1
|
Madsen RR, Le Marois A, Mruk ON, Voliotis M, Yin S, Sufi J, Qin X, Zhao SJ, Gorczynska J, Morelli D, Davidson L, Sahai E, Korolchuk VI, Tape CJ, Vanhaesebroeck B. Oncogenic PIK3CA corrupts growth factor signaling specificity. Mol Syst Biol 2024:10.1038/s44320-024-00078-x. [PMID: 39706867 DOI: 10.1038/s44320-024-00078-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/06/2024] [Accepted: 11/11/2024] [Indexed: 12/23/2024] Open
Abstract
Technical limitations have prevented understanding of how growth factor signals are encoded in distinct activity patterns of the phosphoinositide 3-kinase (PI3K)/AKT pathway, and how this is altered by oncogenic pathway mutations. We introduce a kinetic, single-cell framework for precise calculations of PI3K-specific information transfer for different growth factors. This features live-cell imaging of PI3K/AKT activity reporters and multiplexed CyTOF measurements of PI3K/AKT and RAS/ERK signaling markers over time. Using this framework, we found that the PIK3CAH1047R oncogene was not a simple, constitutive activator of the pathway as often presented. Dose-dependent expression of PIK3CAH1047R in human cervical cancer and induced pluripotent stem cells corrupted the fidelity of growth factor-induced information transfer, with preferential amplification of epidermal growth factor receptor (EGFR) signaling responses compared to insulin-like growth factor 1 (IGF1) and insulin receptor signaling. PIK3CAH1047R did not only shift these responses to a higher mean but also enhanced signaling heterogeneity. We conclude that oncogenic PIK3CAH1047R corrupts information transfer in a growth factor-dependent manner and suggest new opportunities for tuning of receptor-specific PI3K pathway outputs for therapeutic benefit.
Collapse
Affiliation(s)
- Ralitsa R Madsen
- Cell Signaling Laboratory, Department of Oncology, University College London Cancer Institute Paul O'Gorman Building, University College London, London, WC1E 6BT, UK.
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK.
| | - Alix Le Marois
- Tumour Cell Biology Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | - Oliwia N Mruk
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK
| | - Margaritis Voliotis
- Department of Mathematics and Statistics and Living Systems Institute; University of Exeter, Exeter, EX4 4QD, UK
| | - Shaozhen Yin
- Cell Signaling Laboratory, Department of Oncology, University College London Cancer Institute Paul O'Gorman Building, University College London, London, WC1E 6BT, UK
| | - Jahangir Sufi
- Cell Communication Lab, Department of Oncology, University College London Cancer Institute, London, WC1E 6BT, UK
| | - Xiao Qin
- Cell Communication Lab, Department of Oncology, University College London Cancer Institute, London, WC1E 6BT, UK
- MRC Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine, Oxford, OX3 9DS, UK
| | - Salome J Zhao
- Cell Signaling Laboratory, Department of Oncology, University College London Cancer Institute Paul O'Gorman Building, University College London, London, WC1E 6BT, UK
| | - Julia Gorczynska
- Cell Signaling Laboratory, Department of Oncology, University College London Cancer Institute Paul O'Gorman Building, University College London, London, WC1E 6BT, UK
| | - Daniele Morelli
- Cell Signaling Laboratory, Department of Oncology, University College London Cancer Institute Paul O'Gorman Building, University College London, London, WC1E 6BT, UK
| | - Lindsay Davidson
- Human Pluripotent Stem Cell Facility, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK
| | - Erik Sahai
- Tumour Cell Biology Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | - Viktor I Korolchuk
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE4 5PL, UK
| | - Christopher J Tape
- Cell Communication Lab, Department of Oncology, University College London Cancer Institute, London, WC1E 6BT, UK
| | - Bart Vanhaesebroeck
- Cell Signaling Laboratory, Department of Oncology, University College London Cancer Institute Paul O'Gorman Building, University College London, London, WC1E 6BT, UK
| |
Collapse
|
2
|
Sharpe BP, Nazlamova LA, Tse C, Johnston DA, Thomas J, Blyth R, Pickering OJ, Grace B, Harrington J, Rajak R, Rose-Zerilli M, Walters ZS, Underwood TJ. Patient-derived tumor organoid and fibroblast assembloid models for interrogation of the tumor microenvironment in esophageal adenocarcinoma. CELL REPORTS METHODS 2024; 4:100909. [PMID: 39610249 PMCID: PMC11704619 DOI: 10.1016/j.crmeth.2024.100909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 07/01/2024] [Accepted: 11/06/2024] [Indexed: 11/30/2024]
Abstract
The tumor microenvironment (TME) comprises all non-tumor elements of cancer and strongly influences disease progression and phenotype. To understand tumor biology and accurately test new therapeutic strategies, representative models should contain both tumor cells and normal cells of the TME. Here, we describe and characterize co-culture tumor-derived organoids and cancer-associated fibroblasts (CAFs), a major component of the TME, in matrix-embedded assembloid models of esophageal adenocarcinoma (EAC). We demonstrate that the assembloid models faithfully recapitulate the differentiation status of EAC and different CAF phenotypes found in the EAC patient TME. We evaluate cell phenotypes by combining tissue-clearing techniques with whole-mount immunofluorescence and histology, providing a practical framework for the characterization of cancer assembloids.
Collapse
Affiliation(s)
- Benjamin P Sharpe
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
| | - Liliya A Nazlamova
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; Early Cancer Institute, University of Cambridge, Department of Oncology, Box 197 Cambridge Biomedical Campus, Hills Road, Cambridge SB2 0XZ, UK
| | - Carmen Tse
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
| | - David A Johnston
- Biomedical Imaging Unit, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
| | - Jaya Thomas
- Bio-R Bioinformatics Research Facility, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
| | - Rhianna Blyth
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
| | - Oliver J Pickering
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - Ben Grace
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; Southampton Clinical Trials Unit, University of Southampton, Southampton, UK
| | - Jack Harrington
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
| | - Rushda Rajak
- University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - Matthew Rose-Zerilli
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
| | - Zoe S Walters
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
| | - Tim J Underwood
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; Bio-R Bioinformatics Research Facility, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK.
| |
Collapse
|
3
|
Zhang Y, Meng R, Sha D, Gao H, Wang S, Zhou J, Wang X, Li F, Li X, Song W. Advances in the application of colorectal cancer organoids in precision medicine. Front Oncol 2024; 14:1506606. [PMID: 39697234 PMCID: PMC11653019 DOI: 10.3389/fonc.2024.1506606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 11/18/2024] [Indexed: 12/20/2024] Open
Abstract
Colorectal cancer (CRC) ranks among the most prevalent gastrointestinal tumors globally and poses a significant threat to human health. In recent years, tumor organoids have emerged as ideal models for clinical disease research owing to their ability to closely mimic the original tumor tissue and maintain a stable phenotypic structure. Organoid technology has found widespread application in basic tumor research, precision therapy, and new drug development, establishing itself as a reliable preclinical model in CRC research. This has significantly advanced individualized and precise tumor therapies. Additionally, the integration of single-cell technology has enhanced the precision of organoid studies, offering deeper insights into tumor heterogeneity and treatment response, thereby contributing to the development of personalized treatment approaches. This review outlines the evolution of colorectal cancer organoid technology and highlights its strengths in modeling colorectal malignancies. This review also summarizes the progress made in precision tumor medicine and addresses the challenges in organoid research, particularly when organoid research is combined with single-cell technology. Furthermore, this review explores the future potential of organoid technology in the standardization of culture techniques, high-throughput screening applications, and single-cell multi-omics integration, offering novel directions for future colorectal cancer research.
Collapse
Affiliation(s)
- Yanan Zhang
- The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
- Department of Oncology, Zibo Hospital of Traditional Chinese Medicine, Zibo, China
| | - Ruoyu Meng
- Department of Minimally Invasive Comprehensive Treatment of Cancer, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Dan Sha
- Department of Minimally Invasive Comprehensive Treatment of Cancer, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Huiquan Gao
- Department of Radiotherapy, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Shengxi Wang
- Department of Minimally Invasive Comprehensive Treatment of Cancer, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Jun Zhou
- Department of Minimally Invasive Comprehensive Treatment of Cancer, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Xiaoshan Wang
- The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Fuxia Li
- Department of Minimally Invasive Comprehensive Treatment of Cancer, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Xinyu Li
- Department of Minimally Invasive Comprehensive Treatment of Cancer, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Wei Song
- Department of Minimally Invasive Comprehensive Treatment of Cancer, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
4
|
Kabiljo J, Theophil A, Homola J, Renner AF, Stürzenbecher N, Ammon D, Zirnbauer R, Stang S, Tran L, Laengle J, Kulu A, Chen A, Fabits M, Atanasova VS, Pusch O, Weninger W, Walczak H, Herndler Brandstetter D, Egger G, Dolznig H, Kusienicka A, Farlik M, Bergmann M. Cancer-associated fibroblasts shape early myeloid cell response to chemotherapy-induced immunogenic signals in next generation tumor organoid cultures. J Immunother Cancer 2024; 12:e009494. [PMID: 39500527 PMCID: PMC11535717 DOI: 10.1136/jitc-2024-009494] [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] [Accepted: 09/26/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND Patient-derived colorectal cancer (CRC) organoids (PDOs) solely consisting of malignant cells led to major advances in the understanding of cancer treatments. Yet, a major limitation is the absence of cells from the tumor microenvironment, thereby prohibiting potential investigation of treatment responses on immune and structural cells. Currently there are sparse reports describing the interaction of PDOs, cancer-associated fibroblasts (CAFs) and tumor-associated macrophages (TAMs) in complex primary co-culture assay systems. METHODS Primary PDOs and patient matched CAF cultures were generated from surgical resections. Co-culture systems of PDOs, CAFs and monocytic myeloid cells were set up to recapitulate features seen in patient tumors. Single-cell transcriptomics and flow cytometry was used to show effects of culture systems on TAM populations in the co-culture assays under chemotherapeutic and oncolytic viral treatment. RESULTS In contrast to co-cultures of tumor cells and monocytes, CAF/monocyte co-cultures and CAF/monocyte/tumor cell triple cultures resulted in a partial differentiation into macrophages and a phenotypic switch, characterized by the expression of major immunosuppressive markers comparable to TAMs in CRC. Oxaliplatin and 5-fluorouracil, the standard-of-care chemotherapy for CRC, induced polarization of macrophages to a pro-inflammatory phenotype comparable to the immunogenic effects of treatment with an oncolytic virus. Monitoring phagocytosis as a functional proxy to macrophage activation and subsequent onset of an immune response, revealed that chemotherapy-induced cell death, but not virus-mediated cell death, is necessary to induce phagocytosis of CRC cells. Moreover, CAFs enhanced the phagocytic activity in chemotherapy treated CRC triple cultures. CONCLUSIONS Primary CAF-containing triple cultures successfully model TAM-like phenotypes ex vivo and allow the assessment of their functional and phenotypic changes in response to treatments following a precision medicine approach.
Collapse
Affiliation(s)
- Julijan Kabiljo
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute Applied Diagnostics, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Anna Theophil
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute Applied Diagnostics, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Jakob Homola
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Annalena F Renner
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Nathalie Stürzenbecher
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Daphni Ammon
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Rebecca Zirnbauer
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Simone Stang
- Center of Pathobiochemistry and Genetics, Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria
| | - Loan Tran
- Ludwig Boltzmann Institute Applied Diagnostics, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Johannes Laengle
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute Applied Diagnostics, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Askin Kulu
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Anna Chen
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Markus Fabits
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Velina S Atanasova
- Center of Pathobiochemistry and Genetics, Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria
| | - Oliver Pusch
- Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Weninger
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Henning Walczak
- Institute for Biochemistry I, Medical Faculty, University of Cologne, Cologne, Germany
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, London, UK
| | - Dietmar Herndler Brandstetter
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Gerda Egger
- Ludwig Boltzmann Institute Applied Diagnostics, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Helmut Dolznig
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- Center of Pathobiochemistry and Genetics, Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria
| | - Anna Kusienicka
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Matthias Farlik
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Michael Bergmann
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute Applied Diagnostics, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
5
|
Özkan A, LoGrande NT, Feitor JF, Goyal G, Ingber DE. Intestinal organ chips for disease modelling and personalized medicine. Nat Rev Gastroenterol Hepatol 2024; 21:751-773. [PMID: 39192055 DOI: 10.1038/s41575-024-00968-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/10/2024] [Indexed: 08/29/2024]
Abstract
Alterations in intestinal structure, mechanics and physiology underlie acute and chronic intestinal conditions, many of which are influenced by dysregulation of microbiome, peristalsis, stroma or immune responses. Studying human intestinal physiology or pathophysiology is difficult in preclinical animal models because their microbiomes and immune systems differ from those of humans. Although advances in organoid culture partially overcome this challenge, intestinal organoids still lack crucial features that are necessary to study functions central to intestinal health and disease, such as digestion or fluid flow, as well as contributions from long-term effects of living microbiome, peristalsis and immune cells. Here, we review developments in organ-on-a-chip (organ chip) microfluidic culture models of the human intestine that are lined by epithelial cells and interfaced with other tissues (such as stroma or endothelium), which can experience both fluid flow and peristalsis-like motions. Organ chips offer powerful ways to model intestinal physiology and disease states for various human populations and individual patients, and can be used to gain new insight into underlying molecular and biophysical mechanisms of disease. They can also be used as preclinical tools to discover new drugs and then validate their therapeutic efficacy and safety in the same human-relevant model.
