1
|
Baglamis S, Sheraton VM, van Neerven SM, Logiantara A, Nijman LE, Hageman LA, Léveillé N, Elbers CC, Bijlsma MF, Vermeulen L, Krawczyk PM, Lenos KJ. Clonal dispersal is associated with tumor heterogeneity and poor prognosis in colorectal cancer. iScience 2025; 28:112403. [PMID: 40330878 PMCID: PMC12051713 DOI: 10.1016/j.isci.2025.112403] [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: 10/10/2024] [Revised: 01/27/2025] [Accepted: 04/07/2025] [Indexed: 05/08/2025] Open
Abstract
Clonal dispersal, resulting from the intermingling of tumor cell subpopulations, is thought to be a key driver of tumor heterogeneity. Despite advances in spatial modeling of cancer biology, quantification of clonal dispersal has been challenging. This study introduces a straightforward method, relying on fluorescent cell barcoding, to quantify clonal dispersal in various in vitro and in vivo models of colorectal cancer (CRC). Our approach allows for precise localization of clones and uncovering the degree of clonal mixing across different CRC models. Our findings suggest that clonal dispersal is correlated with the expression of genes involved in epithelial-mesenchymal transition and CMS4-related signaling pathways. We further identify a dispersal gene signature, associated with intratumor heterogeneity, which is a robust clinical predictor of poor prognosis and recurrence in CRC, highlighting its potential as a prognostic marker and a putative direction for therapeutic targeting.
Collapse
Affiliation(s)
- Selami Baglamis
- Amsterdam UMC, University of Amsterdam, Laboratory for Experimental Oncology and Radiobiology, 1081 BT Amsterdam, the Netherlands
- Cancer Center Amsterdam, Amsterdam, the Netherlands
- Oncode Institute, Amsterdam, 3521 AL Utrecht, the Netherlands
- Amsterdam UMC, University of Amsterdam, Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, Amsterdam, the Netherlands
| | - Vivek M. Sheraton
- Amsterdam UMC, University of Amsterdam, Laboratory for Experimental Oncology and Radiobiology, 1081 BT Amsterdam, the Netherlands
- Cancer Center Amsterdam, Amsterdam, the Netherlands
- Oncode Institute, Amsterdam, 3521 AL Utrecht, the Netherlands
- Amsterdam UMC, University of Amsterdam, Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, Amsterdam, the Netherlands
- University of Amsterdam, Informatics Institute, Computational Science Lab, 1090 GH Amsterdam, the Netherlands
| | - Sanne M. van Neerven
- Amsterdam UMC, University of Amsterdam, Laboratory for Experimental Oncology and Radiobiology, 1081 BT Amsterdam, the Netherlands
- Cancer Center Amsterdam, Amsterdam, the Netherlands
- Oncode Institute, Amsterdam, 3521 AL Utrecht, the Netherlands
- Amsterdam UMC, University of Amsterdam, Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, Amsterdam, the Netherlands
- University of Cambridge, Wellcome Trust–Cancer Research UK Gurdon Institute, Cambridge CB2 1QN, UK
| | - Adrian Logiantara
- Amsterdam UMC, University of Amsterdam, Laboratory for Experimental Oncology and Radiobiology, 1081 BT Amsterdam, the Netherlands
- Cancer Center Amsterdam, Amsterdam, the Netherlands
- Oncode Institute, Amsterdam, 3521 AL Utrecht, the Netherlands
- Amsterdam UMC, University of Amsterdam, Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, Amsterdam, the Netherlands
| | - Lisanne E. Nijman
- Amsterdam UMC, University of Amsterdam, Laboratory for Experimental Oncology and Radiobiology, 1081 BT Amsterdam, the Netherlands
- Cancer Center Amsterdam, Amsterdam, the Netherlands
- Oncode Institute, Amsterdam, 3521 AL Utrecht, the Netherlands
- Amsterdam UMC, University of Amsterdam, Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, Amsterdam, the Netherlands
| | - Laura A. Hageman
- Amsterdam UMC, University of Amsterdam, Laboratory for Experimental Oncology and Radiobiology, 1081 BT Amsterdam, the Netherlands
- Cancer Center Amsterdam, Amsterdam, the Netherlands
- Oncode Institute, Amsterdam, 3521 AL Utrecht, the Netherlands
| | - Nicolas Léveillé
- Amsterdam UMC, University of Amsterdam, Laboratory for Experimental Oncology and Radiobiology, 1081 BT Amsterdam, the Netherlands
- Cancer Center Amsterdam, Amsterdam, the Netherlands
- Oncode Institute, Amsterdam, 3521 AL Utrecht, the Netherlands
- Amsterdam UMC, University of Amsterdam, Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, Amsterdam, the Netherlands
| | - Clara C. Elbers
- Amsterdam UMC, University of Amsterdam, Laboratory for Experimental Oncology and Radiobiology, 1081 BT Amsterdam, the Netherlands
- Cancer Center Amsterdam, Amsterdam, the Netherlands
- Oncode Institute, Amsterdam, 3521 AL Utrecht, the Netherlands
- Amsterdam UMC, University of Amsterdam, Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, Amsterdam, the Netherlands
| | - Maarten F. Bijlsma
- Amsterdam UMC, University of Amsterdam, Laboratory for Experimental Oncology and Radiobiology, 1081 BT Amsterdam, the Netherlands
- Cancer Center Amsterdam, Amsterdam, the Netherlands
- Oncode Institute, Amsterdam, 3521 AL Utrecht, the Netherlands
- Amsterdam UMC, University of Amsterdam, Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, Amsterdam, the Netherlands
| | - Louis Vermeulen
- Amsterdam UMC, University of Amsterdam, Laboratory for Experimental Oncology and Radiobiology, 1081 BT Amsterdam, the Netherlands
- Cancer Center Amsterdam, Amsterdam, the Netherlands
- Oncode Institute, Amsterdam, 3521 AL Utrecht, the Netherlands
- Amsterdam UMC, University of Amsterdam, Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, Amsterdam, the Netherlands
- Genentech, Department of Discovery Oncology, South San Francisco, CA 94080, USA
| | - Przemek M. Krawczyk
- Cancer Center Amsterdam, Amsterdam, the Netherlands
- Amsterdam UMC, University of Amsterdam, Department of Medical Biology, 1105 AZ Amsterdam, the Netherlands
| | - Kristiaan J. Lenos
- Amsterdam UMC, University of Amsterdam, Laboratory for Experimental Oncology and Radiobiology, 1081 BT Amsterdam, the Netherlands
- Cancer Center Amsterdam, Amsterdam, the Netherlands
- Oncode Institute, Amsterdam, 3521 AL Utrecht, the Netherlands
- Amsterdam UMC, University of Amsterdam, Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, Amsterdam, the Netherlands
| |
Collapse
|
2
|
Jünemann W, Bley I, Rekowski L, Klokow M, Herppich S, Müller I, Cornils K. GD2-CAR NK-92 cell activity against neuroblastoma cells is insusceptible to TIGIT knockout. Cancer Immunol Immunother 2025; 74:191. [PMID: 40317320 PMCID: PMC12049354 DOI: 10.1007/s00262-025-04010-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 03/03/2025] [Indexed: 05/07/2025]
Abstract
Immunotherapy by inhibition of immune checkpoint (IC) molecules has emerged as an important cancer therapy. Among these lC, the poliovirus receptor/poliovirus receptor-like 2 protein (PVR/PVRL2)-TIGIT axis was discovered as potential target for various cancers. For neuroblastoma (NB), the most common extracranial solid cancer in children, no effective IC therapy has been established yet. To investigate the PVR/PVRL2-TIGIT IC axis as a new target for the treatment of NB, we analysed whether PVR and PVRL2 influence the survival of patients and verified the expression of the receptors on NB cell lines. To disrupt the checkpoint axis, we performed single and double knockouts of these receptors on NB cell lines and subsequently removed TIGIT, an inhibitory receptor on immune effector cells, from NK-92 cells. Finally, we combined checkpoint inhibition with GD2-CAR NK-92 cells and investigated changes in cytotoxicity. Using RNA-Seq data we showed that the expression of PVR and PVRL2 on NB cells correlates to a lower event-free survival of patients. CRISPR/Cas9 knockouts of PVR and PVRL2 showed no improved cytotoxic activity of NK-92 cells. We observed enhanced lysis of NB cells using TIGIT-deficient NK-92 cells. However, the cytotoxicity of GD2-CAR NK-92 was not significantly enhanced. In summary, we have shown that in addition to the interaction of PVR/PVRL2 and TIGIT on engineered immune effector cells against NB, pleiotropic ligands appear to be relevant. Deletion of TIGIT from immune effector cells is a promising approach to protect these cells from tumour-associated inhibitory signals but cannot enhance the effect of GD2-CAR-NK-92 cells.
