- b 2 MIT, The David H. Koch Institute for Integrative Cancer Research at MIT, Department of Biology , Cambridge, MA 02139, USA.,c 3 Massachusetts General Hospital and Harvard Medical School, Division of Gastroenterology , Boston, MA 02114, USA
| | - Ömer H Yilmaz
- b 2 MIT, The David H. Koch Institute for Integrative Cancer Research at MIT, Department of Biology , Cambridge, MA 02139, USA.,d 4 Massachusetts General Hospital and Harvard Medical School, Department of Pathology , Boston, MA 02114, USA
| | - Jatin Roper
- a 1 Tufts Medical Center, Division of Gastroenterology and Molecular Oncology Research Institute , Boston, MA 02111, USA .,b 2 MIT, The David H. Koch Institute for Integrative Cancer Research at MIT, Department of Biology , Cambridge, MA 02139, USA
| |
Collapse
|
44
|
Primary Myofibroblasts Maintain Short-Term Viability following Submucosal Injection in Syngeneic, Immune-Competent Mice Utilizing Murine Colonoscopy. PLoS One 2015; 10:e0127258. [PMID: 26016485 PMCID: PMC4445916 DOI: 10.1371/journal.pone.0127258] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 04/13/2015] [Indexed: 01/24/2023] Open
Abstract
The myofibroblast is an important stromal cell of the gastrointestinal tract. Current in vitro and in vivo models either do not accurately recreate stromal-epithelial interactions or are not specific to myofibroblasts. We sought to create an animal model that would allow the study of myofibroblast-epithelial interactions. We isolated and cultured colonic myofibroblasts from FVB mice. Cells were α-SMA and vimentin positive but desmin negative on immunoblot analysis. We injected the myofibroblasts into the colonic submucosa of syngeneic adult mice (n = 8) via a miniendoscopic system. We then isolated green fluorescent protein (GFP) positive colonic myofibroblasts from C57BL/6-Tg(CAG-EGFP)1Osb/J mice and injected them into the colonic lamina propria of C57BL/6J mice at 1x105 (n = 14), 1x106 (n = 9), or 5x106 cells/mL (n = 4). A subset of mice were injected with serum-free media and ink without cells (n = 3). Mice underwent repeat endoscopy and euthanasia one or 7 days after injection. Colons were isolated and either fixed in 10% formalin or the inked sites were individually excised and lysed for DNA. We assessed the injection sites via histology and immunohistochemical stains for α-SMA and GFP. We used qPCR to quantify GFP DNA transcripts at the lamina propria injection sites. Submucosal injection of myofibroblasts resulted in the formation of a subepithelial wheal on endoscopy, which persisted to day 7. Myofibroblasts injected either into the submucosa or lamina propria maintained viability on post-injection day 7 as evidenced by positive α-SMA staining. qPCR of lamina propria injections showed a dose-dependent increase in GFP DNA transcripts on post-injection day 1, whereas the number of transcripts on day 7 was equivalent for the concentrations injected. We demonstrate short-term survival of primary cultured colonic myofibroblasts in syngeneic mice. This may prove to be a valuable model for studying the role of myofibroblasts in states of health and disease.
Collapse
|
45
|
Near-Infrared Confocal Laser Endomicroscopy Detects Colorectal Cancer via an Integrin αvβ3 Optical Probe. Mol Imaging Biol 2015; 17:450-60. [DOI: 10.1007/s11307-015-0825-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
46
|
Brückner M, Lenz P, Nowacki TM, Pott F, Foell D, Bettenworth D. Murine endoscopy for in vivo multimodal imaging of carcinogenesis and assessment of intestinal wound healing and inflammation. J Vis Exp 2014:51875. [PMID: 25226434 PMCID: PMC4828016 DOI: 10.3791/51875] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Mouse models are widely used to study pathogenesis of human diseases and to evaluate diagnostic procedures as well as therapeutic interventions preclinically. However, valid assessment of pathological alterations often requires histological analysis, and when performed ex vivo, necessitates death of the animal. Therefore in conventional experimental settings, intra-individual follow-up examinations are rarely possible. Thus, development of murine endoscopy in live mice enables investigators for the first time to both directly visualize the gastrointestinal mucosa and also repeat the procedure to monitor for alterations. Numerous applications for in vivo murine endoscopy exist, including studying intestinal inflammation or wound healing, obtaining mucosal biopsies repeatedly, and to locally administer diagnostic or therapeutic agents using miniature injection catheters. Most recently, molecular imaging has extended diagnostic imaging modalities allowing specific detection of distinct target molecules using specific photoprobes. In conclusion, murine endoscopy has emerged as a novel cutting-edge technology for diagnostic experimental in vivo imaging and may significantly impact on preclinical research in various fields.
Collapse
Affiliation(s)
| | - Philipp Lenz
- Department of Medicine B, University Hospital Münster
| | | | | | - Dirk Foell
- Department of Pediatric Rheumatology and Immunology, University Children's Hospital Münster
| | | |
Collapse
|
47
|
Roper J, Martin ES, Hung KE. Overview of genetically engineered mouse models of colorectal carcinoma to enable translational biology and drug development. ACTA ACUST UNITED AC 2014; 65:14.29.1-10. [PMID: 24934606 DOI: 10.1002/0471141755.ph1429s65] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Preclinical models for colorectal cancer (CRC) are critical for translational biology and drug development studies to characterize and treat this condition. Mouse models of human cancer are particularly popular because of their relatively low cost, short life span, and ease of use. Genetically engineered mouse models (GEMMs) of CRC are engineered from germline or somatic modification of critical tumor suppressor genes and/or oncogenes that drive mutations in human disease. Detailed in this overview are the salient features of several useful colorectal cancer GEMMs and their value as tools for translational biology and preclinical drug development.