Collapse
Affiliation(s)
- Alican Özkan
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Nina Teresa LoGrande
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Jessica F Feitor
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Girija Goyal
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA.
- Vascular Biology Program and Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA.
- Harvard John A. Paulson School of Engineering and Applied Sciences, Cambridge, MA, USA.
| |
Collapse
|
6
|
Qin X, Tape CJ. Functional analysis of cell plasticity using single-cell technologies. Trends Cell Biol 2024; 34:854-864. [PMID: 38355348 DOI: 10.1016/j.tcb.2024.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 01/09/2024] [Accepted: 01/15/2024] [Indexed: 02/16/2024]
Abstract
Metazoan organisms are heterocellular systems composed of hundreds of different cell types, which arise from an isogenic genome through differentiation. Cellular 'plasticity' further enables cells to alter their fate in response to exogenous cues and is involved in a variety of processes, such as wound healing, infection, and cancer. Recent advances in cellular model systems, high-dimensional single-cell technologies, and lineage tracing have sparked a renaissance in plasticity research. Here, we discuss the definition of cell plasticity, evaluate state-of-the-art model systems and techniques to study cell-fate dynamics, and explore the application of single-cell technologies to obtain functional insights into cell plasticity in healthy and diseased tissues. The integration of advanced biomimetic model systems, single-cell technologies, and high-throughput perturbation studies is enabling a new era of research into non-genetic plasticity in metazoan systems.
Collapse
Affiliation(s)
- Xiao Qin
- MRC Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine, Oxford, OX3 9DS, UK.
| | - Christopher J Tape
- Cell Communication Lab, Department of Oncology, University College London Cancer Institute, 72 Huntley Street, London, WC1E 6DD, UK.
| |
Collapse
|
7
|
Love JR, Karthaus WR. Next-Generation Modeling of Cancer Using Organoids. Cold Spring Harb Perspect Med 2024; 14:a041380. [PMID: 37734867 PMCID: PMC11146310 DOI: 10.1101/cshperspect.a041380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/23/2023]
Abstract
In the last decade, organoid technology has become a cornerstone in cancer research. Organoids are long-term primary cell cultures, usually of epithelial origin, grown in a three-dimensional (3D) protein matrix and a fully defined medium. Organoids can be derived from many organs and cancer types and sites, encompassing both murine and human tissues. Importantly, they can be established from various stages during tumor evolution and recapitulate with high accuracy patient genomics and phenotypes in vitro, offering a platform for personalized medicine. Additionally, organoids are remarkably amendable for experimental manipulation. Taken together, these features make organoids a powerful tool with applications in basic cancer research and personalized medicine. Here, we will discuss the origins of organoid culture, applications in cancer research, and how cancer organoids can synergize with other models of cancer to drive basic discoveries as well as to translate these toward clinical solutions.
Collapse
Affiliation(s)
- Jillian R Love
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, EPFL, 1015 Lausanne, Switzerland
| | - Wouter R Karthaus
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, EPFL, 1015 Lausanne, Switzerland
| |
Collapse
|
8
|
Cadavid JL, Li NT, McGuigan AP. Bridging systems biology and tissue engineering: Unleashing the full potential of complex 3D in vitro tissue models of disease. BIOPHYSICS REVIEWS 2024; 5:021301. [PMID: 38617201 PMCID: PMC11008916 DOI: 10.1063/5.0179125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 03/12/2024] [Indexed: 04/16/2024]
Abstract
Rapid advances in tissue engineering have resulted in more complex and physiologically relevant 3D in vitro tissue models with applications in fundamental biology and therapeutic development. However, the complexity provided by these models is often not leveraged fully due to the reductionist methods used to analyze them. Computational and mathematical models developed in the field of systems biology can address this issue. Yet, traditional systems biology has been mostly applied to simpler in vitro models with little physiological relevance and limited cellular complexity. Therefore, integrating these two inherently interdisciplinary fields can result in new insights and move both disciplines forward. In this review, we provide a systematic overview of how systems biology has been integrated with 3D in vitro tissue models and discuss key application areas where the synergies between both fields have led to important advances with potential translational impact. We then outline key directions for future research and discuss a framework for further integration between fields.
Collapse
|
9
|
Fowler D, Barisa M, Southern A, Nattress C, Hawkins E, Vassalou E, Kanouta A, Counsell J, Rota E, Vlckova P, Draper B, De Mooij T, Farkas A, Brezovjakova H, Baker AT, Scotlandi K, Manara MC, Tape C, Chester K, Anderson J, Fisher J. Payload-delivering engineered γδ T cells display enhanced cytotoxicity, persistence, and efficacy in preclinical models of osteosarcoma. Sci Transl Med 2024; 16:eadg9814. [PMID: 38809963 DOI: 10.1126/scitranslmed.adg9814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 04/23/2024] [Indexed: 05/31/2024]
Abstract
T cell-based cancer immunotherapy has typically relied on membrane-bound cytotoxicity enhancers such as chimeric antigen receptors expressed in autologous αβ T cells. These approaches are limited by tonic signaling of synthetic constructs and costs associated with manufacturing. γδ T cells are an emerging alternative for cellular therapy, having innate antitumor activity, potent antibody-dependent cellular cytotoxicity, and minimal alloreactivity. We present an immunotherapeutic platform technology built around the innate properties of the Vγ9Vδ2 T cell, harnessing specific characteristics of this cell type and offering an allocompatible cellular therapy that recruits bystander immunity. We engineered γδ T cells to secrete synthetic tumor-targeting opsonins in the form of an scFv-Fc fusion protein and a mitogenic IL-15Rα-IL-15 fusion protein (stIL15). Using GD2 as a model antigen, we show that GD2-specific opsonin-secreting Vγ9Vδ2 T cells (stIL15-OPS-γδ T cells) have enhanced cytotoxicity and promote bystander activity of other lymphoid and myeloid cells. Secretion of stIL-15 abrogated the need for exogenous cytokine supplementation and further mediated activation of bystander natural killer cells. Compared with unmodified γδ T cells, stIL15-OPS-γδ T cells exhibited superior in vivo control of subcutaneous tumors and persistence in the blood. Moreover, stIL15-OPS-γδ T cells were efficacious against patient-derived osteosarcomas in animal models and in vitro, where efficacy could be boosted with the addition of zoledronic acid. Together, the data identify stIL15-OPS-γδ T cells as a candidate allogeneic cell therapy platform combining direct cytolysis with bystander activation to promote tumor control.
Collapse
Affiliation(s)
- Daniel Fowler
- UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research, 20 Guilford Street, WC1N 1DZ London, UK
| | - Marta Barisa
- UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research, 20 Guilford Street, WC1N 1DZ London, UK
| | - Alba Southern
- UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research, 20 Guilford Street, WC1N 1DZ London, UK
| | - Callum Nattress
- UCL Cancer Institute, Paul O'Gorman Building, 72 Huntley Street, WC1E 6DD London, UK
| | - Elizabeth Hawkins
- UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research, 20 Guilford Street, WC1N 1DZ London, UK
| | - Eleni Vassalou
- UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research, 20 Guilford Street, WC1N 1DZ London, UK
| | - Angeliki Kanouta
- UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research, 20 Guilford Street, WC1N 1DZ London, UK
| | | | - Enrique Rota
- UCL Cancer Institute, Paul O'Gorman Building, 72 Huntley Street, WC1E 6DD London, UK
| | - Petra Vlckova
- UCL Cancer Institute, Paul O'Gorman Building, 72 Huntley Street, WC1E 6DD London, UK
| | - Benjamin Draper
- UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research, 20 Guilford Street, WC1N 1DZ London, UK
| | - Tessa De Mooij
- UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research, 20 Guilford Street, WC1N 1DZ London, UK
| | - Andrea Farkas
- UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research, 20 Guilford Street, WC1N 1DZ London, UK
| | - Helena Brezovjakova
- UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research, 20 Guilford Street, WC1N 1DZ London, UK
| | - Alfie T Baker
- UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research, 20 Guilford Street, WC1N 1DZ London, UK
| | - Katia Scotlandi
- IRCCS Istituto Ortopedico Rizzoli, Experimental Oncology Laboratory, Via di Barbiano 1/10, 40136 Bologna Italy
| | - Maria C Manara
- IRCCS Istituto Ortopedico Rizzoli, Experimental Oncology Laboratory, Via di Barbiano 1/10, 40136 Bologna Italy
| | - Chris Tape
- UCL Cancer Institute, Paul O'Gorman Building, 72 Huntley Street, WC1E 6DD London, UK
| | - Kerry Chester
- UCL Cancer Institute, Paul O'Gorman Building, 72 Huntley Street, WC1E 6DD London, UK
| | - John Anderson
- UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research, 20 Guilford Street, WC1N 1DZ London, UK
| | - Jonathan Fisher
- UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research, 20 Guilford Street, WC1N 1DZ London, UK
| |
Collapse
|
10
|
Kalla J, Pfneissl J, Mair T, Tran L, Egger G. A systematic review on the culture methods and applications of 3D tumoroids for cancer research and personalized medicine. Cell Oncol (Dordr) 2024:10.1007/s13402-024-00960-8. [PMID: 38806997 DOI: 10.1007/s13402-024-00960-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2024] [Indexed: 05/30/2024] Open
Abstract
Cancer is a highly heterogeneous disease, and thus treatment responses vary greatly between patients. To improve therapy efficacy and outcome for cancer patients, more representative and patient-specific preclinical models are needed. Organoids and tumoroids are 3D cell culture models that typically retain the genetic and epigenetic characteristics, as well as the morphology, of their tissue of origin. Thus, they can be used to understand the underlying mechanisms of cancer initiation, progression, and metastasis in a more physiological setting. Additionally, co-culture methods of tumoroids and cancer-associated cells can help to understand the interplay between a tumor and its tumor microenvironment. In recent years, tumoroids have already helped to refine treatments and to identify new targets for cancer therapy. Advanced culturing systems such as chip-based fluidic devices and bioprinting methods in combination with tumoroids have been used for high-throughput applications for personalized medicine. Even though organoid and tumoroid models are complex in vitro systems, validation of results in vivo is still the common practice. Here, we describe how both animal- and human-derived tumoroids have helped to identify novel vulnerabilities for cancer treatment in recent years, and how they are currently used for precision medicine.
Collapse
Affiliation(s)
- Jessica Kalla
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Janette Pfneissl
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Theresia Mair
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Loan Tran
- Department of Pathology, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute Applied Diagnostics, Vienna, Austria
| | - Gerda Egger
- Department of Pathology, Medical University of Vienna, Vienna, Austria.
- Ludwig Boltzmann Institute Applied Diagnostics, Vienna, Austria.
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
11
|
Tape CJ. Plastic persisters: revival stem cells in colorectal cancer. Trends Cancer 2024; 10:185-195. [PMID: 38071119 DOI: 10.1016/j.trecan.2023.11.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/03/2023] [Accepted: 11/07/2023] [Indexed: 03/16/2024]
Abstract
Colorectal cancer (CRC) is traditionally considered to be a genetically driven disease. However, nongenetic plasticity has recently emerged as a major driver of tumour initiation, metastasis, and therapy response in CRC. Central to these processes is a recently discovered cell type, the revival colonic stem cell (revCSC). In contrast to traditional proliferative CSCs (proCSCs), revCSCs prioritise survival over propagation. revCSCs play an essential role in primary tumour formation, metastatic dissemination, and nongenetic chemoresistance. Current evidence suggests that CRC tumours leverage intestinal stem cell plasticity to both proliferate (via proCSCs) when unchallenged and survive (via revCSCs) in response to cell-extrinsic pressures. Although revCSCs likely represent a major source of therapeutic failure in CRC, our increasing knowledge of this important stem cell fate provides novel opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- Christopher J Tape
- Cell Communication Lab, Department of Oncology, University College London Cancer Institute, 72 Huntley Street, London, WC1E 6DD, UK.
| |
Collapse
|
12
|
Ramos Zapatero M, Tong A, Opzoomer JW, O'Sullivan R, Cardoso Rodriguez F, Sufi J, Vlckova P, Nattress C, Qin X, Claus J, Hochhauser D, Krishnaswamy S, Tape CJ. Trellis tree-based analysis reveals stromal regulation of patient-derived organoid drug responses. Cell 2023; 186:5606-5619.e24. [PMID: 38065081 DOI: 10.1016/j.cell.2023.11.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 07/27/2023] [Accepted: 11/02/2023] [Indexed: 12/18/2023]
Abstract
Patient-derived organoids (PDOs) can model personalized therapy responses; however, current screening technologies cannot reveal drug response mechanisms or how tumor microenvironment cells alter therapeutic performance. To address this, we developed a highly multiplexed mass cytometry platform to measure post-translational modification (PTM) signaling, DNA damage, cell-cycle activity, and apoptosis in >2,500 colorectal cancer (CRC) PDOs and cancer-associated fibroblasts (CAFs) in response to clinical therapies at single-cell resolution. To compare patient- and microenvironment-specific drug responses in thousands of single-cell datasets, we developed "Trellis"-a highly scalable, tree-based treatment effect analysis method. Trellis single-cell screening revealed that on-target cell-cycle blockage and DNA-damage drug effects are common, even in chemorefractory PDOs. However, drug-induced apoptosis is rarer, patient-specific, and aligns with cancer cell PTM signaling. We find that CAFs can regulate PDO plasticity-shifting proliferative colonic stem cells (proCSCs) to slow-cycling revival colonic stem cells (revCSCs) to protect cancer cells from chemotherapy.