Collapse
MESH Headings
- Humans
- Neuroblastoma/immunology
- Neuroblastoma/therapy
- Neuroblastoma/pathology
- Receptors, Immunologic/genetics
- Receptors, Immunologic/metabolism
- Receptors, Immunologic/immunology
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Gangliosides/immunology
- Cell Line, Tumor
- Immunotherapy, Adoptive/methods
- Gene Knockout Techniques
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/genetics
- Receptors, Virus/genetics
Collapse
Affiliation(s)
- Wiebke Jünemann
- Children's Cancer Centre Research Institute Hamburg, Hamburg, Germany
- Division of Paediatric Stem Cell Transplantation and Immunology, Department of Paediatric Haematology and Oncology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Isabelle Bley
- Children's Cancer Centre Research Institute Hamburg, Hamburg, Germany
- Division of Paediatric Stem Cell Transplantation and Immunology, Department of Paediatric Haematology and Oncology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Laura Rekowski
- Children's Cancer Centre Research Institute Hamburg, Hamburg, Germany
- Division of Paediatric Stem Cell Transplantation and Immunology, Department of Paediatric Haematology and Oncology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Marianne Klokow
- Children's Cancer Centre Research Institute Hamburg, Hamburg, Germany
- Division of Paediatric Stem Cell Transplantation and Immunology, Department of Paediatric Haematology and Oncology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Susanne Herppich
- Children's Cancer Centre Research Institute Hamburg, Hamburg, Germany
- Division of Paediatric Stem Cell Transplantation and Immunology, Department of Paediatric Haematology and Oncology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Ingo Müller
- Children's Cancer Centre Research Institute Hamburg, Hamburg, Germany
- Division of Paediatric Stem Cell Transplantation and Immunology, Department of Paediatric Haematology and Oncology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Kerstin Cornils
- Children's Cancer Centre Research Institute Hamburg, Hamburg, Germany.
- Division of Paediatric Stem Cell Transplantation and Immunology, Department of Paediatric Haematology and Oncology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
3
|
van Ooijen H, Verron Q, Zhang H, Sandoz PA, Frisk TW, Carannante V, Olofsson K, Wagner AK, Sandström N, Önfelt B. A thermoplastic chip for 2D and 3D correlative assays combining screening and high-resolution imaging of immune cell responses. CELL REPORTS METHODS 2025; 5:100965. [PMID: 39826552 PMCID: PMC11841093 DOI: 10.1016/j.crmeth.2025.100965] [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: 08/10/2023] [Revised: 10/02/2024] [Accepted: 12/31/2024] [Indexed: 01/22/2025]
Abstract
We present an easy-to-use, disposable, thermoplastic microwell chip designed to support screening and high-resolution imaging of single-cell behavior in two- and three-dimensional (2D and 3D) cell cultures. We show that the chip has excellent optical properties and provide simple protocols for efficient long-term cell culture of suspension and adherent cells, the latter grown either as monolayers or as hundreds of single, uniformly sized spheroids. We then demonstrate the applicability of the system for single-cell analysis by correlating the dynamic cytotoxic response of single immune cells grown under different metabolic conditions to their intracellular cytolytic load at the end of the assay. Additionally, we illustrate highly multiplex cytotoxicity screening of tumor spheroids in the chip, comparing the effect of environment cues characteristic of the tumor microenvironment on natural killer (NK)-cell-induced killing. Following the functional screening, we perform high-resolution 3D immunofluorescent imaging of infiltrating NK cells within the spheroid volumes.
Collapse
Affiliation(s)
- Hanna van Ooijen
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Quentin Verron
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Hanqing Zhang
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Patrick A Sandoz
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Thomas W Frisk
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Valentina Carannante
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Karl Olofsson
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Arnika K Wagner
- Department of Medicine, Center for Hematology and Regenerative Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden; Haematology Center, Karolinska University Hospital, Stockholm, Sweden
| | - Niklas Sandström
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Björn Önfelt
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden; Department of Medicine, Center for Infectious Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
4
|
Maltseva D, Kirillov I, Zhiyanov A, Averinskaya D, Suvorov R, Gubani D, Kudriaeva A, Belogurov A, Tonevitsky A. Incautious design of shRNAs for stable overexpression of miRNAs could result in generation of undesired isomiRs. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2024; 1867:195046. [PMID: 38876159 DOI: 10.1016/j.bbagrm.2024.195046] [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: 11/17/2023] [Revised: 06/03/2024] [Accepted: 06/06/2024] [Indexed: 06/16/2024]
Abstract
shRNA-mediated strategy of miRNA overexpression based on RNA Polymerase III (Pol III) expression cassettes is widely used for miRNA functional studies. For some miRNAs, e.g., encoded in the genome as a part of a polycistronic miRNA cluster, it is most likely the only way for their individual stable overexpression. Here we have revealed that expression of miRNAs longer than 19 nt (e.g. 23 nt in length hsa-miR-93-5p) using such approach could be accompanied by undesired predominant generation of 5' end miRNA isoforms (5'-isomiRs). Extra U residues (up to five) added by Pol III at the 3' end of the transcribed shRNA during transcription termination could cause a shift in the Dicer cleavage position of the shRNA. This results in the formation of 5'-isomiRs, which have a significantly altered seed region compared to the initially encoded canonical hsa-miR-93-5p. We demonstrated that the commonly used qPCR method is insensitive to the formation of 5'-isomiRs and cannot be used to confirm miRNA overexpression. However, the predominant expression of 5'-isomiRs without three or four first nucleotides instead of the canonical isoform could be disclosed based on miRNA-Seq analysis. Moreover, mRNA sequencing data showed that the 5'-isomiRs of hsa-miR-93-5p presumably regulate their own mRNA targets. Thus, omitting miRNA-Seq analysis may lead to erroneous conclusions regarding revealed mRNA targets and possible molecular mechanisms in which studied miRNA is involved. Overall, the presented results show that structures of shRNAs for stable overexpression of miRNAs requires careful design to avoid generation of undesired 5'-isomiRs.
Collapse
Affiliation(s)
- Diana Maltseva
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Moscow 101000, Russia
| | - Ivan Kirillov
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Moscow 101000, Russia
| | - Anton Zhiyanov
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Moscow 101000, Russia
| | - Daria Averinskaya
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Moscow 101000, Russia
| | - Roman Suvorov
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Moscow 101000, Russia
| | - Daria Gubani
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Moscow 101000, Russia
| | - Anna Kudriaeva
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| | - Alexey Belogurov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| | - Alexander Tonevitsky
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Moscow 101000, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia; Art Photonics GmbH, Berlin 12489, Germany.
| |
Collapse
|
5
|
Shulgin A, Spirin P, Lebedev T, Kravchenko A, Glasunov V, Yermak I, Prassolov V. Comparative study of HIV-1 inhibition efficiency by carrageenans from red seaweeds family gigartinaceae, Tichocarpaceae and Phyllophoraceae. Heliyon 2024; 10:e33407. [PMID: 39050420 PMCID: PMC11267007 DOI: 10.1016/j.heliyon.2024.e33407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 06/14/2024] [Accepted: 06/20/2024] [Indexed: 07/27/2024] Open
Abstract
The efficiency of human immunodeficiency virus-1 (HIV-1) inhibition by sulfated polysaccharides isolated from the various families of red algae of the Far East Pacific coast were studied. The anti-HIV-1 activity of kappa and lambda-carrageenans from Chondrus armatus, original highly sulfated X-carrageenan with low content of 3,6-anhydrogalactose from Tichocarpus crinitus and i/κ-carrageenan with hybrid structure isolated from Ahnfeltiopsis flabelliformis was found. The antiviral action of these polysaccharides and its low-weight oligosaccharide was compared with commercial κ-carrageenan. Here we used the HIV-1-based lentiviral particles and evaluated that these carrageenans in non-toxic concentrations significantly suppress the transduction potential of lentiviral particles pseudotyped with different envelope proteins, targeting cells of neuronal or T-cell origin. The antiviral action of these carrageenans was confirmed using the chimeric replication competent Mo-MuLV (Moloney murine leukemia retrovirus) encoding marker eGFP protein. We found that X-carrageenans from T. crinitus and its low weight derivative and λ-carrageenan from C. armatus effectively suppress the infection caused by retrovirus. The obtained data suggest that the differences in the suppressive effect of carrageenans on the transduction efficiency of HIV-1 based lentiviral particles may be related to the structural features of the studied polysaccharides.