Collapse
Affiliation(s)
- Jatin Roper
- Division of Gastroenterology and Molecular Oncology Research Institute, Tufts Medical Center, Boston, Massachusetts
| | | | | |
Collapse
|
48
|
Zaytseva YY, Elliott VA, Rychahou P, Mustain WC, Kim JT, Valentino J, Gao T, O'Connor KL, Neltner JM, Lee EY, Weiss HL, Evers BM. Cancer cell-associated fatty acid synthase activates endothelial cells and promotes angiogenesis in colorectal cancer. Carcinogenesis 2014; 35:1341-51. [PMID: 24510238 DOI: 10.1093/carcin/bgu042] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Upregulation of fatty acid synthase (FASN), a key enzyme of de novo lipogenesis, is associated with metastasis in colorectal cancer (CRC). However, the mechanisms of regulation are unknown. Since angiogenesis is crucial for metastasis, we investigated the role of FASN in the neovascularization of CRC. The effect of FASN on tumor vasculature was studied in orthotopic CRCs, the chick embryo chorioallantoic membrane (CAM) and Matrigel plug models using immunohistochemistry, immunofluorescent staining and confocal microscopy. Cell secretion was evaluated by ELISA and antibody arrays. Proliferation, migration and tubulogenesis of endothelial cells (ECs) were assessed in CRC-EC coculture models. In this study, we found that stable knockdown of FASN decreased microvessel density in HT29 and HCT116 orthotopic CRCs and resulted in 'normalization' of tumor vasculature in both orthotopic and CAM models. Furthermore, FASN regulated secretion of pro- and antiangiogenic factors, including vascular endothelial growth factor-A (VEGF-A). Mechanisms associated with the antiangiogenic activity noted with knockdown of FASN included: downregulation of VEGF(189), upregulation of antiangiogenic isoform VEGF(165b) and a decrease in expression and activity of matrix metalloproteinase-9. Furthermore, conditioned medium from FASN knockdown CRC cells inhibited activation of vascular endothelial growth factor receptor-2 and its downstream signaling and decreased proliferation, migration and tubulogenesis of ECs as compared with control medium. Together, these results suggest that cancer cell-associated FASN regulates tumor vasculature through alteration of the profile of secreted angiogenic factors and regulation of their bioavailability. Inhibition of FASN upstream of VEGF-A and other angiogenic pathways can be a novel therapeutic strategy to prevent or inhibit metastasis in CRC.
Collapse
Affiliation(s)
| | | | | | | | - Ji Tae Kim
- Markey Cancer Center, Department of Surgery
| | | | - Tianyan Gao
- Department of Molecular and Cellular Biochemistry
| | | | | | - Eun Y Lee
- Markey Cancer Center, Department of Surgery, Department of Pathology and Laboratory Medicine and
| | - Heidi L Weiss
- Markey Cancer Center, Department of Biostatistics, University of Kentucky, Lexington, KY 40536, USA
| | | |
Collapse
|
49
|
Ward JM, Treuting PM. Rodent intestinal epithelial carcinogenesis: pathology and preclinical models. Toxicol Pathol 2013; 42:148-61. [PMID: 24178574 DOI: 10.1177/0192623313505156] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Colon cancer is a major human malignancy that afflicts millions of people throughout the world each year. Genetics and diet play large roles in colon carcinogenesis although chemicals may also contribute. For the past 40 years, scientists have studied experimentally induced intestinal carcinogenesis in rodents in order to elucidate the etiology and mechanisms involved. Comparative histopathology has revealed many similarities of rodent and human intestinal cancers. Comparative molecular pathology has also shown genetic similarities. More recently, genetically engineered mice and inflammatory colon cancer models have been used for investigating mechanisms and potential chemopreventive and treatment modalities. This review will focus on comparative histopathology and nonclinical models.
Collapse
|
50
|
Karim BO, Huso DL. Mouse models for colorectal cancer. Am J Cancer Res 2013; 3:240-50. [PMID: 23841024 PMCID: PMC3696531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Accepted: 05/23/2013] [Indexed: 06/02/2023] Open
Abstract
Colorectal cancer (CRC) is the third leading cause of cancer-related death in the United States, with the number of affected people increasing. There are many risk factors that increase CRC risk, including family or personal history of CRC, smoking, consumption of red meat, obesity, and alcohol consumption. Conversely, increased screening, maintaining healthy body weight, not smoking, and limiting intake of red meat are all associated with reduced CRC morbidity and mortality. Mouse models of CRC were first used in 1928 and have played an important role in understanding CRC biology and treatment and have long been instrumental in clarifying the pathobiology of CRC formation and inhibition. This review focuses on advancements in modeling CRC in mice.
Collapse
Affiliation(s)
- Baktiar O Karim
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University Baltimore, MD 21205, USA
| | | |
Collapse
|