Collapse
Affiliation(s)
- María Ramos Zapatero
- Cell Communication Lab, Department of Oncology, University College London Cancer Institute, London WC1E 6DD, UK
| | - Alexander Tong
- Department of Computer Science, Yale University, New Haven, CT, USA; Department of Computer Science and Operations Research, Université de Montréal, Montreal, QC, Canada; Mila - Quebec AI Institute, Montréal, QC, Canada
| | - James W Opzoomer
- Cell Communication Lab, Department of Oncology, University College London Cancer Institute, London WC1E 6DD, UK
| | - Rhianna O'Sullivan
- Cell Communication Lab, Department of Oncology, University College London Cancer Institute, London WC1E 6DD, UK
| | - Ferran Cardoso Rodriguez
- Cell Communication Lab, Department of Oncology, University College London Cancer Institute, London WC1E 6DD, UK
| | - Jahangir Sufi
- Cell Communication Lab, Department of Oncology, University College London Cancer Institute, London WC1E 6DD, UK
| | - Petra Vlckova
- Cell Communication Lab, Department of Oncology, University College London Cancer Institute, London WC1E 6DD, UK
| | - Callum Nattress
- Cell Communication Lab, Department of Oncology, University College London Cancer Institute, London WC1E 6DD, UK
| | - Xiao Qin
- Cell Communication Lab, Department of Oncology, University College London Cancer Institute, London WC1E 6DD, UK
| | - Jeroen Claus
- Phospho Biomedical Animation, The Greenhouse Studio 6, London N17 9QU, UK
| | - Daniel Hochhauser
- Drug-DNA Interactions Group, Department of Oncology, University College London Cancer Institute, London WC1E 6DD, UK
| | - Smita Krishnaswamy
- Department of Computer Science, Yale University, New Haven, CT, USA; Department of Genetics, Yale University, New Haven, CT, USA; Program for Computational Biology & Bioinformatics, Yale University, New Haven, CT, USA; Program for Applied Math, Yale University, New Haven, CT, USA; Wu-Tsai Institute, Yale University, New Haven, CT, USA.
| | - Christopher J Tape
- Cell Communication Lab, Department of Oncology, University College London Cancer Institute, London WC1E 6DD, UK.
| |
Collapse
|
13
|
Qin X, Cardoso Rodriguez F, Sufi J, Vlckova P, Claus J, Tape CJ. An oncogenic phenoscape of colonic stem cell polarization. Cell 2023; 186:5554-5568.e18. [PMID: 38065080 DOI: 10.1016/j.cell.2023.11.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 09/14/2023] [Accepted: 11/02/2023] [Indexed: 12/18/2023]
Abstract
Cancer cells are regulated by oncogenic mutations and microenvironmental signals, yet these processes are often studied separately. To functionally map how cell-intrinsic and cell-extrinsic cues co-regulate cell fate, we performed a systematic single-cell analysis of 1,107 colonic organoid cultures regulated by (1) colorectal cancer (CRC) oncogenic mutations, (2) microenvironmental fibroblasts and macrophages, (3) stromal ligands, and (4) signaling inhibitors. Multiplexed single-cell analysis revealed a stepwise epithelial differentiation phenoscape dictated by combinations of oncogenes and stromal ligands, spanning from fibroblast-induced Clusterin (CLU)+ revival colonic stem cells (revCSCs) to oncogene-driven LRIG1+ hyper-proliferative CSCs (proCSCs). The transition from revCSCs to proCSCs is regulated by decreasing WNT3A and TGF-β-driven YAP signaling and increasing KRASG12D or stromal EGF/Epiregulin-activated MAPK/PI3K flux. We find that APC loss and KRASG12D collaboratively limit access to revCSCs and disrupt stromal-epithelial communication-trapping epithelia in the proCSC fate. These results reveal that oncogenic mutations dominate homeostatic differentiation by obstructing cell-extrinsic regulation of cell-fate plasticity.
Collapse
Affiliation(s)
- Xiao Qin
- Cell Communication Lab, Department of Oncology, University College London Cancer Institute, 72 Huntley Street, London WC1E 6DD, UK
| | - Ferran Cardoso Rodriguez
- Cell Communication Lab, Department of Oncology, University College London Cancer Institute, 72 Huntley Street, London WC1E 6DD, UK
| | - Jahangir Sufi
- Cell Communication Lab, Department of Oncology, University College London Cancer Institute, 72 Huntley Street, London WC1E 6DD, UK
| | - Petra Vlckova
- Cell Communication Lab, Department of Oncology, University College London Cancer Institute, 72 Huntley Street, London WC1E 6DD, UK
| | - Jeroen Claus
- Phospho Biomedical Animation, The Greenhouse Studio 6, London N17 9QU, UK
| | - Christopher J Tape
- Cell Communication Lab, Department of Oncology, University College London Cancer Institute, 72 Huntley Street, London WC1E 6DD, UK.
| |
Collapse
|
14
|
Madsen RR, Toker A. PI3K signaling through a biochemical systems lens. J Biol Chem 2023; 299:105224. [PMID: 37673340 PMCID: PMC10570132 DOI: 10.1016/j.jbc.2023.105224] [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: 05/08/2023] [Revised: 08/25/2023] [Accepted: 08/28/2023] [Indexed: 09/08/2023] Open
Abstract
Following 3 decades of extensive research into PI3K signaling, it is now evidently clear that the underlying network does not equate to a simple ON/OFF switch. This is best illustrated by the multifaceted nature of the many diseases associated with aberrant PI3K signaling, including common cancers, metabolic disease, and rare developmental disorders. However, we are still far from a complete understanding of the fundamental control principles that govern the numerous phenotypic outputs that are elicited by activation of this well-characterized biochemical signaling network, downstream of an equally diverse set of extrinsic inputs. At its core, this is a question on the role of PI3K signaling in cellular information processing and decision making. Here, we review the determinants of accurate encoding and decoding of growth factor signals and discuss outstanding questions in the PI3K signal relay network. We emphasize the importance of quantitative biochemistry, in close integration with advances in single-cell time-resolved signaling measurements and mathematical modeling.
Collapse
Affiliation(s)
- Ralitsa R Madsen
- MRC-Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, Scotland, United Kingdom.
| | - Alex Toker
- Department of Pathology and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
15
|
Landon‐Brace N, Li NT, McGuigan AP. Exploring New Dimensions of Tumor Heterogeneity: The Application of Single Cell Analysis to Organoid-Based 3D In Vitro Models. Adv Healthc Mater 2023; 12:e2300903. [PMID: 37589373 PMCID: PMC11468421 DOI: 10.1002/adhm.202300903] [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: 03/21/2023] [Revised: 06/28/2023] [Indexed: 08/18/2023]
Abstract
Modeling the heterogeneity of the tumor microenvironment (TME) in vitro is essential to investigating fundamental cancer biology and developing novel treatment strategies that holistically address the factors affecting tumor progression and therapeutic response. Thus, the development of new tools for both in vitro modeling, such as patient-derived organoids (PDOs) and complex 3D in vitro models, and single cell omics analysis, such as single-cell RNA-sequencing, represents a new frontier for investigating tumor heterogeneity. Specifically, the integration of PDO-based 3D in vitro models and single cell analysis offers a unique opportunity to explore the intersecting effects of interpatient, microenvironmental, and tumor cell heterogeneity on cell phenotypes in the TME. In this review, the current use of PDOs in complex 3D in vitro models of the TME is discussed and the emerging directions in the development of these models are highlighted. Next, work that has successfully applied single cell analysis to PDO-based models is examined and important experimental considerations are identified for this approach. Finally, open questions are highlighted that may be amenable to exploration using the integration of PDO-based models and single cell analysis. Ultimately, such investigations may facilitate the identification of novel therapeutic targets for cancer that address the significant influence of tumor-TME interactions.
Collapse
Affiliation(s)
- Natalie Landon‐Brace
- Institute of Biomedical EngineeringUniversity of Toronto200 College StreetTorontoM5S3E5Canada
| | - Nancy T. Li
- Department of Chemical Engineering and Applied ChemistryUniversity of Toronto200 College StTorontoM5S3E5Canada
| | - Alison P. McGuigan
- Department of Chemical Engineering and Applied ChemistryInstitute of Biomedical EngineeringUniversity of Toronto200 College StTorontoM5S3E5Canada
| |
Collapse
|
16
|
Sockell A, Wong W, Longwell S, Vu T, Karlsson K, Mokhtari D, Schaepe J, Lo YH, Cornelius V, Kuo C, Van Valen D, Curtis C, Fordyce PM. A microwell platform for high-throughput longitudinal phenotyping and selective retrieval of organoids. Cell Syst 2023; 14:764-776.e6. [PMID: 37734323 DOI: 10.1016/j.cels.2023.08.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 02/24/2023] [Accepted: 08/22/2023] [Indexed: 09/23/2023]
Abstract
Organoids are powerful experimental models for studying the ontogeny and progression of various diseases including cancer. Organoids are conventionally cultured in bulk using an extracellular matrix mimic. However, bulk-cultured organoids physically overlap, making it impossible to track the growth of individual organoids over time in high throughput. Moreover, local spatial variations in bulk matrix properties make it difficult to assess whether observed phenotypic heterogeneity between organoids results from intrinsic cell differences or differences in the microenvironment. Here, we developed a microwell-based method that enables high-throughput quantification of image-based parameters for organoids grown from single cells, which can further be retrieved from their microwells for molecular profiling. Coupled with a deep learning image-processing pipeline, we characterized phenotypic traits including growth rates, cellular movement, and apical-basal polarity in two CRISPR-engineered human gastric organoid models, identifying genomic changes associated with increased growth rate and changes in accessibility and expression correlated with apical-basal polarity. A record of this paper's transparent peer review process is included in the supplemental information.
Collapse
Affiliation(s)
- Alexandra Sockell
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Wing Wong
- Department of Genetics, Stanford University, Stanford, CA 94305, USA; Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Scott Longwell
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Thy Vu
- Department of Biochemistry, UT Austin, Austin, TX 78712, USA
| | - Kasper Karlsson
- Department of Genetics, Stanford University, Stanford, CA 94305, USA; Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Daniel Mokhtari
- Department of Biochemistry, Stanford University, Stanford, CA 94305, USA
| | - Julia Schaepe
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Yuan-Hung Lo
- Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Vincent Cornelius
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Calvin Kuo
- Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - David Van Valen
- Division of Biology and Bioengineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Christina Curtis
- Department of Genetics, Stanford University, Stanford, CA 94305, USA; Department of Medicine, Stanford University, Stanford, CA 94305, USA; Department of Biomedical Data Science, Stanford University, Stanford, CA 94305, USA; Stanford Cancer Institute, Stanford University, Stanford, CA 94305, USA; Chan Zuckerberg Biohub, San Francisco, CA 94110, USA.
| | - Polly M Fordyce
- Department of Genetics, Stanford University, Stanford, CA 94305, USA; Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Chan Zuckerberg Biohub, San Francisco, CA 94110, USA; ChEM-H Institute, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
17
|
Koladiya A, Davis KL. Advances in Clinical Mass Cytometry. Clin Lab Med 2023; 43:507-519. [PMID: 37481326 DOI: 10.1016/j.cll.2023.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/24/2023]
Abstract
The advent of high-dimensional single-cell technologies has enabled detection of cellular heterogeneity and functional diversity of immune cells during health and disease conditions. Because of its multiplexing capabilities and limited compensation requirements, mass cytometry or cytometry by time of flight (CyTOF) has played a superior role in immune monitoring compared with flow cytometry. Further, it has higher throughput and lower cost compared with other single-cell techniques. Several published articles have utilized CyTOF to identify cellular phenotypes and features associated with disease outcomes. This article introduces CyTOF-based assays to profile immune cell-types, cell-states, and their applications in clinical research.
Collapse
Affiliation(s)
- Abhishek Koladiya
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Kara L Davis
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA; Center for Cancer Cell Therapy, Stanford University, Stanford, CA, USA.
| |
Collapse
|
18
|
Sell T, Klotz C, Fischer MM, Astaburuaga-García R, Krug S, Drost J, Clevers H, Sers C, Morkel M, Blüthgen N. Oncogenic signaling is coupled to colorectal cancer cell differentiation state. J Cell Biol 2023; 222:e202204001. [PMID: 37017636 PMCID: PMC10082329 DOI: 10.1083/jcb.202204001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 12/23/2022] [Accepted: 03/17/2023] [Indexed: 04/06/2023] Open
Abstract
Colorectal cancer progression is intrinsically linked to stepwise deregulation of the intestinal differentiation trajectory. In this process, sequential mutations of APC, KRAS, TP53, and SMAD4 enable oncogenic signaling and establish the hallmarks of cancer. Here, we use mass cytometry of isogenic human colon organoids and patient-derived cancer organoids to capture oncogenic signaling, cell phenotypes, and differentiation states in a high-dimensional single-cell map. We define a differentiation axis in all tumor progression states from normal to cancer. Our data show that colorectal cancer driver mutations shape the distribution of cells along the differentiation axis. In this regard, subsequent mutations can have stem cell promoting or restricting effects. Individual nodes of the cancer cell signaling network remain coupled to the differentiation state, regardless of the presence of driver mutations. We use single-cell RNA sequencing to link the (phospho-)protein signaling network to transcriptomic states with biological and clinical relevance. Our work highlights how oncogenes gradually shape signaling and transcriptomes during tumor progression.