Collapse
Affiliation(s)
- Andrey Shulgin
- Department of Cancer Cell Biology, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova 32, 119991, Moscow, Russia
| | - Pavel Spirin
- Department of Cancer Cell Biology, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova 32, 119991, Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova 32, 119991, Moscow, Russia
| | - Timofey Lebedev
- Department of Cancer Cell Biology, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova 32, 119991, Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova 32, 119991, Moscow, Russia
| | - Anna Kravchenko
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Sciences, 690022, Vladivostok, Russia
| | - Valery Glasunov
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Sciences, 690022, Vladivostok, Russia
| | - Irina Yermak
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Sciences, 690022, Vladivostok, Russia
| | - Vladimir Prassolov
- Department of Cancer Cell Biology, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova 32, 119991, Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova 32, 119991, Moscow, Russia
| |
Collapse
|
6
|
Xhaxho S, Chen-Wichmann L, Kreissig S, Windisch R, Gottschlich A, Nandi S, Schabernack S, Kohler I, Kellner C, Kobold S, Humpe A, Wichmann C. Efficient Chimeric Antigen Receptor T-Cell Generation Starting with Leukoreduction System Chambers of Thrombocyte Apheresis Sets. Transfus Med Hemother 2024; 51:111-118. [PMID: 38584695 PMCID: PMC10996058 DOI: 10.1159/000532130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 07/17/2023] [Indexed: 04/09/2024] Open
Abstract
Introduction Primary human blood cells represent an essential model system to study physiology and disease. However, human blood is a limited resource. During healthy donor plateletpheresis, the leukoreduction system chamber (LRSC) reduces the leukocyte amount within the subsequent platelet concentrate through saturated, fluidized, particle bed filtration technology. Normally, the LRSC is discarded after apheresis is completed. Compared to peripheral blood, LRSC yields 10-fold mononuclear cell concentration. Methods To explore if those retained leukocytes are attractive for research purposes, we isolated CD3+ T cells from the usually discarded LRSCs via density gradient centrifugation in order to manufacture CD19-targeted chimeric antigen receptor (CAR) T cells. Results Immunophenotypic characterization revealed viable and normal CD4+ and CD8+ T-cell populations within LRSC, with low CD19+ B cell counts. Magnetic-activated cell sorting (MACS) purified CD3+ T cells were transduced with CD19 CAR-encoding lentiviral self-inactivating vectors using concentrated viral supernatants. Robust CD19 CAR cell surface expression on transduced T cells was confirmed by flow cytometry. CD19 CAR T cells were further enriched through anti-CAR MACS, yielding 80% CAR+ T-cell populations. In vitro CAR T cell expansion to clinically relevant numbers was achieved. To prove functionality, CAR T cells were co-incubated with the human CD19+ B cell precursor leukemia cell line Nalm6. Compared to unmodified T cells, CD19 CAR T cells effectively eradicated Nalm6 cells. Conclusion Taken together, we can show that lymphocytes isolated from LRSCs of plateletpheresis sets can be efficiently used for the generation of functional CAR T cells for experimental purposes.
Collapse
Affiliation(s)
- Stefani Xhaxho
- Division of Transfusion Medicine, Cell Therapeutics and Haemostaseology, University Hospital, LMU Munich, Munich, Germany
| | - Linping Chen-Wichmann
- Division of Transfusion Medicine, Cell Therapeutics and Haemostaseology, University Hospital, LMU Munich, Munich, Germany
| | - Sophie Kreissig
- Division of Transfusion Medicine, Cell Therapeutics and Haemostaseology, University Hospital, LMU Munich, Munich, Germany
| | - Roland Windisch
- Division of Transfusion Medicine, Cell Therapeutics and Haemostaseology, University Hospital, LMU Munich, Munich, Germany
| | - Adrian Gottschlich
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, LMU Munich, Munich, Germany
| | - Sayantan Nandi
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, LMU Munich, Munich, Germany
| | - Sophie Schabernack
- Division of Transfusion Medicine, Cell Therapeutics and Haemostaseology, University Hospital, LMU Munich, Munich, Germany
| | - Irmgard Kohler
- Division of Transfusion Medicine, Cell Therapeutics and Haemostaseology, University Hospital, LMU Munich, Munich, Germany
| | - Christian Kellner
- Division of Transfusion Medicine, Cell Therapeutics and Haemostaseology, University Hospital, LMU Munich, Munich, Germany
| | - Sebastian Kobold
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, LMU Munich, Munich, Germany
- Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Munich, Research Center for Environmental Health (HMGU), Neuherberg, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Andreas Humpe
- Division of Transfusion Medicine, Cell Therapeutics and Haemostaseology, University Hospital, LMU Munich, Munich, Germany
| | - Christian Wichmann
- Division of Transfusion Medicine, Cell Therapeutics and Haemostaseology, University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
7
|
Baglamis S, Sheraton VM, Meijer D, Qian H, Hoebe RA, Lenos KJ, Betjes MA, Betjes MA, Tans S, van Zon J, Vermeulen L, Krawczyk PM. Using picoliter droplet deposition to track clonal competition in adherent and organoid cancer cell cultures. Sci Rep 2023; 13:18832. [PMID: 37914743 PMCID: PMC10620187 DOI: 10.1038/s41598-023-42849-w] [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: 02/12/2023] [Accepted: 09/15/2023] [Indexed: 11/03/2023] Open
Abstract
Clonal growth and competition underlie processes of key relevance in etiology, progression and therapy response across all cancers. Here, we demonstrate a novel experimental approach, based on multi-color, fluorescent tagging of cell nuclei, in combination with picoliter droplet deposition, to study the clonal dynamics in two- and three-dimensional cell cultures. The method allows for the simultaneous visualization and analysis of multiple clones in individual multi-clonal colonies, providing a powerful tool for studying clonal dynamics and identifying clonal populations with distinct characteristics. Results of our experiments validate the utility of the method in studying clonal dynamics in vitro, and reveal differences in key aspects of clonal behavior of different cancer cell lines in monoculture conditions, as well as in co-cultures with stromal fibroblasts.
Collapse
Affiliation(s)
- Selami Baglamis
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Cancer Center Amsterdam, 1081 HV, Amsterdam, The Netherlands
- Oncode Institute, 3521 AL, Utrecht, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, 1105 AZ, Amsterdam, The Netherlands
| | - Vivek M Sheraton
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Cancer Center Amsterdam, 1081 HV, Amsterdam, The Netherlands
- Oncode Institute, 3521 AL, Utrecht, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, 1105 AZ, Amsterdam, The Netherlands
- Institute for Advanced Study, University of Amsterdam, 1012 WX, Amsterdam, The Netherlands
| | - Debora Meijer
- Cancer Center Amsterdam, 1081 HV, Amsterdam, The Netherlands
- Department of Medical Biology, Amsterdam University Medical Centers (location AMC), University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Haibin Qian
- Cancer Center Amsterdam, 1081 HV, Amsterdam, The Netherlands
- Department of Medical Biology, Amsterdam University Medical Centers (location AMC), University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Ron A Hoebe
- Cancer Center Amsterdam, 1081 HV, Amsterdam, The Netherlands
- Department of Medical Biology, Amsterdam University Medical Centers (location AMC), University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Kristiaan J Lenos
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Cancer Center Amsterdam, 1081 HV, Amsterdam, The Netherlands
- Oncode Institute, 3521 AL, Utrecht, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, 1105 AZ, Amsterdam, The Netherlands
| | - Max A Betjes
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Cancer Center Amsterdam, 1081 HV, Amsterdam, The Netherlands
- Oncode Institute, 3521 AL, Utrecht, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, 1105 AZ, Amsterdam, The Netherlands
| | | | | | | | - Louis Vermeulen
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
- Cancer Center Amsterdam, 1081 HV, Amsterdam, The Netherlands.
- Oncode Institute, 3521 AL, Utrecht, The Netherlands.