Collapse
Affiliation(s)
- Thomas Sell
- Institute of Pathology, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Institute of Biology, Humboldt University of Berlin, Berlin, Germany
| | - Christian Klotz
- Department of Infectious Diseases, Robert Koch-Institute, Unit 16 Mycotic and Parasitic Agents and Mycobacteria, Berlin, Germany
| | - Matthias M. Fischer
- Institute of Pathology, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Institute of Biology, Humboldt University of Berlin, Berlin, Germany
| | - Rosario Astaburuaga-García
- Institute of Pathology, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Institute of Biology, Humboldt University of Berlin, Berlin, Germany
| | - Susanne Krug
- Department of Gastroenterology, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Rheumatology and Infectious Diseases, Clinical Physiology/Nutritional Medicine, Berlin, Germany
| | - Jarno Drost
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Hans Clevers
- Oncode Institute, Utrecht, Netherlands
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, Utrecht, Netherlands
| | - Christine Sers
- Institute of Pathology, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Institute of Biology, Humboldt University of Berlin, Berlin, Germany
- German Cancer Consortium Partner Site Berlin, German Cancer Research Center, Heidelberg, Germany
| | - Markus Morkel
- Institute of Pathology, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Cancer Consortium Partner Site Berlin, German Cancer Research Center, Heidelberg, Germany
- Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Bioportal Single Cells, Berlin, Germany
| | - Nils Blüthgen
- Institute of Pathology, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Institute of Biology, Humboldt University of Berlin, Berlin, Germany
- German Cancer Consortium Partner Site Berlin, German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
19
|
Lin M, Hartl K, Heuberger J, Beccaceci G, Berger H, Li H, Liu L, Müllerke S, Conrad T, Heymann F, Woehler A, Tacke F, Rajewsky N, Sigal M. Establishment of gastrointestinal assembloids to study the interplay between epithelial crypts and their mesenchymal niche. Nat Commun 2023; 14:3025. [PMID: 37230989 DOI: 10.1038/s41467-023-38780-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 05/15/2023] [Indexed: 05/27/2023] Open
Abstract
The cellular organization of gastrointestinal crypts is orchestrated by different cells of the stromal niche but available in vitro models fail to fully recapitulate the interplay between epithelium and stroma. Here, we establish a colon assembloid system comprising the epithelium and diverse stromal cell subtypes. These assembloids recapitulate the development of mature crypts resembling in vivo cellular diversity and organization, including maintenance of a stem/progenitor cell compartment in the base and their maturation into secretory/absorptive cell types. This process is supported by self-organizing stromal cells around the crypts that resemble in vivo organization, with cell types that support stem cell turnover adjacent to the stem cell compartment. Assembloids that lack BMP receptors either in epithelial or stromal cells fail to undergo proper crypt formation. Our data highlight the crucial role of bidirectional signaling between epithelium and stroma, with BMP as a central determinant of compartmentalization along the crypt axis.
Collapse
Affiliation(s)
- Manqiang Lin
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 10115, Berlin, Germany
| | - Kimberly Hartl
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 10115, Berlin, Germany
| | - Julian Heuberger
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 10115, Berlin, Germany
| | - Giulia Beccaceci
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 10115, Berlin, Germany
| | - Hilmar Berger
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
| | - Hao Li
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 10115, Berlin, Germany
| | - Lichao Liu
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
| | - Stefanie Müllerke
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 10115, Berlin, Germany
| | - Thomas Conrad
- Genomics Technology Platform, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125, Berlin, Germany
- Core Facility Genomics, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 10178, Berlin, Germany
| | - Felix Heymann
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
| | - Andrew Woehler
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 10115, Berlin, Germany
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
| | - Nikolaus Rajewsky
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 10115, Berlin, Germany
| | - Michael Sigal
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany.
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 10115, Berlin, Germany.
| |
Collapse
|
20
|
Arnett LP, Rana R, Chung WWY, Li X, Abtahi M, Majonis D, Bassan J, Nitz M, Winnik MA. Reagents for Mass Cytometry. Chem Rev 2023; 123:1166-1205. [PMID: 36696538 DOI: 10.1021/acs.chemrev.2c00350] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Mass cytometry (cytometry by time-of-flight detection [CyTOF]) is a bioanalytical technique that enables the identification and quantification of diverse features of cellular systems with single-cell resolution. In suspension mass cytometry, cells are stained with stable heavy-atom isotope-tagged reagents, and then the cells are nebulized into an inductively coupled plasma time-of-flight mass spectrometry (ICP-TOF-MS) instrument. In imaging mass cytometry, a pulsed laser is used to ablate ca. 1 μm2 spots of a tissue section. The plume is then transferred to the CyTOF, generating an image of biomarker expression. Similar measurements are possible with multiplexed ion bean imaging (MIBI). The unit mass resolution of the ICP-TOF-MS detector allows for multiparametric analysis of (in principle) up to 130 different parameters. Currently available reagents, however, allow simultaneous measurement of up to 50 biomarkers. As new reagents are developed, the scope of information that can be obtained by mass cytometry continues to increase, particularly due to the development of new small molecule reagents which enable monitoring of active biochemistry at the cellular level. This review summarizes the history and current state of mass cytometry reagent development and elaborates on areas where there is a need for new reagents. Additionally, this review provides guidelines on how new reagents should be tested and how the data should be presented to make them most meaningful to the mass cytometry user community.
Collapse
Affiliation(s)
- Loryn P Arnett
- Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, OntarioM5S 3H6, Canada
| | - Rahul Rana
- Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, OntarioM5S 3H6, Canada
| | - Wilson Wai-Yip Chung
- Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, OntarioM5S 3H6, Canada
| | - Xiaochong Li
- Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, OntarioM5S 3H6, Canada
| | - Mahtab Abtahi
- Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, OntarioM5S 3H6, Canada
| | - Daniel Majonis
- Standard BioTools Canada Inc. (formerly Fluidigm Canada Inc.), 1380 Rodick Road, Suite 400, Markham, OntarioL3R 4G5, Canada
| | - Jay Bassan
- Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, OntarioM5S 3H6, Canada
| | - Mark Nitz
- Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, OntarioM5S 3H6, Canada
| | - Mitchell A Winnik
- Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, OntarioM5S 3H6, Canada.,Department of Chemical Engineering and Applied Chemistry, 200 College Street, Toronto, OntarioM5S 3E5, Canada
| |
Collapse
|
21
|
Yuan J, Li X, Yu S. Cancer organoid co-culture model system: Novel approach to guide precision medicine. Front Immunol 2023; 13:1061388. [PMID: 36713421 PMCID: PMC9877297 DOI: 10.3389/fimmu.2022.1061388] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 12/22/2022] [Indexed: 01/14/2023] Open
Abstract
Three-dimensional cancer organoids derived from self-organizing cancer stems are ex vivo miniatures of tumors that faithfully recapitulate their structure, distinctive cancer features, and genetic signatures. As novel tools, current cancer organoids have been well established and rapidly applied in drug testing, genome editing, and transplantation, with the ultimate aim of entering clinical practice for guiding personalized therapy. However, given that the lack of a tumor microenvironment, including immune cells and fibrous cells, is a major limitation of this emerging methodology, co-culture models inspire high hope for further application of this technology in cancer research. Co-culture of cancer organoids and immune cells or fibroblasts is available to investigate the tumor microenvironment, molecular interactions, and chimeric antigen receptor-engineered lymphocytes in cancer treatment. In light of the recent progress in cancer organoid co-culture models, it is only possible to recognize the advantages and drawbacks of this novel model to exploit its full potential. In this review, we summarize the recent advances in the application of cancer organoids and co-culture models and how they could be improved in the future to benefit cancer research, especially precision medicine.
Collapse
|
22
|
Kumar A, Cai S, Allam M, Henderson S, Ozbeyler M, Saiontz L, Coskun AF. Single-Cell and Spatial Analysis of Emergent Organoid Platforms. Methods Mol Biol 2023; 2660:311-344. [PMID: 37191807 DOI: 10.1007/978-1-0716-3163-8_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Organoids have emerged as a promising advancement of the two-dimensional (2D) culture systems to improve studies in organogenesis, drug discovery, precision medicine, and regenerative medicine applications. Organoids can self-organize as three-dimensional (3D) tissues derived from stem cells and patient tissues to resemble organs. This chapter presents growth strategies, molecular screening methods, and emerging issues of the organoid platforms. Single-cell and spatial analysis resolve organoid heterogeneity to obtain information about the structural and molecular cellular states. Culture media diversity and varying lab-to-lab practices have resulted in organoid-to-organoid variability in morphology and cell compositions. An essential resource is an organoid atlas that can catalog protocols and standardize data analysis for different organoid types. Molecular profiling of individual cells in organoids and data organization of the organoid landscape will impact biomedical applications from basic science to translational use.
Collapse
Affiliation(s)
- Aditi Kumar
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Shuangyi Cai
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Mayar Allam
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Samuel Henderson
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Melissa Ozbeyler
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Lilly Saiontz
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Ahmet F Coskun
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.
- Interdisciplinary Bioengineering Graduate Program, Georgia Institute of Technology, Atlanta, GA, USA.
- Parker H. Petit Institute for Bioengineering and Bioscience, , Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
23
|
Focus on organoids: cooperation and interconnection with extracellular vesicles - Is this the future of in vitro modeling? Semin Cancer Biol 2022; 86:367-381. [PMID: 34896267 DOI: 10.1016/j.semcancer.2021.12.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/29/2021] [Accepted: 12/07/2021] [Indexed: 01/27/2023]
Abstract
Organoids are simplified in vitro model systems of organs that are used for modeling tissue development and disease, drug screening, cell therapy, and personalized medicine. Despite considerable success in the design of organoids, challenges remain in achieving real-life applications. Organoids serve as unique and organized groups of micro physiological systems that are capable of self-renewal and self-organization. Moreover, they exhibit similar organ functionality(ies) as that of tissue(s) of origin. Organoids can be designed from adult stem cells, induced pluripotent stem cells, or embryonic stem cells. They consist of most of the important cell types of the desired tissue/organ along with the topology and cell-cell interactions that are highly similar to those of an in vivo tissue/organ. Organoids have gained interest in human biomedical research, as they demonstrate high promise for use in basic, translational, and applied research. As in vitro models, organoids offer significant opportunities for reducing the reliance and use of experimental animals. In this review, we will provide an overview of organoids, as well as those intercellular communications mediated by extracellular vesicles (EVs), and discuss the importance of organoids in modeling a tumor immune microenvironment (TIME). Organoids can also be exploited to develop a better understanding of intercellular communications mediated by EVs. Also, organoids are useful in mimicking TIME, thereby offering a better-controlled environment for studying various associated biological processes and immune cell types involved in tumor immunity, such as T-cells, macrophages, dendritic cells, and myeloid-derived suppressor cells, among others.
Collapse
|
24
|
Pan XT, Yang XY, Mao TQ, Liu K, Chen ZZ, Ji LN, Jiang DC, Wang K, Gu ZZ, Xia XH. Super-Long SERS Active Single Silver Nanowires for Molecular Imaging in 2D and 3D Cell Culture Models. BIOSENSORS 2022; 12:bios12100875. [PMID: 36291012 PMCID: PMC9599576 DOI: 10.3390/bios12100875] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 10/06/2022] [Accepted: 10/14/2022] [Indexed: 05/21/2023]
Abstract
Establishing a systematic molecular information analysis strategy for cell culture models is of great significance for drug development and tissue engineering technologies. Here, we fabricated single silver nanowires with high surface-enhanced Raman scattering activity to extract SERS spectra in situ from two-dimensional (2D) and three-dimensional (3D) cell culture models. The silver nanowires were super long, flexible and thin enough to penetrate through multiple cells. A single silver nanowire was used in combination with a four-dimensional microcontroller as a cell endoscope for spectrally analyzing the components in cell culture models. Then, we adopted a machine learning algorithm to analyze the obtained spectra. Our results show that the abundance of proteins differs significantly between the 2D and 3D models, and that nucleic acid-rich and protein-rich regions can be distinguished with satisfactory accuracy.
Collapse
Affiliation(s)
- Xiao-Tong Pan
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Xuan-Ye Yang
- Institute of Theoretical and Computational Chemistry, Key Laboratory of Mesoscopic Chemistry of the Ministry of Education (MOE), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Tian-Qi Mao
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Kang Liu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Zao-Zao Chen
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Li-Na Ji
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
- Correspondence: (L.-N.J.); (D.-C.J.); (K.W.)
| | - De-Chen Jiang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
- Correspondence: (L.-N.J.); (D.-C.J.); (K.W.)
| | - Kang Wang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
- Correspondence: (L.-N.J.); (D.-C.J.); (K.W.)
| | - Zhong-Ze Gu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Xing-Hua Xia
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| |
Collapse
|
25
|
Freckmann EC, Sandilands E, Cumming E, Neilson M, Román-Fernández A, Nikolatou K, Nacke M, Lannagan TRM, Hedley A, Strachan D, Salji M, Morton JP, McGarry L, Leung HY, Sansom OJ, Miller CJ, Bryant DM. Traject3d allows label-free identification of distinct co-occurring phenotypes within 3D culture by live imaging. Nat Commun 2022; 13:5317. [PMID: 36085324 PMCID: PMC9463449 DOI: 10.1038/s41467-022-32958-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 08/25/2022] [Indexed: 11/09/2022] Open
Abstract
Single cell profiling by genetic, proteomic and imaging methods has expanded the ability to identify programmes regulating distinct cell states. The 3-dimensional (3D) culture of cells or tissue fragments provides a system to study how such states contribute to multicellular morphogenesis. Whether cells plated into 3D cultures give rise to a singular phenotype or whether multiple biologically distinct phenotypes arise in parallel is largely unknown due to a lack of tools to detect such heterogeneity. Here we develop Traject3d (Trajectory identification in 3D), a method for identifying heterogeneous states in 3D culture and how these give rise to distinct phenotypes over time, from label-free multi-day time-lapse imaging. We use this to characterise the temporal landscape of morphological states of cancer cell lines, varying in metastatic potential and drug resistance, and use this information to identify drug combinations that inhibit such heterogeneity. Traject3d is therefore an important companion to other single-cell technologies by facilitating real-time identification via live imaging of how distinct states can lead to alternate phenotypes that occur in parallel in 3D culture.
Collapse
Affiliation(s)
- Eva C Freckmann
- Institute of Cancer Sciences, University of Glasgow, Glasgow, G61 1HQ, United Kingdom
- The CRUK Beatson Institute, Glasgow, G61 1BD, United Kingdom
| | - Emma Sandilands
- Institute of Cancer Sciences, University of Glasgow, Glasgow, G61 1HQ, United Kingdom
- The CRUK Beatson Institute, Glasgow, G61 1BD, United Kingdom
| | - Erin Cumming
- Institute of Cancer Sciences, University of Glasgow, Glasgow, G61 1HQ, United Kingdom
- The CRUK Beatson Institute, Glasgow, G61 1BD, United Kingdom
| | - Matthew Neilson
- The CRUK Beatson Institute, Glasgow, G61 1BD, United Kingdom
| | - Alvaro Román-Fernández
- Institute of Cancer Sciences, University of Glasgow, Glasgow, G61 1HQ, United Kingdom
- The CRUK Beatson Institute, Glasgow, G61 1BD, United Kingdom
| | - Konstantina Nikolatou
- Institute of Cancer Sciences, University of Glasgow, Glasgow, G61 1HQ, United Kingdom
- The CRUK Beatson Institute, Glasgow, G61 1BD, United Kingdom
| | - Marisa Nacke
- Institute of Cancer Sciences, University of Glasgow, Glasgow, G61 1HQ, United Kingdom
- The CRUK Beatson Institute, Glasgow, G61 1BD, United Kingdom
| | | | - Ann Hedley
- The CRUK Beatson Institute, Glasgow, G61 1BD, United Kingdom
| | - David Strachan
- The CRUK Beatson Institute, Glasgow, G61 1BD, United Kingdom
| | - Mark Salji
- The CRUK Beatson Institute, Glasgow, G61 1BD, United Kingdom
| | - Jennifer P Morton
- Institute of Cancer Sciences, University of Glasgow, Glasgow, G61 1HQ, United Kingdom
- The CRUK Beatson Institute, Glasgow, G61 1BD, United Kingdom
| | - Lynn McGarry
- The CRUK Beatson Institute, Glasgow, G61 1BD, United Kingdom
| | - Hing Y Leung
- Institute of Cancer Sciences, University of Glasgow, Glasgow, G61 1HQ, United Kingdom
- The CRUK Beatson Institute, Glasgow, G61 1BD, United Kingdom
| | - Owen J Sansom
- Institute of Cancer Sciences, University of Glasgow, Glasgow, G61 1HQ, United Kingdom
- The CRUK Beatson Institute, Glasgow, G61 1BD, United Kingdom
| | - Crispin J Miller
- Institute of Cancer Sciences, University of Glasgow, Glasgow, G61 1HQ, United Kingdom
- The CRUK Beatson Institute, Glasgow, G61 1BD, United Kingdom
| | - David M Bryant
- Institute of Cancer Sciences, University of Glasgow, Glasgow, G61 1HQ, United Kingdom.
- The CRUK Beatson Institute, Glasgow, G61 1BD, United Kingdom.
| |
Collapse
|
26
|
Wang Y, Song W, Yu S, Liu Y, Chen YG. Intestinal cellular heterogeneity and disease development revealed by single-cell technology. CELL REGENERATION 2022; 11:26. [PMID: 36045190 PMCID: PMC9433512 DOI: 10.1186/s13619-022-00127-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 07/15/2022] [Indexed: 11/10/2022]
Abstract
The intestinal epithelium is responsible for food digestion and nutrient absorption and plays a critical role in hormone secretion, microorganism defense, and immune response. These functions depend on the integral single-layered intestinal epithelium, which shows diversified cell constitution and rapid self-renewal and presents powerful regeneration plasticity after injury. Derailment of homeostasis of the intestine epithelium leads to the development of diseases, most commonly including enteritis and colorectal cancer. Therefore, it is important to understand the cellular characterization of the intestinal epithelium at the molecular level and the mechanisms underlying its homeostatic maintenance. Single-cell technologies allow us to gain molecular insights at the single-cell level. In this review, we summarize the single-cell RNA sequencing applications to understand intestinal cell characteristics, spatiotemporal evolution, and intestinal disease development.
Collapse
|
27
|
Delgado-Gonzalez A, Laz-Ruiz JA, Cano-Cortes MV, Huang YW, Gonzalez VD, Diaz-Mochon JJ, Fantl WJ, Sanchez-Martin RM. Hybrid Fluorescent Mass-Tag Nanotrackers as Universal Reagents for Long-Term Live-Cell Barcoding. Anal Chem 2022; 94:10626-10635. [PMID: 35866879 PMCID: PMC9352147 DOI: 10.1021/acs.analchem.2c00795] [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] [Indexed: 11/29/2022]
Abstract
![]()
Barcoding and pooling cells for processing as a composite
sample
are critical to minimize technical variability in multiplex technologies.
Fluorescent cell barcoding has been established as a standard method
for multiplexing in flow cytometry analysis. In parallel, mass-tag
barcoding is routinely used to label cells for mass cytometry. Barcode
reagents currently used label intracellular proteins in fixed and
permeabilized cells and, therefore, are not suitable for studies with
live cells in long-term culture prior to analysis. In this study,
we report the development of fluorescent palladium-based hybrid-tag
nanotrackers to barcode live cells for flow and mass cytometry dual-modal
readout. We describe the preparation, physicochemical characterization,
efficiency of cell internalization, and durability of these nanotrackers
in live cells cultured over time. In addition, we demonstrate their
compatibility with standardized cytometry reagents and protocols.
Finally, we validated these nanotrackers for drug response assays
during a long-term coculture experiment with two barcoded cell lines.
This method represents a new and widely applicable advance for fluorescent
and mass-tag barcoding that is independent of protein expression levels
and can be used to label cells before long-term drug studies.
Collapse
Affiliation(s)
- Antonio Delgado-Gonzalez
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Gov-ernment, PTS Granada, Avda. Ilustración 114, 18016 Granada, Spain.,Department of Medicinal & Organic Chemistry and Excellence Research Unit of "Chemistry applied to Biomedi-cine and the Environment", Faculty of Pharmacy, University of Granada, Campus de Cartuja s/n, 18071 Granada, Spain.,Biosanitary Research Institute of Granada (ibs.GRANADA), University Hospitals of Granada-University of Grana-da, 18012 Granada, Spain.,Department of Urology, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Jose Antonio Laz-Ruiz
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Gov-ernment, PTS Granada, Avda. Ilustración 114, 18016 Granada, Spain.,Department of Medicinal & Organic Chemistry and Excellence Research Unit of "Chemistry applied to Biomedi-cine and the Environment", Faculty of Pharmacy, University of Granada, Campus de Cartuja s/n, 18071 Granada, Spain.,Biosanitary Research Institute of Granada (ibs.GRANADA), University Hospitals of Granada-University of Grana-da, 18012 Granada, Spain
| | - M Victoria Cano-Cortes
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Gov-ernment, PTS Granada, Avda. Ilustración 114, 18016 Granada, Spain.,Department of Medicinal & Organic Chemistry and Excellence Research Unit of "Chemistry applied to Biomedi-cine and the Environment", Faculty of Pharmacy, University of Granada, Campus de Cartuja s/n, 18071 Granada, Spain.,Biosanitary Research Institute of Granada (ibs.GRANADA), University Hospitals of Granada-University of Grana-da, 18012 Granada, Spain
| | - Ying-Wen Huang
- Department of Urology, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Veronica D Gonzalez
- Department of Urology, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Juan Jose Diaz-Mochon
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Gov-ernment, PTS Granada, Avda. Ilustración 114, 18016 Granada, Spain.,Department of Medicinal & Organic Chemistry and Excellence Research Unit of "Chemistry applied to Biomedi-cine and the Environment", Faculty of Pharmacy, University of Granada, Campus de Cartuja s/n, 18071 Granada, Spain.,Biosanitary Research Institute of Granada (ibs.GRANADA), University Hospitals of Granada-University of Grana-da, 18012 Granada, Spain
| | - Wendy J Fantl
- Department of Urology, Stanford University School of Medicine, Stanford, California 94305, United States.,Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California 94305, United States.,Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, California 94304, United States
| | - Rosario M Sanchez-Martin
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Gov-ernment, PTS Granada, Avda. Ilustración 114, 18016 Granada, Spain.,Department of Medicinal & Organic Chemistry and Excellence Research Unit of "Chemistry applied to Biomedi-cine and the Environment", Faculty of Pharmacy, University of Granada, Campus de Cartuja s/n, 18071 Granada, Spain.,Biosanitary Research Institute of Granada (ibs.GRANADA), University Hospitals of Granada-University of Grana-da, 18012 Granada, Spain
| |
Collapse
|
28
|
Mohr E, Thum T, Bär C. Accelerating Cardiovascular Research: Recent Advances in Translational 2D and 3D Heart Models. Eur J Heart Fail 2022; 24:1778-1791. [PMID: 35867781 DOI: 10.1002/ejhf.2631] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 06/30/2022] [Accepted: 07/20/2022] [Indexed: 11/11/2022] Open
Abstract
In vitro modelling the complex (patho-) physiological conditions of the heart is a major challenge in cardiovascular research. In recent years, methods based on three-dimensional (3D) cultivation approaches have steadily evolved to overcome the major limitations of conventional adherent monolayer cultivation (2D). These 3D approaches aim to study, reproduce or modify fundamental native features of the heart such as tissue organization and cardiovascular microenvironment. Therefore, these systems have great potential for (patient-specific) disease research, for the development of new drug screening platforms, and for the use in regenerative and replacement therapy applications. Consequently, continuous improvement and adaptation is required with respect to fundamental limitations such as cardiomyocyte maturation, scalability, heterogeneity, vascularization, and reproduction of native properties. In this review, 2D monolayer culturing and the 3D in vitro systems of cardiac spheroids, organoids, engineered cardiac microtissue and bioprinting as well as the ex vivo technique of myocardial slicing are introduced with their basic concepts, advantages, and limitations. Furthermore, recent advances of various new approaches aiming to extend as well as to optimize these in vitro and ex vivo systems are presented. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Elisa Mohr
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Str.1, 30625, Hannover, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Str.1, 30625, Hannover, Germany.,REBIRTH Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany.,Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hannover, Germany
| | - Christian Bär
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Str.1, 30625, Hannover, Germany.,REBIRTH Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany.,Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hannover, Germany
| |
Collapse
|
29
|
Wu W, Li X, Yu S. Patient-derived Tumour Organoids: A Bridge between Cancer Biology and Personalised Therapy. Acta Biomater 2022; 146:23-36. [PMID: 35533925 DOI: 10.1016/j.actbio.2022.04.050] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/28/2022] [Accepted: 04/29/2022] [Indexed: 02/06/2023]
Abstract
Patient-derived tumour organoids (PDOs) have revolutionised our understanding of cancer biology and the applications of personalised therapies. These advancements are principally ascribed to the ability of PDOs to consistently recapitulate and maintain the genomic, proteomic and morphological characteristics of parental tumours. Given these characteristics, PDOs (and their extended biobanks) are a representative preclinical model eminently suited to translate relevant scientific findings into personalized therapies rapidly. Here, we summarise recent advancements in PDOs from the perspective of cancer biology and clinical applications, focusing on the current challenges and opportunities of reconstructing and standardising more sophisticated PDO models. STATEMENT OF SIGNIFICANCE: Patient-derived tumour organoids (PDOs), three-dimensional (3D) self-assembled organotypic structures, have revolutionised our understanding of cancer biology and the applications of personalised therapies. These advancements are principally ascribed to the ability of PDOs to consistently recapitulate and maintain the genomic, proteomic and morphological characteristics of parental tumours. Given these characteristics, PDOs (and their extended biobanks) are a representative preclinical model eminently suited to translate relevant scientific findings into personalized therapies rapidly.