- Amsterdam Gastroenterology Endocrinology Metabolism, 1105 AZ, Amsterdam, The Netherlands.
| | - Przemek M Krawczyk
- Cancer Center Amsterdam, 1081 HV, Amsterdam, The Netherlands.
- Department of Medical Biology, Amsterdam University Medical Centers (location AMC), University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| |
Collapse
|
8
|
Qian H, Baglamis S, Redeker F, Raaijman J, Hoebe RA, Sheraton VM, Vermeulen L, Krawczyk PM. High-Content and High-Throughput Clonogenic Survival Assay Using Fluorescence Barcoding. Cancers (Basel) 2023; 15:4772. [PMID: 37835466 PMCID: PMC10571559 DOI: 10.3390/cancers15194772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 09/25/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
The Clonogenic Survival Assay (CSA) is a fundamental tool employed to assess cell survival and proliferative potential in cancer research. Despite its importance, CSA faces limitations, primarily its time- and labor-intensive nature and its binary output. To overcome these limitations and enhance CSA's utility, several approaches have been developed, focusing on increasing the throughput. However, achieving both high-content and high-throughput analyses simultaneously has remained a challenge. In this paper, we introduce LeGO-CSA, an extension of the classical CSA that employs the imaging of cell nuclei barcoded with fluorescent lentiviral gene ontology markers, enabling both high-content and high-throughput analysis. To validate our approach, we contrasted it with results from a classical assay and conducted a proof-of-concept screen of small-molecule inhibitors targeting various pathways relevant to cancer treatment. Notably, our results indicate that the classical CSA may underestimate clonogenicity and unveil intriguing aspects of clonal cell growth. We demonstrate the potential of LeGO-CSA to offer a robust approach for assessing cell survival and proliferation with enhanced precision and throughput, with promising implications for accelerating drug discovery and contributing to a more comprehensive understanding of cellular behavior in cancer.
Collapse
Affiliation(s)
- Haibin Qian
- Department of Medical Biology, Amsterdam University Medical Centers (Location AMC), Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (H.Q.); (R.A.H.)
- Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands; (S.B.); (V.M.S.); (L.V.)
| | - Selami Baglamis
- Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands; (S.B.); (V.M.S.); (L.V.)
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
- Oncode Institute, 3521 AL Utrecht, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, 1105 AZ Amsterdam, The Netherlands
| | - Fumei Redeker
- Department of Medical Biology, Amsterdam University Medical Centers (Location AMC), Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (H.Q.); (R.A.H.)
- Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands; (S.B.); (V.M.S.); (L.V.)
| | - Julia Raaijman
- Department of Medical Biology, Amsterdam University Medical Centers (Location AMC), Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (H.Q.); (R.A.H.)
- Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands; (S.B.); (V.M.S.); (L.V.)
| | - Ron A. Hoebe
- Department of Medical Biology, Amsterdam University Medical Centers (Location AMC), Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (H.Q.); (R.A.H.)
- Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands; (S.B.); (V.M.S.); (L.V.)
| | - Vivek M. Sheraton
- Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands; (S.B.); (V.M.S.); (L.V.)
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
- Oncode Institute, 3521 AL Utrecht, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, 1105 AZ Amsterdam, The Netherlands
- Institute for Advanced Study, University of Amsterdam, 1012 WX Amsterdam, The Netherlands
| | - Louis Vermeulen
- Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands; (S.B.); (V.M.S.); (L.V.)
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
- Oncode Institute, 3521 AL Utrecht, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, 1105 AZ Amsterdam, The Netherlands
| | - Przemek M. Krawczyk
- Department of Medical Biology, Amsterdam University Medical Centers (Location AMC), Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (H.Q.); (R.A.H.)
- Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands; (S.B.); (V.M.S.); (L.V.)
| |
Collapse
|
9
|
Glumakova K, Ivanov G, Vedernikova V, Shyrokova L, Lebedev T, Stomakhin A, Zenchenko A, Oslovsky V, Drenichev M, Prassolov V, Spirin P. Nucleoside Analog 2',3'-Isopropylidene-5-Iodouridine as Novel Efficient Inhibitor of HIV-1. Pharmaceutics 2023; 15:2389. [PMID: 37896149 PMCID: PMC10610023 DOI: 10.3390/pharmaceutics15102389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 09/18/2023] [Accepted: 09/23/2023] [Indexed: 10/29/2023] Open
Abstract
Nucleoside reverse transcriptase inhibitors are the first class of drugs to be approved by the FDA for the suppression of HIV-1 and are widely used for this purpose in combination with drugs of other classes. Despite the progress in HIV-1 treatment, there is still the need to develop novel efficient antivirals. Here the efficiency of HIV-1 inhibition by a set of original 5-substituted uridine nucleosides was studied. We used the replication deficient human immunodeficiency virus (HIV-1)-based lentiviral particles and identified that among the studied compounds, 2',3'-isopropylidene-5-iodouridine was shown to cause anti-HIV-1 activity. Importantly, no toxic action of this compound against the cells of T-cell origin was found. We determined that this compound is significantly more efficient at suppressing HIV-1 compared to Azidothymidine (AZT) when taken at the high non-toxic concentrations. We did not find any profit when using AZT in combination with 2',3'-isopropylidene-5-iodouridine. 2',3'-Isopropylidene-5-iodouridine acts synergistically to repress HIV-1 when combined with the CDK4/6 inhibitor Palbociclib in low non-toxic concentration. No synergistic antiviral action was detected when AZT was combined with Palbociclib. We suggest 2',3'-isopropylidene-5-iodouridine as a novel perspective non-toxic compound that may be used for HIV-l suppression.
Collapse
Affiliation(s)
- Ksenia Glumakova
- Department of Cancer Cell Biology, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova 32, 119991 Moscow, Russia; (K.G.); (G.I.); (V.V.); (T.L.); (A.S.); (A.Z.); (V.O.); (M.D.)
- Moscow Institute of Physics and Technology, National Research University, Institutskiy per. 9, 141701 Dolgoprudny, Russia
| | - Georgy Ivanov
- Department of Cancer Cell Biology, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova 32, 119991 Moscow, Russia; (K.G.); (G.I.); (V.V.); (T.L.); (A.S.); (A.Z.); (V.O.); (M.D.)
| | - Valeria Vedernikova
- Department of Cancer Cell Biology, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova 32, 119991 Moscow, Russia; (K.G.); (G.I.); (V.V.); (T.L.); (A.S.); (A.Z.); (V.O.); (M.D.)
- Moscow Institute of Physics and Technology, National Research University, Institutskiy per. 9, 141701 Dolgoprudny, Russia
| | - Lena Shyrokova
- Department of Experimental Medical Science, Lund University, 221 84 Lund, Sweden;
| | - Timofey Lebedev
- Department of Cancer Cell Biology, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova 32, 119991 Moscow, Russia; (K.G.); (G.I.); (V.V.); (T.L.); (A.S.); (A.Z.); (V.O.); (M.D.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova 32, 119991 Moscow, Russia
| | - Andrei Stomakhin
- Department of Cancer Cell Biology, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova 32, 119991 Moscow, Russia; (K.G.); (G.I.); (V.V.); (T.L.); (A.S.); (A.Z.); (V.O.); (M.D.)
| | - Anastasia Zenchenko
- Department of Cancer Cell Biology, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova 32, 119991 Moscow, Russia; (K.G.); (G.I.); (V.V.); (T.L.); (A.S.); (A.Z.); (V.O.); (M.D.)
| | - Vladimir Oslovsky
- Department of Cancer Cell Biology, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova 32, 119991 Moscow, Russia; (K.G.); (G.I.); (V.V.); (T.L.); (A.S.); (A.Z.); (V.O.); (M.D.)
| | - Mikhail Drenichev
- Department of Cancer Cell Biology, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova 32, 119991 Moscow, Russia; (K.G.); (G.I.); (V.V.); (T.L.); (A.S.); (A.Z.); (V.O.); (M.D.)
| | - Vladimir Prassolov
- Department of Cancer Cell Biology, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova 32, 119991 Moscow, Russia; (K.G.); (G.I.); (V.V.); (T.L.); (A.S.); (A.Z.); (V.O.); (M.D.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova 32, 119991 Moscow, Russia
| | - Pavel Spirin
- Department of Cancer Cell Biology, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova 32, 119991 Moscow, Russia; (K.G.); (G.I.); (V.V.); (T.L.); (A.S.); (A.Z.); (V.O.); (M.D.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova 32, 119991 Moscow, Russia
| |
Collapse
|
10
|
Hershey BJ, Barozzi S, Orsenigo F, Pompei S, Iannelli F, Kamrad S, Matafora V, Pisati F, Calabrese L, Fragale G, Salvadori G, Martini E, Totaro MG, Magni S, Guan R, Parazzoli D, Maiuri P, Bachi A, Patil KR, Cosentino Lagomarsino M, Havas KM. Clonal cooperation through soluble metabolite exchange facilitates metastatic outgrowth by modulating Allee effect. SCIENCE ADVANCES 2023; 9:eadh4184. [PMID: 37713487 PMCID: PMC10881076 DOI: 10.1126/sciadv.adh4184] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 08/14/2023] [Indexed: 09/17/2023]
Abstract
Cancers feature substantial intratumoral heterogeneity of genetic and phenotypically distinct lineages. Although interactions between coexisting lineages are emerging as a potential contributor to tumor evolution, the extent and nature of these interactions remain largely unknown. We postulated that tumors develop ecological interactions that sustain diversity and facilitate metastasis. Using a combination of fluorescent barcoding, mathematical modeling, metabolic analysis, and in vivo models, we show that the Allee effect, i.e., growth dependency on population size, is a feature of tumor lineages and that cooperative ecological interactions between lineages alleviate the Allee barriers to growth in a model of triple-negative breast cancer. Soluble metabolite exchange formed the basis for these cooperative interactions and catalyzed the establishment of a polyclonal community that displayed enhanced metastatic dissemination and outgrowth in xenograft models. Our results highlight interclonal metabolite exchange as a key modulator of tumor ecology and a contributing factor to overcoming Allee effect-associated growth barriers to metastasis.
Collapse
Affiliation(s)
| | - Sara Barozzi
- IFOM ETS The AIRC Institute of Molecular Oncology, Milan, Italy
| | | | - Simone Pompei
- IFOM ETS The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Fabio Iannelli
- IFOM ETS The AIRC Institute of Molecular Oncology, Milan, Italy
| | | | | | | | | | | | | | | | | | - Serena Magni
- IFOM ETS The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Rui Guan
- Medical Research Council Toxicology Unit, Cambridge, UK
| | - Dario Parazzoli
- IFOM ETS The AIRC Institute of Molecular Oncology, Milan, Italy
| | | | - Angela Bachi
- IFOM ETS The AIRC Institute of Molecular Oncology, Milan, Italy
| | | | | | | |
Collapse
|
11
|
Barry-Carroll L, Greulich P, Marshall AR, Riecken K, Fehse B, Askew KE, Li K, Garaschuk O, Menassa DA, Gomez-Nicola D. Microglia colonize the developing brain by clonal expansion of highly proliferative progenitors, following allometric scaling. Cell Rep 2023; 42:112425. [PMID: 37099424 DOI: 10.1016/j.celrep.2023.112425] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 01/25/2023] [Accepted: 04/06/2023] [Indexed: 04/27/2023] Open
Abstract
Microglia arise from the yolk sac and enter the brain during early embryogenesis. Upon entry, microglia undergo in situ proliferation and eventually colonize the entire brain by the third postnatal week in mice. However, the intricacies of their developmental expansion remain unclear. Here, we characterize the proliferative dynamics of microglia during embryonic and postnatal development using complementary fate-mapping techniques. We demonstrate that the developmental colonization of the brain is facilitated by clonal expansion of highly proliferative microglial progenitors that occupy spatial niches throughout the brain. Moreover, the spatial distribution of microglia switches from a clustered to a random pattern between embryonic and late postnatal development. Interestingly, the developmental increase in microglial numbers follows the proportional growth of the brain in an allometric manner until a mosaic distribution has been established. Overall, our findings offer insight into how the competition for space may drive microglial colonization by clonal expansion during development.
Collapse
Affiliation(s)
- Liam Barry-Carroll
- School of Biological Sciences, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Philip Greulich
- School of Mathematical Sciences, University of Southampton, Southampton, UK; Institute for Life Sciences (IfLS), University of Southampton, Southampton, UK
| | - Abigail R Marshall
- School of Biological Sciences, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Kristoffer Riecken
- Research Department Cell and Gene Therapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Boris Fehse
- Research Department Cell and Gene Therapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Katharine E Askew
- School of Biological Sciences, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Kaizhen Li
- Department of Neurophysiology, University of Tübingen, Tübingen, Germany
| | - Olga Garaschuk
- Department of Neurophysiology, University of Tübingen, Tübingen, Germany
| | - David A Menassa
- School of Biological Sciences, University of Southampton, Southampton General Hospital, Southampton, UK; The Queen's College, University of Oxford, Oxford, UK
| | - Diego Gomez-Nicola
- School of Biological Sciences, University of Southampton, Southampton General Hospital, Southampton, UK; Institute for Life Sciences (IfLS), University of Southampton, Southampton, UK.
| |
Collapse
|
12
|
Cotner M, Meng S, Jost T, Gardner A, De Santiago C, Brock A. Integration of quantitative methods and mathematical approaches for the modeling of cancer cell proliferation dynamics. Am J Physiol Cell Physiol 2023; 324:C247-C262. [PMID: 36503241 PMCID: PMC9886359 DOI: 10.1152/ajpcell.00185.2022] [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: 05/02/2022] [Revised: 11/21/2022] [Accepted: 11/21/2022] [Indexed: 12/15/2022]
Abstract
Physiological processes rely on the control of cell proliferation, and the dysregulation of these processes underlies various pathological conditions, including cancer. Mathematical modeling can provide new insights into the complex regulation of cell proliferation dynamics. In this review, we first examine quantitative experimental approaches for measuring cell proliferation dynamics in vitro and compare the various types of data that can be obtained in these settings. We then explore the toolbox of common mathematical modeling frameworks that can describe cell behavior, dynamics, and interactions of proliferation. We discuss how these wet-laboratory studies may be integrated with different mathematical modeling approaches to aid the interpretation of the results and to enable the prediction of cell behaviors, specifically in the context of cancer.
Collapse
Affiliation(s)
- Michael Cotner
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas
| | - Sarah Meng
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas
| | - Tyler Jost
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas
| | - Andrea Gardner
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas
| | - Carolina De Santiago
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas
| | - Amy Brock
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas
| |
Collapse
|
13
|
Tissue factor-induced fibrinogenesis mediates cancer cell clustering and multiclonal peritoneal metastasis. Cancer Lett 2023; 553:215983. [PMID: 36404569 DOI: 10.1016/j.canlet.2022.215983] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/13/2022] [Accepted: 10/23/2022] [Indexed: 11/02/2022]
Abstract
Peritoneal metastasis is one of the most frequent causes of death in several types of advanced cancers; however, the underlying molecular mechanisms remain largely unknown. In this study, we exploited multicolor fluorescent lineage tracking to investigate the clonality of peritoneal metastasis in mouse xenograft models. When peritoneal metastasis was induced by intraperitoneal or orthotopic injection of multicolored cancer cells, each peritoneally metastasized tumor displayed multicolor fluorescence regardless of metastasis sites, indicating that it consists of multiclonal cancer cell populations. Multicolored cancer cell clusters form within the peritoneal cavity and collectively attach to the peritoneum. In vitro, peritoneal lavage fluid or cleared ascitic fluid derived from cancer patients induces cancer cell clustering, which is inhibited by anticoagulants. Cancer cell clusters formed in vitro and in vivo are associated with fibrin formation. Furthermore, tissue factor knockout in cancer cells abrogates cell clustering, peritoneal attachment, and peritoneal metastasis. Thus, we propose that cancer cells activate the coagulation cascade via tissue factor to form fibrin-mediated cell clusters and promote peritoneal attachment; these factors lead to the development of multiclonal peritoneal metastasis and may be therapeutic targets.