Collapse
Affiliation(s)
- Wence Wu
- Departments of Orthopedics, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Nanli, Panjiayuan, Chaoyang District, Beijing, 100021, China
| | - Xiaoyang Li
- Departments of Orthopedics, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Nanli, Panjiayuan, Chaoyang District, Beijing, 100021, China
| | - Shengji Yu
- Departments of Orthopedics, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Nanli, Panjiayuan, Chaoyang District, Beijing, 100021, China.
| |
Collapse
|
30
|
Wang K, Yu Y, Han R, Wang X, Zhao Y, Tang H, Li G. [Establishment of a culture system for human nasal mucosa organoids with controllable differentiation]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2022; 42:868-877. [PMID: 35790437 DOI: 10.12122/j.issn.1673-4254.2022.06.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To establish a culture system for human nasal mucosal organoids with controllable differentiation to reproduce the structure and function of the source tissue through staged expansion-differentiation culture. METHODS Fresh samples of surgically resected middle turbinate and nasal polyp tissues were collected, from which the nasal mucosa epithelial cells were isolated by enzymatic digestion and filtration for continuous culture at the air-liquid interface for expansion (EO group) or staged culture for expansion and differentiation (DO group). Immunohistochemical staining was used to characterize the structure, cellular composition and ciliary function of nasal mucosal organoids in the two groups. The secretion function of the differentiated nasal mucosal organoids in DO group was evaluated using PAS staining. RESULTS Both of the two organoid culture systems yielded vacuolar or solid spherical 3D organoids, and their diameters increased progressively with time. On day 16 of culture, more vacuolar organoids occurred in DO group, while more solid spherical organoids were seen in EO group, and the proportion of vacuoles was significantly greater in DO group than in EO group [(54.67±13.26)% vs (21.67±8.57)%, P < 0.05]. Short tandem repeat (STR) test of the nasal mucosal organoids and the source tissue showed a 100% match between them. On day 21 of culture, scanning and transmission electron microscopy of the nasal mucosal organoids identified ultrastructure of cilia in DO group and short villi structure in most of the organoids in EO group. Immunohistochemical staining showed positivity for P63 (basal cells), β-tubulin (ciliated columnar cells), and MUC5AC (goblet cells) in the organoids. Compared with those in EO group, the organoids in DO group showed significantly greater percentages of ciliated cells [(7.95±1.81)% vs (27.04±5.91)%, P < 0.05] and goblet cells [(14.46±0.93)% vs (39.85±5.43)%, P < 0.05) with a similar percentage of basal cells [(56.91±14.12)% vs (53.42±15.77)%, P > 0.05]. The differentiated nasal mucosal organoids in DO group were positively stained for glycogen. CONCLUSION The staged expansion-differentiation culture method allows more stable and prolonged growth of the cultured cells in vitro to produce organoids with controllable differentiation closely resembling the morphological structure and functions (ciliary function and secretory function) of the source tissue.
Collapse
Affiliation(s)
- K Wang
- Department of Otolaryngology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Y Yu
- Department of Otolaryngology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - R Han
- Department of Otolaryngology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - X Wang
- Department of Otolaryngology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Y Zhao
- Department of Otolaryngology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - H Tang
- Department of Otolaryngology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - G Li
- Department of Otolaryngology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
31
|
Hautefort I, Poletti M, Papp D, Korcsmaros T. Everything You Always Wanted to Know About Organoid-Based Models (and Never Dared to Ask). Cell Mol Gastroenterol Hepatol 2022; 14:311-331. [PMID: 35643188 PMCID: PMC9233279 DOI: 10.1016/j.jcmgh.2022.04.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 04/28/2022] [Accepted: 04/29/2022] [Indexed: 12/12/2022]
Abstract
Homeostatic functions of a living tissue, such as the gastrointestinal tract, rely on highly sophisticated and finely tuned cell-to-cell interactions. These crosstalks evolve and continuously are refined as the tissue develops and give rise to specialized cells performing general and tissue-specific functions. To study these systems, stem cell-based in vitro models, often called organoids, and non-stem cell-based primary cell aggregates (called spheroids) appeared just over a decade ago. These models still are evolving and gaining complexity, making them the state-of-the-art models for studying cellular crosstalk in the gastrointestinal tract, and to investigate digestive pathologies, such as inflammatory bowel disease, colorectal cancer, and liver diseases. However, the use of organoid- or spheroid-based models to recapitulate in vitro the highly complex structure of in vivo tissue remains challenging, and mainly restricted to expert developmental cell biologists. Here, we condense the founding knowledge and key literature information that scientists adopting the organoid technology for the first time need to consider when using these models for novel biological questions. We also include information that current organoid/spheroid users could use to add to increase the complexity to their existing models. We highlight the current and prospective evolution of these models through bridging stem cell biology with biomaterial and scaffold engineering research areas. Linking these complementary fields will increase the in vitro mimicry of in vivo tissue, and potentially lead to more successful translational biomedical applications. Deepening our understanding of the nature and dynamic fine-tuning of intercellular crosstalks will enable identifying novel signaling targets for new or repurposed therapeutics used in many multifactorial diseases.
Collapse
Affiliation(s)
- Isabelle Hautefort
- Earlham Institute, Organisms and Ecosystems Programme, Norwich, United Kingdom
| | - Martina Poletti
- Earlham Institute, Organisms and Ecosystems Programme, Norwich, United Kingdom; Quadram Institute Bioscience, Gut Microbes and Health Programme, Norwich, United Kingdom
| | - Diana Papp
- Quadram Institute Bioscience, Gut Microbes and Health Programme, Norwich, United Kingdom
| | - Tamas Korcsmaros
- Earlham Institute, Organisms and Ecosystems Programme, Norwich, United Kingdom; Quadram Institute Bioscience, Gut Microbes and Health Programme, Norwich, United Kingdom; Imperial College London, Department of Metabolism, Digestion and Reproduction, London, United Kingdom.
| |
Collapse
|
32
|
Baskar R, Kimmey SC, Bendall SC. Revealing new biology from multiplexed, metal-isotope-tagged, single-cell readouts. Trends Cell Biol 2022; 32:501-512. [PMID: 35181197 DOI: 10.1016/j.tcb.2022.01.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 11/26/2022]
Abstract
Mass cytometry (MC) is a recent technology that pairs plasma-based ionization of cells in suspension with time-of-flight (TOF) mass spectrometry to sensitively quantify the single-cell abundance of metal-isotope-tagged affinity reagents to key proteins, RNA, and peptides. Given the ability to multiplex readouts (~50 per cell) and capture millions of cells per experiment, MC offers a robust way to assay rare, transitional cell states that are pertinent to human development and disease. Here, we review MC approaches that let us probe the dynamics of cellular regulation across multiple conditions and sample types in a single experiment. Additionally, we discuss current limitations and future extensions of MC as well as computational tools commonly used to extract biological insight from single-cell proteomic datasets.
Collapse
Affiliation(s)
- Reema Baskar
- Department of Pathology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Sam C Kimmey
- Department of Pathology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Sean C Bendall
- Department of Pathology, School of Medicine, Stanford University, Stanford, CA, USA.
| |
Collapse
|
33
|
Wang J, Feng X, Li Z, Chen Y, Huang W. Patient-derived organoids as a model for tumor research. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 189:259-326. [PMID: 35595351 DOI: 10.1016/bs.pmbts.2022.03.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Cancer represents a leading cause of death, despite the rapid progress of cancer research, leading to urgent need for accurate preclinical model to further study of tumor mechanism and accelerate translational applications. Cancer cell lines cannot fully recapitulate tumors of different patients due to the lack of tumor complexity and specification, while the high technical difficulty, long time, and substantial cost of patient-derived xenograft model makes it unable to be used extensively for all types of tumors and large-scale drug screening. Patient-derived organoids can be established rapidly with a high success rate from many tumors, and precisely replicate the key histopathological, genetic, and phenotypic features, as well as therapeutic response of patient tumor. Therefore, they are extensively used in cancer basic research, biobanking, disease modeling and precision medicine. The combinations of cancer organoids with other advanced technologies, such as 3D bio-printing, organ-on-a-chip, and CRISPR-Cas9, contributes to the more complete replication of complex tumor microenvironment and tumorigenesis. In this review, we discuss the various methods of the establishment and the application of patient-derived organoids in diverse tumors as well as the limitations and future prospects of these models. Further advances of tumor organoids are expected to bridge the huge gap between bench and bedside and provide the unprecedented opportunities to advance cancer research.
Collapse
Affiliation(s)
- Jia Wang
- The First Affiliated Hospital of Shantou University, Shantou University Medical College, Shantou, China
| | - Xiaoying Feng
- The First Affiliated Hospital of Shantou University, Shantou University Medical College, Shantou, China
| | - Zhichao Li
- Department of Urology, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China; Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen, China; International Cancer Center of Shenzhen University, Shenzhen, China
| | - Yongsong Chen
- The First Affiliated Hospital of Shantou University, Shantou University Medical College, Shantou, China
| | - Weiren Huang
- The First Affiliated Hospital of Shantou University, Shantou University Medical College, Shantou, China; Department of Urology, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China; Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen, China; International Cancer Center of Shenzhen University, Shenzhen, China; Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
| |
Collapse
|
34
|
Below CR, Kelly J, Brown A, Humphries JD, Hutton C, Xu J, Lee BY, Cintas C, Zhang X, Hernandez-Gordillo V, Stockdale L, Goldsworthy MA, Geraghty J, Foster L, O'Reilly DA, Schedding B, Askari J, Burns J, Hodson N, Smith DL, Lally C, Ashton G, Knight D, Mironov A, Banyard A, Eble JA, Morton JP, Humphries MJ, Griffith LG, Jørgensen C. A microenvironment-inspired synthetic three-dimensional model for pancreatic ductal adenocarcinoma organoids. NATURE MATERIALS 2022; 21:110-119. [PMID: 34518665 PMCID: PMC7612137 DOI: 10.1038/s41563-021-01085-1] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 07/21/2021] [Indexed: 05/09/2023]
Abstract
Experimental in vitro models that capture pathophysiological characteristics of human tumours are essential for basic and translational cancer biology. Here, we describe a fully synthetic hydrogel extracellular matrix designed to elicit key phenotypic traits of the pancreatic environment in culture. To enable the growth of normal and cancerous pancreatic organoids from genetically engineered murine models and human patients, essential adhesive cues were empirically defined and replicated in the hydrogel scaffold, revealing a functional role of laminin-integrin α3/α6 signalling in establishment and survival of pancreatic organoids. Altered tissue stiffness-a hallmark of pancreatic cancer-was recapitulated in culture by adjusting the hydrogel properties to engage mechano-sensing pathways and alter organoid growth. Pancreatic stromal cells were readily incorporated into the hydrogels and replicated phenotypic traits characteristic of the tumour environment in vivo. This model therefore recapitulates a pathologically remodelled tumour microenvironment for studies of normal and pancreatic cancer cells in vitro.
Collapse
Affiliation(s)
- Christopher R Below
- Cancer Research UK Manchester Institute, The University of Manchester, Manchester, UK
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Joanna Kelly
- Cancer Research UK Manchester Institute, The University of Manchester, Manchester, UK
| | - Alexander Brown
- Centre for Gynepathology Research, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jonathan D Humphries
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Department of Life Science, Manchester Metropolitan University, Manchester, UK
| | - Colin Hutton
- Cancer Research UK Manchester Institute, The University of Manchester, Manchester, UK
| | - Jingshu Xu
- Cancer Research UK Manchester Institute, The University of Manchester, Manchester, UK
| | - Brian Y Lee
- Cancer Research UK Manchester Institute, The University of Manchester, Manchester, UK
| | - Celia Cintas
- Cancer Research UK Manchester Institute, The University of Manchester, Manchester, UK
| | - Xiaohong Zhang
- Cancer Research UK Manchester Institute, The University of Manchester, Manchester, UK
| | - Victor Hernandez-Gordillo
- Centre for Gynepathology Research, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Linda Stockdale
- Centre for Gynepathology Research, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | | | | | | | - Barbara Schedding
- Institute for Physiological Chemistry and Pathobiochemistry, University of Muenster, Muenster, Germany
| | - Janet Askari
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Jessica Burns
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Nigel Hodson
- BioAFM Laboratory, Bioimaging Core Facility, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Duncan L Smith
- Cancer Research UK Manchester Institute, The University of Manchester, Manchester, UK
| | - Catherine Lally
- Cancer Research UK Manchester Institute, The University of Manchester, Manchester, UK
| | - Garry Ashton
- Cancer Research UK Manchester Institute, The University of Manchester, Manchester, UK
| | - David Knight
- Biological Mass Spectrometry Core Facility, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Aleksandr Mironov
- Electron Microscopy Core Facility, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Antonia Banyard
- Cancer Research UK Manchester Institute, The University of Manchester, Manchester, UK
| | - Johannes A Eble
- Institute for Physiological Chemistry and Pathobiochemistry, University of Muenster, Muenster, Germany
| | - Jennifer P Morton
- Cancer Research UK Beatson Institute, Glasgow, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Martin J Humphries
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Linda G Griffith
- Centre for Gynepathology Research, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Claus Jørgensen
- Cancer Research UK Manchester Institute, The University of Manchester, Manchester, UK.
| |
Collapse
|
35
|
Flores-Téllez TDNJ, Baena E. Experimental challenges to modeling prostate cancer heterogeneity. Cancer Lett 2022; 524:194-205. [PMID: 34688843 DOI: 10.1016/j.canlet.2021.10.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 09/23/2021] [Accepted: 10/09/2021] [Indexed: 12/24/2022]
Abstract
Tumor heterogeneity plays a key role in prostate cancer prognosis, therapy selection, relapse, and acquisition of treatment resistance. Prostate cancer presents a heterogeneous diversity at inter- and intra-tumor and inter-patient levels which are influenced by multiple intrinsic and/or extrinsic factors. Recent studies have started to characterize the complexity of prostate tumors and these different tiers of heterogeneity. In this review, we discuss the most common factors that contribute to tumoral diversity. Moreover, we focus on the description of the in vitro and in vivo approaches, as well as high-throughput technologies, that help to model intra-tumoral diversity. Further understanding tumor heterogeneities and the challenges they present will guide enhanced patient risk stratification, aid the design of more precise therapies, and ultimately help beat this chameleon-like disease.