Collapse
|
14
|
Nikulin S, Razumovskaya A, Poloznikov A, Zakharova G, Alekseev B, Tonevitsky A. ELOVL5 and IGFBP6 genes modulate sensitivity of breast cancer cells to ferroptosis. Front Mol Biosci 2023; 10:1075704. [PMID: 36714261 PMCID: PMC9880435 DOI: 10.3389/fmolb.2023.1075704] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 01/03/2023] [Indexed: 01/14/2023] Open
Abstract
Introduction: Relapse of breast cancer is one of the key obstacles to successful treatment. Previously we have shown that low expression of ELOVL5 and IGFBP6 genes in breast cancer tissue corresponded to poor prognosis. ELOVL5 participates directly in the elongation of polyunsaturated fatty acids (PUFAs) that are considered to play an important role in cancer cell metabolism. Thus, in this work we studied the changes in lipid metabolism in breast cancer cells with reduced expression of either ELOVL5 or IGFBP6 gene. Methods: MDA-MB-231 cells with a stable knockdown of either ELOVL5 or IGFBP6 gene were used in this study. Transcriptomic and proteomic analysis as well as RT-PCR were utilized to assess gene expression. Content of individual fatty acids in the cells was measured with HPLC-MS. HPLC was used for analysis of the kinetics of PUFAs uptake. Cell viability was measured with MTS assay. Flow cytometry was used to measure activation of apoptosis. Fluorescent microscopy was utilized to assess accumulation of ROS and formation of lipid droplets. Glutathione peroxidase activity was measured with a colorimetric assay. Results: We found that the knockdown of IGFBP6 gene led to significant changes in the profile of fatty acids in the cells and in the expression of many genes associated with lipid metabolism. As some PUFAs are known to inhibit proliferation and cause death of cancer cells, we also tested the response of the cells to single PUFAs and to combinations of docosahexaenoic acid (DHA, a n-3 PUFA) with standard chemotherapeutic drugs. Our data suggest that external PUFAs cause cell death by activation of ferroptosis, an iron-dependent mechanism of cell death with excessive lipid peroxidation. Moreover, both knockdowns increased cells' sensitivity to ferroptosis, probably due to a significant decrease in the activity of the antioxidant enzyme GPX4. Addition of DHA to commonly used chemotherapeutic drugs enhanced their effect significantly, especially for the cells with low expression of IGFBP6 gene. Discussion: The results of this study suggest that addition of PUFAs to the treatment regimen for the patients with low expression of IGFBP6 and ELOVL5 genes can be potentially beneficial and is worth testing in a clinically relevant setting.
Collapse
Affiliation(s)
- Sergey Nikulin
- Faculty of Biology and Biotechnologies, Higher School of Economics, Moscow, Russia,*Correspondence: Sergey Nikulin,
| | | | - Andrey Poloznikov
- P. A. Hertsen Moscow Oncology Research Center, Branch of the National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Galina Zakharova
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Boris Alekseev
- P. A. Hertsen Moscow Oncology Research Center, Branch of the National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Alexander Tonevitsky
- Faculty of Biology and Biotechnologies, Higher School of Economics, Moscow, Russia,Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
15
|
Serrano A, Berthelet J, Naik SH, Merino D. Mastering the use of cellular barcoding to explore cancer heterogeneity. Nat Rev Cancer 2022; 22:609-624. [PMID: 35982229 DOI: 10.1038/s41568-022-00500-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/13/2022] [Indexed: 11/09/2022]
Abstract
Tumours are often composed of a multitude of malignant clones that are genomically unique, and only a few of them may have the ability to escape cancer therapy and grow as symptomatic lesions. As a result, tumours with a large degree of genomic diversity have a higher chance of leading to patient death. However, clonal fate can be driven by non-genomic features. In this context, new technologies are emerging not only to track the spatiotemporal fate of individual cells and their progeny but also to study their molecular features using various omics analysis. In particular, the recent development of cellular barcoding facilitates the labelling of tens to millions of cancer clones and enables the identification of the complex mechanisms associated with clonal fate in different microenvironments and in response to therapy. In this Review, we highlight the recent discoveries made using lentiviral-based cellular barcoding techniques, namely genetic and optical barcoding. We also emphasize the strengths and limitations of each of these technologies and discuss some of the key concepts that must be taken into consideration when one is designing barcoding experiments. Finally, we suggest new directions to further improve the use of these technologies in cancer research.
Collapse
Affiliation(s)
- Antonin Serrano
- Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, Victoria, Australia
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, Faculty of Medicine, Dentistry and Health Science, The University of Melbourne, Parkville, Victoria, Australia
| | - Jean Berthelet
- Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, Victoria, Australia
| | - Shalin H Naik
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, Faculty of Medicine, Dentistry and Health Science, The University of Melbourne, Parkville, Victoria, Australia
| | - Delphine Merino
- Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia.
- School of Cancer Medicine, La Trobe University, Bundoora, Victoria, Australia.
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.
- Department of Medical Biology, Faculty of Medicine, Dentistry and Health Science, The University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
16
|
Combined Targeting of AKT and mTOR Inhibits Tumor Formation of EpCAM+ and CD90+ Human Hepatocellular Carcinoma Cells in an Orthotopic Mouse Model. Cancers (Basel) 2022; 14:cancers14081882. [PMID: 35454789 PMCID: PMC9024696 DOI: 10.3390/cancers14081882] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 12/12/2022] Open
Abstract
The epithelial cell adhesion molecule (EpCAM) and Thy-1 cell surface antigen (CD90) have been implicated as cancer stem cell (CSC) markers in hepatocellular carcinoma (HCC). Expression of EpCAM and CD90 on HCC cells is associated with increased tumorigenicity, metastasis and poor prognosis. In this study, we demonstrate that combined treatment with AKT and mTOR inhibitors—i.e., MK2206 and RAD001—results in a synergistic reduction in proliferation of EpCAM+ and CD90+ HCC cells cultured either as adherent cells or as tumoroids in vitro. In addition, tumor growth was reduced by combined treatment with AKT and mTOR inhibitors in an orthotopic xenograft mouse model of an EpCAM+ HCC cell line (Huh7) and primary patient-derived EpCAM+ HCC cells (HCC1) as well as a CD90+ HCC-related cell line (SK-HEP1) in vivo. However, during AKT/mTOR treatment, outgrowth of therapy-resistant tumors was observed in all mice analyzed within a few weeks. Resistance was associated in most cases with restoration of AKT signaling in the tumors, intrahepatic metastases and distant metastases. In addition, an upregulation of the p38 MAPK pathway was identified in the AKT/mTOR inhibitor-resistant tumor cells by kinome profiling. The development of resistant cells during AKT/mTOR therapy was further analyzed by red-green-blue (RGB) marking of HCC cells, which revealed an outgrowth of a large number of Huh7 cells over a period of 6 months. In summary, our data demonstrate that combined treatment with AKT and mTOR inhibitors exhibits synergistic effects on proliferation of EpCAM+ as well as CD90+ HCC cells in vitro. However, the fast development of large numbers of resistant clones under AKT/mTOR therapy observed in vitro and in the orthotopic xenotransplantation mouse model in vivo strongly suggests that this therapy alone will not be sufficient to eliminate EpCAM+ or CD90+ cancer stem cells from HCC patients.
Collapse
|
17
|
Yurchenko AA, Pop OT, Ighilahriz M, Padioleau I, Rajabi F, Sharpe HJ, Poulalhon N, Dreno B, Khammari A, Delord M, Alberti A, Soufir N, Battistella M, Mourah S, Bouquet F, Savina A, Besse A, Mendez-Lopez M, Grange F, Monestier S, Mortier L, Meyer N, Dutriaux C, Robert C, Saiag P, Herms F, Lambert J, de Sauvage FJ, Dumaz N, Flatz L, Basset-Seguin N, Nikolaev SI. Frequency and Genomic Aspects of Intrinsic Resistance to Vismodegib in Locally Advanced Basal Cell Carcinoma. Clin Cancer Res 2022; 28:1422-1432. [PMID: 35078858 PMCID: PMC9365352 DOI: 10.1158/1078-0432.ccr-21-3764] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/03/2021] [Accepted: 01/20/2022] [Indexed: 01/07/2023]
Abstract
PURPOSE Vismodegib is approved for the treatment of locally advanced basal cell carcinoma (laBCC), but some cases demonstrate intrinsic resistance (IR) to the drug. We sought to assess the frequency of IR to vismodegib in laBCC and its underlying genomic mechanisms. EXPERIMENTAL DESIGN Response to vismodegib was evaluated in a cohort of 148 laBCC patients. Comprehensive genomic and transcriptomic profiling was performed in a subset of five intrinsically resistant BCC (IR-BCC). RESULTS We identified that IR-BCC represents 6.1% of laBCC in the studied cohort. Prior treatment with chemotherapy was associated with IR. Genetic events that were previously associated with acquired resistance (AR) in BCC or medulloblastoma were observed in three out of five IR-BCC. However, IR-BCCs were distinct by highly rearranged polyploid genomes. Functional analyses identified hyperactivation of the HIPPO-YAP and WNT pathways at RNA and protein levels in IR-BCC. In vitro assay on the BCC cell line further confirmed that YAP1 overexpression increases the cell proliferation rate. CONCLUSIONS IR to vismodegib is a rare event in laBCC. IR-BCCs frequently harbor resistance mutations in the Hh pathway, but also are characterized by hyperactivation of the HIPPO-YAP and WNT pathways.
Collapse
Affiliation(s)
- Andrey A. Yurchenko
- INSERM U981, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
| | - Oltin T. Pop
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | | | - Ismael Padioleau
- INSERM U981, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
| | - Fatemeh Rajabi
- INSERM U981, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
| | | | - Nicolas Poulalhon
- Service de dermatologie, Hôpital Lyon Sud, Hospices Civils de Lyon, Pierre Bénite, France
| | - Brigitte Dreno
- Department of Dermato-Oncology, CHU Nantes, Nantes Université, CIC 1413, Inserm UMR 1302/EMR6001 INCIT, F-44000 Nantes, France
| | - Amir Khammari
- Department of Dermato-Oncology, CHU Nantes, Nantes Université, CIC 1413, Inserm UMR 1302/EMR6001 INCIT, F-44000 Nantes, France
| | - Marc Delord
- Université de Paris, Hôpital Saint-Louis, Paris, France
- Department of Population Health Sciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | | | | | - Maxime Battistella
- INSERM U976, Hôpital Saint-Louis, Paris, France
- Université de Paris, Hôpital Saint-Louis, Paris, France
- Service d'anatomie pathologique, Hôpital Saint-Louis, Claude Vellefaux, Paris, France
| | - Samia Mourah
- INSERM U976, Hôpital Saint-Louis, Paris, France
- Université de Paris, Hôpital Saint-Louis, Paris, France
- Département de Génomique des Tumeurs Solides, Hôpital Saint-Louis, Claude Vellefaux, Paris, France
| | | | | | - Andrej Besse
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Max Mendez-Lopez
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Florent Grange
- Service de dermatologie, CHU Reims, Rue du general Koenig, Reims, France
- Service de Dermatologie, centre hospitalier de Valence, Valence, France
| | | | - Laurent Mortier
- Service de dermatologie, CHU Lille, Clin Dermato Hop Huriez, Rue Michel Polonovski, Lille, France
| | - Nicolas Meyer
- Service de dermatologie, Institut Univeristaire du Cancer et CHU de Toulouse, Hôpital Larrey, Toulouse, France
| | | | - Caroline Robert
- INSERM U981, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
- Department of Medical Oncology, Gustave Roussy and Paris-Saclay University, Villejuif, France
| | - Philippe Saiag
- Department of General and Oncologic Dermatology, Ambroise-Paré hospital, APHP, and EA 4340 “Biomarkers in Cancerology and Hemato-oncology,” UVSQ, Université Paris-Saclay, Boulogne-Billancourt, France
| | - Florian Herms
- Service de dermatologie, Hôpital Saint-Louis, Paris, France
| | - Jerome Lambert
- Université de Paris, Hôpital Saint-Louis, Paris, France
- Service de Biostatistique et Information Médicale, Hôpital Saint-Louis, Paris, France
| | | | | | - Lukas Flatz
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
- Department of Dermatology, University Hospital Tübingen, Tübingen, Germany
| | - Nicole Basset-Seguin
- INSERM U976, Hôpital Saint-Louis, Paris, France
- Université de Paris, Hôpital Saint-Louis, Paris, France
- Service de dermatologie, Hôpital Saint-Louis, Paris, France
| | - Sergey I. Nikolaev
- INSERM U981, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
| |
Collapse
|
18
|
Gotzhein F, Aranyossy T, Thielecke L, Sonntag T, Thaden V, Fehse B, Müller I, Glauche I, Cornils K. The Reconstitution Dynamics of Cultivated Hematopoietic Stem Cells and Progenitors Is Independent of Age. Int J Mol Sci 2022; 23:ijms23063160. [PMID: 35328579 PMCID: PMC8948791 DOI: 10.3390/ijms23063160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/08/2022] [Accepted: 03/10/2022] [Indexed: 12/10/2022] Open
Abstract
Hematopoietic stem cell transplantation (HSCT) represents the only curative treatment option for numerous hematologic malignancies. While the influence of donor age and the composition of the graft have already been examined in clinical and preclinical studies, little information is available on the extent to which different hematological subpopulations contribute to the dynamics of the reconstitution process and on whether and how these contributions are altered with age. In a murine model of HSCT, we therefore simultaneously tracked different cultivated and transduced hematopoietic stem and progenitor cell (HSPC) populations using a multicolor-coded barcode system (BC32). We studied a series of age-matched and age-mismatched transplantations and compared the influence of age on the reconstitution dynamics. We show that reconstitution from these cultured and assembled grafts was substantially driven by hematopoietic stem cells (HSCs) and multipotent progenitors (MPPs) independent of age. The reconstitution patterns were polyclonal and stable in all age groups independently of the variability between individual animals, with higher output rates from MPPs than from HSCs. Our experiments suggest that the dynamics of reconstitution and the contribution of cultured and individually transduced HSPC subpopulations are largely independent of age. Our findings support ongoing efforts to expand the application of HSCT in older individuals as a promising strategy to combat hematological diseases, including gene therapy applications.
Collapse
Affiliation(s)
- Frauke Gotzhein
- Clinic of Pediatric Hematology and Oncology, Division of Pediatric Stem Cell Transplantation and Immunology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (F.G.); (V.T.); (I.M.)
- Research Institute Children’s Cancer Center Hamburg, 20251 Hamburg, Germany
| | - Tim Aranyossy
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (T.A.); (T.S.); (B.F.)
| | - Lars Thielecke
- Institute for Medical Informatics and Biometry, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (L.T.); (I.G.)
| | - Tanja Sonntag
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (T.A.); (T.S.); (B.F.)
| | - Vanessa Thaden
- Clinic of Pediatric Hematology and Oncology, Division of Pediatric Stem Cell Transplantation and Immunology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (F.G.); (V.T.); (I.M.)
- Research Institute Children’s Cancer Center Hamburg, 20251 Hamburg, Germany
| | - Boris Fehse
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (T.A.); (T.S.); (B.F.)
| | - Ingo Müller
- Clinic of Pediatric Hematology and Oncology, Division of Pediatric Stem Cell Transplantation and Immunology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (F.G.); (V.T.); (I.M.)
- Research Institute Children’s Cancer Center Hamburg, 20251 Hamburg, Germany
| | - Ingmar Glauche
- Institute for Medical Informatics and Biometry, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (L.T.); (I.G.)
| | - Kerstin Cornils
- Clinic of Pediatric Hematology and Oncology, Division of Pediatric Stem Cell Transplantation and Immunology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (F.G.); (V.T.); (I.M.)
- Research Institute Children’s Cancer Center Hamburg, 20251 Hamburg, Germany
- Correspondence: ; Tel.: +49-40-7410-52721
| |
Collapse
|
19
|
Immunoproteasome Activity in Chronic Lymphocytic Leukemia as a Target of the Immunoproteasome-Selective Inhibitors. Cells 2022; 11:cells11050838. [PMID: 35269460 PMCID: PMC8909520 DOI: 10.3390/cells11050838] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/21/2022] [Accepted: 02/22/2022] [Indexed: 12/21/2022] Open
Abstract
Targeting proteasome with proteasome inhibitors (PIs) is an approved treatment strategy in multiple myeloma that has also been explored pre-clinically and clinically in other hematological malignancies. The approved PIs target both the constitutive and the immunoproteasome, the latter being present predominantly in cells of lymphoid origin. Therapeutic targeting of the immunoproteasome in cells with sole immunoproteasome activity may be selectively cytotoxic in malignant cells, while sparing the non-lymphoid tissues from the on-target PIs toxicity. Using activity-based probes to assess the proteasome activity profile and correlating it with the cytotoxicity assays, we identified B-cell chronic lymphocytic leukemia (B-CLL) to express predominantly immunoproteasome activity, which is associated with high sensitivity to approved proteasome inhibitors and, more importantly, to the immunoproteasome selective inhibitors LU005i and LU035i, targeting all immunoproteasome active subunits or only the immunoproteasome β5i, respectively. At the same time, LU102, a proteasome β2 inhibitor, sensitized B-CLL or immunoproteasome inhibitor-inherently resistant primary cells of acute myeloid leukemia, B-cell acute lymphoblastic leukemia, multiple myeloma and plasma cell leukemia to low doses of LU035i. The immunoproteasome thus represents a novel therapeutic target, which warrants further testing with clinical stage immunoproteasome inhibitors in monotherapy or in combinations.