Collapse
Affiliation(s)
- Teresita Del N J Flores-Téllez
- Prostate Oncobiology Group, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, Alderley Edge, Macclesfield, SK10 4TG, UK
| | - Esther Baena
- Prostate Oncobiology Group, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, Alderley Edge, Macclesfield, SK10 4TG, UK; Belfast-Manchester Movember Centre of Excellence, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, SK10 4TG, UK.
| |
Collapse
|
36
|
Shen Z, Zhao H, Yao H, Pan X, Yang J, Zhang S, Han G, Zhang X. Dynamic metabolic change of cancer cells induced by natural killer cells at single-cell level studied by label-free mass cytometry. Chem Sci 2022; 13:1641-1647. [PMID: 35282636 PMCID: PMC8827047 DOI: 10.1039/d1sc06366a] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 12/31/2021] [Indexed: 01/10/2023] Open
Abstract
Natural killer cells (NK cells) are important immune cells which have attracted increasing attention in cancer immunotherapy. Due to the heterogeneity of cells, individual cancer cells show different resistance to NK cytotoxicity, which has been revealed by flow cytometry. Here we used label-free mass cytometry (CyESI-MS) as a new tool to analyze the metabolites in Human Hepatocellular Carcinoma (HepG2) cells at the single-cell level after the interaction with different numbers of NK92 MI cells. A large amount of chemical information from individual HepG2 cells was obtained showing the process of cell apoptosis induced by NK cells. Nineteen metabolites which consecutively change during cell apoptosis were revealed by calculating their average relative intensity. Four metabolic pathways were impacted during cell apoptosis which hit 4 metabolites including glutathione (GSH), creatine, glutamic acid and taurine. We found that the HepG2 cells could be divided into two phenotypes after co-culturing with NK cells according to the bimodal distribution of concentration of these 4 metabolites. The correlation between metabolites and different apoptotic pathways in the early apoptosis cell group was established by the 4 metabolites at the single-cell level. This is a new idea of using single-cell specific metabolites to reveal the metabolic heterogeneity in cell apoptosis which would be a powerful means for evaluating the cytotoxicity of NK cells. Label-free mass cytometry is utilized to study the dynamic metabolic change during apoptosis in HepG2 cells induced by NK92 MI cells at the single-cell level. The metabolic heterogeneity of individual HepG2 cells during apoptosis was revealed.![]()
Collapse
Affiliation(s)
- Zizheng Shen
- Department of Chemistry, Tsinghua University Beijing 100084 China
| | - Hansen Zhao
- Department of Chemistry, Tsinghua University Beijing 100084 China
| | - Huan Yao
- Department of Chemistry, Tsinghua University Beijing 100084 China
| | - Xingyu Pan
- Department of Chemistry, Tsinghua University Beijing 100084 China
| | - Jinlei Yang
- Department of Chemistry, Tsinghua University Beijing 100084 China
| | - Sichun Zhang
- Department of Chemistry, Tsinghua University Beijing 100084 China
| | - Guojun Han
- Institute of Medical Technology, Peking University Health Science Center Beijing 100191 China
- Peking University School and Hospital of Stomatology Beijing 100081 P. R. China
- Department of Biomedical Engineering, Peking University Health Science Center Beijing 100191 P. R. China
| | - Xinrong Zhang
- Department of Chemistry, Tsinghua University Beijing 100084 China
| |
Collapse
|
37
|
Melliou S, Sangster KT, Djuric U, Diamandis P. The promise of organoids for unraveling the proteomic landscape of the developing human brain. Mol Psychiatry 2022; 27:73-80. [PMID: 34703024 DOI: 10.1038/s41380-021-01354-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 10/04/2021] [Accepted: 10/05/2021] [Indexed: 12/13/2022]
Abstract
Cerebral organoids offer an opportunity to bioengineer experimental avatars of the developing human brain and have already begun garnering relevant insights into complex neurobiological processes and disease. Thus far, investigations into their heterogeneous cellular composition and developmental trajectories have been largely limited to transcriptional readouts. Recent advances in global proteomic technologies have enabled a new range of techniques to explore dynamic and non-overlapping spatiotemporal protein-level programs operational in these humanoid neural structures. Here we discuss these early protein-based studies and their potentially essential role for unraveling critical secreted paracrine signals, processes with poor proteogenomic correlations, or neurodevelopmental proteins requiring post-translational modification for biological activity. Integrating emerging proteomic tools with these faithful human-derived neurodevelopmental models could transform our understanding of complex neural cell phenotypes and neurobiological processes, not exclusively driven by transcriptional regulation. These insights, less accessible by exclusive RNA-based approaches, could reveal new knowledge into human brain development and guide improvements in neural regenerative medicine efforts.
Collapse
Affiliation(s)
- Sofia Melliou
- Princess Margaret Cancer Centre, 101 College Street, Toronto, ON, M5G 1L7, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Kevin T Sangster
- Princess Margaret Cancer Centre, 101 College Street, Toronto, ON, M5G 1L7, Canada
| | - Ugljesa Djuric
- Princess Margaret Cancer Centre, 101 College Street, Toronto, ON, M5G 1L7, Canada
| | - Phedias Diamandis
- Princess Margaret Cancer Centre, 101 College Street, Toronto, ON, M5G 1L7, Canada. .,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada. .,Laboratory Medicine Program, University Health Network, 200 Elizabeth Street, Toronto, ON, M5G 2C4, Canada. .,Department of Medical Biophysics, University of Toronto, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
38
|
Tristan CA, Ormanoglu P, Slamecka J, Malley C, Chu PH, Jovanovic VM, Gedik Y, Jethmalani Y, Bonney C, Barnaeva E, Braisted J, Mallanna SK, Dorjsuren D, Iannotti MJ, Voss TC, Michael S, Simeonov A, Singeç I. Robotic high-throughput biomanufacturing and functional differentiation of human pluripotent stem cells. Stem Cell Reports 2021; 16:3076-3092. [PMID: 34861164 PMCID: PMC8693769 DOI: 10.1016/j.stemcr.2021.11.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 11/02/2021] [Accepted: 11/04/2021] [Indexed: 12/21/2022] Open
Abstract
Efficient translation of human induced pluripotent stem cells (hiPSCs) requires scalable cell manufacturing strategies for optimal self-renewal and functional differentiation. Traditional manual cell culture is variable and labor intensive, posing challenges for high-throughput applications. Here, we established a robotic platform and automated all essential steps of hiPSC culture and differentiation under chemically defined conditions. This approach allowed rapid and standardized manufacturing of billions of hiPSCs that can be produced in parallel from up to 90 different patient- and disease-specific cell lines. Moreover, we established automated multi-lineage differentiation and generated functional neurons, cardiomyocytes, and hepatocytes. To validate our approach, we compared robotic and manual cell culture operations and performed comprehensive molecular and cellular characterizations (e.g., single-cell transcriptomics, mass cytometry, metabolism, electrophysiology) to benchmark industrial-scale cell culture operations toward building an integrated platform for efficient cell manufacturing for disease modeling, drug screening, and cell therapy.
Collapse
Affiliation(s)
- Carlos A Tristan
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation (DPI), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Pinar Ormanoglu
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation (DPI), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Jaroslav Slamecka
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation (DPI), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Claire Malley
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation (DPI), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Pei-Hsuan Chu
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation (DPI), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Vukasin M Jovanovic
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation (DPI), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Yeliz Gedik
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation (DPI), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Yogita Jethmalani
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation (DPI), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Charles Bonney
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation (DPI), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Elena Barnaeva
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation (DPI), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - John Braisted
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation (DPI), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Sunil K Mallanna
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation (DPI), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Dorjbal Dorjsuren
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation (DPI), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Michael J Iannotti
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation (DPI), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Ty C Voss
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation (DPI), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Sam Michael
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation (DPI), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Anton Simeonov
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation (DPI), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Ilyas Singeç
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation (DPI), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), 9800 Medical Center Drive, Rockville, MD 20850, USA.
| |
Collapse
|
39
|
Fostering experimental and computational synergy to modulate hyperinflammation. Trends Immunol 2021; 43:4-7. [PMID: 34844849 PMCID: PMC9375712 DOI: 10.1016/j.it.2021.11.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 11/03/2021] [Accepted: 11/04/2021] [Indexed: 11/23/2022]
Abstract
The molecular underpinnings of the uncontrolled release of proinflammatory cytokines and chemokines (‘cytokine storm’), which can cause organ damage and even mortality, are not completely understood. Furthermore, targeted therapeutic options to dampen such hyperinflammation are scarce. Here, we highlight the ways in which technological advances have set the stage for a new age of synergy between experimental and computational researchers to guide the discovery of novel therapeutic targets for modulating hyperinflammation.
Collapse
|
40
|
Qu J, Kalyani FS, Liu L, Cheng T, Chen L. Tumor organoids: synergistic applications, current challenges, and future prospects in cancer therapy. Cancer Commun (Lond) 2021; 41:1331-1353. [PMID: 34713636 PMCID: PMC8696219 DOI: 10.1002/cac2.12224] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/29/2021] [Accepted: 09/17/2021] [Indexed: 02/06/2023] Open
Abstract
Patient-derived cancer cells (PDCs) and patient-derived xenografts (PDXs) are often used as tumor models, but have many shortcomings. PDCs not only lack diversity in terms of cell type, spatial organization, and microenvironment but also have adverse effects in stem cell cultures, whereas PDX are expensive with a low transplantation success rate and require a long culture time. In recent years, advances in three-dimensional (3D) organoid culture technology have led to the development of novel physiological systems that model the tissues of origin more precisely than traditional culture methods. Patient-derived cancer organoids bridge the conventional gaps in PDC and PDX models and closely reflect the pathophysiological features of natural tumorigenesis and metastasis, and have led to new patient-specific drug screening techniques, development of individualized treatment regimens, and discovery of prognostic biomarkers and mechanisms of resistance. Synergistic combinations of cancer organoids with other technologies, for example, organ-on-a-chip, 3D bio-printing, and CRISPR-Cas9-mediated homology-independent organoid transgenesis, and with treatments, such as immunotherapy, have been useful in overcoming their limitations and led to the development of more suitable model systems that recapitulate the complex stroma of cancer, inter-organ and intra-organ communications, and potentially multiorgan metastasis. In this review, we discuss various methods for the creation of organ-specific cancer organoids and summarize organ-specific advances and applications, synergistic technologies, and treatments as well as current limitations and future prospects for cancer organoids. Further advances will bring this novel 3D organoid culture technique closer to clinical practice in the future.
Collapse
Affiliation(s)
- Jingjing Qu
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, P. R. China.,Lung Cancer and Gastroenterology Department, Hunan Cancer Hospital, Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, Hunan, 410008, P. R. China
| | - Farhin Shaheed Kalyani
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, P. R. China
| | - Li Liu
- Lung Cancer and Gastroenterology Department, Hunan Cancer Hospital, Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, Hunan, 410008, P. R. China
| | - Tianli Cheng
- Thoracic Medicine Department 1, Hunan Cancer Hospital, Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, Hunan, 410008, P. R. China
| | - Lijun Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, P. R. China
| |
Collapse
|
41
|
Sufi J, Qin X, Rodriguez FC, Bu YJ, Vlckova P, Zapatero MR, Nitz M, Tape CJ. Multiplexed single-cell analysis of organoid signaling networks. Nat Protoc 2021; 16:4897-4918. [PMID: 34497385 DOI: 10.1038/s41596-021-00603-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 07/06/2021] [Indexed: 02/08/2023]
Abstract
Organoids are biomimetic tissue models comprising multiple cell types and cell states. Post-translational modification (PTM) signaling networks control cellular phenotypes and are frequently dysregulated in diseases such as cancer. Although signaling networks vary across cell types, there are limited techniques to study cell type-specific PTMs in heterocellular organoids. Here, we present a multiplexed mass cytometry (MC) protocol for single-cell analysis of PTM signaling and cell states in organoids and organoids co-cultured with fibroblasts and leukocytes. We describe how thiol-reactive organoid barcoding in situ (TOBis) enables 35-plex and 126-plex single-cell comparison of organoid cultures and provide a cytometry by time of flight (CyTOF) signaling analysis pipeline (CyGNAL) for computing cell type-specific PTM signaling networks. The TOBis MC protocol takes ~3 d from organoid fixation to data acquisition and can generate single-cell data for >40 antibodies from millions of cells across 126 organoid cultures in a single MC run.
Collapse
Affiliation(s)
- Jahangir Sufi
- Cell Communication Lab, Department of Oncology, University College London Cancer Institute, London, UK
| | - Xiao Qin
- Cell Communication Lab, Department of Oncology, University College London Cancer Institute, London, UK
| | - Ferran Cardoso Rodriguez
- Cell Communication Lab, Department of Oncology, University College London Cancer Institute, London, UK
| | - Yong Jia Bu
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Petra Vlckova
- Cell Communication Lab, Department of Oncology, University College London Cancer Institute, London, UK
| | - María Ramos Zapatero
- Cell Communication Lab, Department of Oncology, University College London Cancer Institute, London, UK
| | - Mark Nitz
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Christopher J Tape
- Cell Communication Lab, Department of Oncology, University College London Cancer Institute, London, UK.
| |
Collapse
|
42
|
Abstract
Organoids are self-organizing, expanding 3D cultures derived from stem cells. Using tissue derived from patients, these miniaturized models recapitulate various aspects of patient physiology and disease phenotypes including genetic profiles and drug sensitivities. As such, patient-derived organoid (PDO) platforms provide an unprecedented opportunity for improving preclinical drug discovery, clinical trial validation, and ultimately patient care. This article reviews the evolution and scope of organoid technology, highlights recent encouraging results using PDOs as potential patient "avatars" to predict drug response and outcomes, and discusses critical parameters for widespread clinical adoption. These include improvements in assay speed, reproducibility, standardization, and automation which are necessary to realize the translational potential of PDOs as clinical tools. The multiple entry points where PDOs may contribute valuable insights in drug discovery and lessen the risks associated with clinical trials are also discussed.
Collapse
Affiliation(s)
- Shree Bose
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, North Carolina 27705, USA
| | - Hans Clevers
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, the Netherlands
- Oncode, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center (UMC) Utrecht, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands
| | - Xiling Shen
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, North Carolina 27705, USA
| |
Collapse
|
43
|
Qin X, Tape CJ. Deciphering Organoids: High-Dimensional Analysis of Biomimetic Cultures. Trends Biotechnol 2021; 39:774-787. [PMID: 33279281 DOI: 10.1016/j.tibtech.2020.10.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 10/21/2020] [Accepted: 10/29/2020] [Indexed: 12/29/2022]
Abstract
Organoids are self-organising stem cell-derived ex vivo cultures widely adopted as biomimetic models of healthy and diseased tissues. Traditional low-dimensional experimental methods such as microscopy and bulk molecular analysis have generated remarkable biological insights from organoids. However, as complex heterocellular systems, organoids are especially well-positioned to take advantage of emerging high-dimensional technologies. In particular, single-cell methods offer considerable opportunities to analyse organoids at unprecedented scale and depth, enabling comprehensive characterisation of cellular processes and spatial organisation underpinning organoid heterogeneity. This review evaluates state-of-the-art analytical methods applied to organoids, discusses the latest advances in single-cell technologies, and speculates on the integration of these two rapidly developing fields.
Collapse
Affiliation(s)
- Xiao Qin
- Cell Communication Lab, Department of Oncology, University College London Cancer Institute, London, UK
| | - Christopher J Tape
- Cell Communication Lab, Department of Oncology, University College London Cancer Institute, London, UK.
| |
Collapse
|
44
|
Carter EP, Roozitalab R, Gibson SV, Grose RP. Tumour microenvironment 3D-modelling: simplicity to complexity and back again. Trends Cancer 2021; 7:1033-1046. [PMID: 34312120 DOI: 10.1016/j.trecan.2021.06.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 06/16/2021] [Accepted: 06/28/2021] [Indexed: 02/07/2023]
Abstract
Tumours are surrounded by a host of noncancerous cells that fulfil both supportive and suppressive roles within the tumour microenvironment (TME). The drive to understand the biology behind each of these components has led to a rapid expansion in the number and use of 3D in vitro models, as researchers find ways to incorporate multiple cell types into physiomimetic configurations. The use and increasing complexity of these models does however demand many considerations. In this review we discuss approaches adopted to recapitulate complex tumour biology in tractable 3D models. We consider how these cell types can be sourced and combined and examine methods for the deconvolution of complex multicellular models into manageable and informative outputs.
Collapse
Affiliation(s)
- Edward P Carter
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Reza Roozitalab
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Shayin V Gibson
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Richard P Grose
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK.
| |
Collapse
|
45
|
Roe CE, Hayes MJ, Barone SM, Irish JM. Training Novices in Generation and Analysis of High-Dimensional Human Cell Phospho-Flow Cytometry Data. ACTA ACUST UNITED AC 2021; 93:e71. [PMID: 32250555 DOI: 10.1002/cpcy.71] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
This article presents a single experiment designed to introduce a trainee to multiple advanced bench and analysis techniques, including high-dimensional cytometry, profiling cell signaling networks, functional assays with primary human tissue, and single-cell analysis with machine learning tools. The trainee is expected to have only minimal laboratory experience and is not required to have any prior training in flow cytometry, immunology, or data science. This article aims to introduce the advanced research areas with a design that is robust enough that novice trainees will succeed, flexible enough to allow some project customization, and fundamental enough that the skills and knowledge gained will provide a template for future experiments. For advanced users, the updated phospho-flow protocol and the established controls, best practices, and expected outcomes presented here also provide a framework for adapting these tools in new areas with unexplored biology. © 2020 by John Wiley & Sons, Inc. Basic Protocol: Phospho-protein stimulation and mass cytometry data collection Support Protocol: Analysis of signaling mass cytometry data.
Collapse
Affiliation(s)
- Caroline E Roe
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee.,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Madeline J Hayes
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee.,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Sierra M Barone
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee.,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jonathan M Irish
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee.,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
46
|
Maia A, Wiemann S. Cancer-Associated Fibroblasts: Implications for Cancer Therapy. Cancers (Basel) 2021; 13:3526. [PMID: 34298736 PMCID: PMC8307167 DOI: 10.3390/cancers13143526] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/09/2021] [Accepted: 07/12/2021] [Indexed: 12/12/2022] Open
Abstract
Tumour cells do not exist as an isolated entity. Instead, they are surrounded by and closely interact with cells of the environment they are emerged in. The tumour microenvironment (TME) is not static and several factors, including cancer cells and therapies, have been described to modulate several of its components. Fibroblasts are key elements of the TME with the capacity to influence tumour progression, invasion and response to therapy, which makes them attractive targets in cancer treatment. In this review, we focus on fibroblasts and their numerous roles in the TME with a special attention to recent findings describing their heterogeneity and role in therapy response. Furthermore, we explore how different therapies can impact these cells and their communication with cancer cells. Finally, we highlight potential strategies targeting this cell type that can be employed for improving patient outcome.
Collapse
Affiliation(s)
- Ana Maia
- German Cancer Research Center (DKFZ), Division of Molecular Genome Analysis, Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| | - Stefan Wiemann
- German Cancer Research Center (DKFZ), Division of Molecular Genome Analysis, Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| |
Collapse
|
47
|
Integrated multi-omics analyses on patient-derived CRC organoids highlight altered molecular pathways in colorectal cancer progression involving PTEN. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:198. [PMID: 34154611 PMCID: PMC8215814 DOI: 10.1186/s13046-021-01986-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 05/16/2021] [Indexed: 12/23/2022]
Abstract
Background Colorectal cancer (CRC) represents the fourth leading cause of cancer-related deaths. The heterogeneity of CRC identity limits the usage of cell lines to study this type of tumor because of the limited representation of multiple features of the original malignancy. Patient-derived colon organoids (PDCOs) are a promising 3D-cell model to study tumor identity for personalized medicine, although this approach still lacks detailed characterization regarding molecular stability during culturing conditions. Correlation analysis that considers genomic, transcriptomic, and proteomic data, as well as thawing, timing, and culturing conditions, is missing. Methods Through integrated multi–omics strategies, we characterized PDCOs under different growing and timing conditions, to define their ability to recapitulate the original tumor. Results Whole Exome Sequencing allowed detecting temporal acquisition of somatic variants, in a patient-specific manner, having deleterious effects on driver genes CRC-associated. Moreover, the targeted NGS approach confirmed that organoids faithfully recapitulated patients’ tumor tissue. Using RNA-seq experiments, we identified 5125 differentially expressed transcripts in tumor versus normal organoids at different time points, in which the PTEN pathway resulted of particular interest, as also confirmed by further phospho-proteomics analysis. Interestingly, we identified the PTEN c.806_817dup (NM_000314) mutation, which has never been reported previously and is predicted to be deleterious according to the American College of Medical Genetics and Genomics (ACMG) classification. Conclusion The crosstalk of genomic, transcriptomic and phosphoproteomic data allowed to observe that PDCOs recapitulate, at the molecular level, the tumor of origin, accumulating mutations over time that potentially mimic the evolution of the patient’s tumor, underlining relevant potentialities of this 3D model. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-021-01986-8.
Collapse
|
48
|
Guo F, Hall AR, Tape CJ, Ling S, Pointon A. Intra- and intercellular signaling pathways associated with drug-induced cardiac pathophysiology. Trends Pharmacol Sci 2021; 42:675-687. [PMID: 34092416 DOI: 10.1016/j.tips.2021.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 04/20/2021] [Accepted: 05/06/2021] [Indexed: 11/30/2022]
Abstract
Cardiac physiology and homeostasis are maintained by the interaction of multiple cell types, via both intra- and intercellular signaling pathways. Perturbations in these signaling pathways induced by oncology therapies can reduce cardiac function, ultimately leading to heart failure. As cancer survival increases, related cardiovascular complications are becoming increasingly prevalent, thus identifying the perturbations and cell signaling drivers of cardiotoxicity is increasingly important. Here, we discuss the homotypic and heterotypic cellular interactions that form the basis of intra- and intercellular cardiac signaling pathways, and how oncological agents disrupt these pathways, leading to heart failure. We also highlight the emerging systems biology techniques that can be applied, enabling a deeper understanding of the intra- and intercellular signaling pathways across multiple cell types associated with cardiovascular toxicity.
Collapse
Affiliation(s)
- Fei Guo
- Functional and Mechanistic Safety, Clinical Pharmacology and Safety Sciences, Research and Development, AstraZeneca, Cambridge, UK; Cell Communication Laboratory, Department of Oncology, University College London Cancer Institute, London, WC1E 6DD, UK
| | - Andrew R Hall
- Functional and Mechanistic Safety, Clinical Pharmacology and Safety Sciences, Research and Development, AstraZeneca, Cambridge, UK
| | - Christopher J Tape
- Cell Communication Laboratory, Department of Oncology, University College London Cancer Institute, London, WC1E 6DD, UK
| | - Stephanie Ling
- Imaging and Data Analytics, Clinical Pharmacology and Safety Sciences, Research and Development, AstraZeneca, Cambridge, UK
| | - Amy Pointon
- Functional and Mechanistic Safety, Clinical Pharmacology and Safety Sciences, Research and Development, AstraZeneca, Cambridge, UK.
| |
Collapse
|
49
|
Organoids and Colorectal Cancer. Cancers (Basel) 2021; 13:cancers13112657. [PMID: 34071313 PMCID: PMC8197877 DOI: 10.3390/cancers13112657] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/20/2021] [Accepted: 05/24/2021] [Indexed: 12/12/2022] Open
Abstract
Organoids were first established as a three-dimensional cell culture system from mouse small intestine. Subsequent development has made organoids a key system to study many human physiological and pathological processes that affect a variety of tissues and organs. In particular, organoids are becoming very useful tools to dissect colorectal cancer (CRC) by allowing the circumvention of classical problems and limitations, such as the impossibility of long-term culture of normal intestinal epithelial cells and the lack of good animal models for CRC. In this review, we describe the features and current knowledge of intestinal organoids and how they are largely contributing to our better understanding of intestinal cell biology and CRC genetics. Moreover, recent data show that organoids are appropriate systems for antitumoral drug testing and for the personalized treatment of CRC patients.
Collapse
|
50
|
Pape J, Emberton M, Cheema U. 3D Cancer Models: The Need for a Complex Stroma, Compartmentalization and Stiffness. Front Bioeng Biotechnol 2021; 9:660502. [PMID: 33912551 PMCID: PMC8072339 DOI: 10.3389/fbioe.2021.660502] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 03/22/2021] [Indexed: 12/19/2022] Open
Abstract
The use of tissue-engineered 3D models of cancer has grown in popularity with recent advances in the field of cancer research. 3D models are inherently more biomimetic compared to 2D cell monolayers cultured on tissue-culture plastic. Nevertheless 3D models still lack the cellular and matrix complexity of native tissues. This review explores different 3D models currently used, outlining their benefits and limitations. Specifically, this review focuses on stiffness and collagen density, compartmentalization, tumor-stroma cell population and extracellular matrix composition. Furthermore, this review explores the methods utilized in different models to directly measure cancer invasion and growth. Of the models evaluated, with PDX and in vivo as a relative "gold standard", tumoroids were deemed as comparable 3D cancer models with a high degree of biomimicry, in terms of stiffness, collagen density and the ability to compartmentalize the tumor and stroma. Future 3D models for different cancer types are proposed in order to improve the biomimicry of cancer models used for studying disease progression.
Collapse
Affiliation(s)
- Judith Pape
- Division of Surgery and Interventional Science, Department of Targeted Intervention, Centre for 3D Models of Health and Disease, University College London, London, United Kingdom
| | - Mark Emberton
- Faculty of Medical Sciences, University College London, London, United Kingdom
| | - Umber Cheema
- Division of Surgery and Interventional Science, Department of Targeted Intervention, Centre for 3D Models of Health and Disease, University College London, London, United Kingdom
| |
Collapse
|