Collapse
|
20
|
Multicolor strategies for investigating clonal expansion and tissue plasticity. Cell Mol Life Sci 2022; 79:141. [PMID: 35187598 PMCID: PMC8858928 DOI: 10.1007/s00018-021-04077-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 09/27/2021] [Accepted: 10/14/2021] [Indexed: 12/20/2022]
Abstract
Understanding the generation of complexity in living organisms requires the use of lineage tracing tools at a multicellular scale. In this review, we describe the different multicolor strategies focusing on mouse models expressing several fluorescent reporter proteins, generated by classical (MADM, Brainbow and its multiple derivatives) or acute (StarTrack, CLoNe, MAGIC Markers, iOn, viral vectors) transgenesis. After detailing the multi-reporter genetic strategies that serve as a basis for the establishment of these multicolor mouse models, we briefly mention other animal and cellular models (zebrafish, chicken, drosophila, iPSC) that also rely on these constructs. Then, we highlight practical applications of multicolor mouse models to better understand organogenesis at single progenitor scale (clonal analyses) in the brain and briefly in several other tissues (intestine, skin, vascular, hematopoietic and immune systems). In addition, we detail the critical contribution of multicolor fate mapping strategies in apprehending the fine cellular choreography underlying tissue morphogenesis in several models with a particular focus on brain cytoarchitecture in health and diseases. Finally, we present the latest technological advances in multichannel and in-depth imaging, and automated analyses that enable to better exploit the large amount of data generated from multicolored tissues.
Collapse
|
21
|
Innes JA, Lowe AS, Fonseca R, Aley N, El-Hassan T, Constantinou M, Lau J, Eddaoudi A, Marino S, Brandner S. Phenotyping clonal populations of glioma stem cell reveals a high degree of plasticity in response to changes of microenvironment. J Transl Med 2022; 102:172-184. [PMID: 34782726 PMCID: PMC8784315 DOI: 10.1038/s41374-021-00695-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 10/23/2021] [Accepted: 10/25/2021] [Indexed: 11/09/2022] Open
Abstract
The phenotype of glioma-initiating cells (GIC) is modulated by cell-intrinsic and cell-extrinsic factors. Phenotypic heterogeneity and plasticity of GIC is an important limitation to therapeutic approaches targeting cancer stem cells. Plasticity also presents a challenge to the identification, isolation, and propagation of purified cancer stem cells. Here we use a barcode labelling approach of GIC to generate clonal populations over a number of passages, in combination with phenotyping using the established stem cell markers CD133, CD15, CD44, and A2B5. Using two cell lines derived from isocitrate dehydrogenase (IDH)-wildtype glioblastoma, we identify a remarkable heterogeneity of the phenotypes between the cell lines. During passaging, clonal expansion manifests as the emergence of a limited number of barcoded clones and a decrease in the overall number of clones. Dual-labelled GIC are capable of forming traceable clonal populations which emerge after as few as two passages from mixed cultures and through analyses of similarity of relative proportions of 16 surface markers we were able to pinpoint the fate of such populations. By generating tumour organoids we observed a remarkable persistence of dominant clones but also a significant plasticity of stemness marker expression. Our study presents an experimental approach to simultaneously barcode and phenotype glioma-initiating cells to assess their functional properties, for example to screen newly established GIC for tumour-specific therapeutic vulnerabilities.
Collapse
Affiliation(s)
- James A Innes
- Department of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, Queen Square, London, WC1N 3BG, UK
| | - Andrew S Lowe
- Department of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, Queen Square, London, WC1N 3BG, UK
| | - Raquel Fonseca
- Department of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, Queen Square, London, WC1N 3BG, UK
| | - Natasha Aley
- Department of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, Queen Square, London, WC1N 3BG, UK
| | - Tedani El-Hassan
- Department of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, Queen Square, London, WC1N 3BG, UK
| | - Myrianni Constantinou
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, E1 2AT, UK
| | - Joanne Lau
- Department of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, Queen Square, London, WC1N 3BG, UK
| | - Ayad Eddaoudi
- Zayed Centre for Research Into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, University College London, London, WC1N 1DZ, UK
| | - Silvia Marino
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, E1 2AT, UK
| | - Sebastian Brandner
- Department of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, Queen Square, London, WC1N 3BG, UK.
| |
Collapse
|
22
|
Bow AJ, Masi TJ, Dhar MS. Etched 3D-Printed Polycaprolactone Constructs Functionalized with Reduced Graphene Oxide for Enhanced Attachment of Dental Pulp-Derived Stem Cells. Pharmaceutics 2021; 13:2146. [PMID: 34959426 PMCID: PMC8704510 DOI: 10.3390/pharmaceutics13122146] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 12/03/2021] [Accepted: 12/10/2021] [Indexed: 01/08/2023] Open
Abstract
A core challenge in the field of tissue engineering is the ability to establish pipeline workflows for the design and characterization of scaffold technologies with clinically translatable attributes. The parallel development of biomaterials and stem cell populations represents a self-sufficient and streamlined approach for establishing such a pipeline. In the current study, rat dental pulp stem cell (rDPSC) populations were established to assess functionalized polycaprolactone (PCL) constructs. Initial optimization and characterization of rDPSC extraction and culture conditions confirmed that cell populations were readily expandable and demonstrated surface markers associated with multi-potency. Subset populations were transduced to express DsRed fluorescent protein as a mechanism of tracking both cells and cell-derived extracellular matrix content on complex scaffold architecture. Thermoplastic constructs included reduced graphene oxide (rGO) as an additive to promote cellular attachment and were further modified by surface etching a weak acetic acid solution to roughen surface topographical features, which was observed to dramatically improve cell surface coverage in vitro. Based on these data, the modified rGO-functionalized PCL constructs represent a versatile platform for bone tissue engineering, capable of being applied as a standalone matrix or in conjunction with bio-active payloads such as DPSCs or other bio-inks.
Collapse
Affiliation(s)
- Austin J. Bow
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37931, USA;
| | - Thomas J. Masi
- School of Medicine, University of Tennessee Graduate, Knoxville, TN 37920, USA;
| | - Madhu S. Dhar
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37931, USA;
| |
Collapse
|
23
|
Pishas KI, Cowley KJ, Pandey A, Hoang T, Beach JA, Luu J, Vary R, Smith LK, Shembrey CE, Rashoo N, White MO, Simpson KJ, Bild A, Griffiths JI, Cheasley D, Campbell I, Bowtell DDL, Christie EL. Phenotypic Consequences of SLC25A40-ABCB1 Fusions beyond Drug Resistance in High-Grade Serous Ovarian Cancer. Cancers (Basel) 2021; 13:cancers13225644. [PMID: 34830797 PMCID: PMC8616176 DOI: 10.3390/cancers13225644] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/05/2021] [Accepted: 11/05/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Among the plethora of malignancies affecting the female reproductive tract, those concerning the ovary are the most frequently fatal. In particular, chemotherapy-resistant High-Grade Serous Ovarian Cancer (HGSOC) remains a clinically intractable disease with a high rate of mortality. We previously identified SLC25A40-ABCB1 transcriptional fusions as the driving force behind drug resistance in HGSOC. As success in the clinical arena will only be achieved by enhancing our fundamental understanding of the drivers that mediate cellular drug resistance, this report sought to elucidate the phenotypic, metabolomic and transcriptional consequences of SLC25A40-ABCB1 fusions beyond drug resistance. High-throughput FDA drug screening was also undertaken to identify new therapeutic avenues against drug-resistant cellular populations. Abstract Despite high response rates to initial chemotherapy, the majority of women diagnosed with High-Grade Serous Ovarian Cancer (HGSOC) ultimately develop drug resistance within 1–2 years of treatment. We previously identified the most common mechanism of acquired resistance in HGSOC to date, transcriptional fusions involving the ATP-binding cassette (ABC) transporter ABCB1, which has well established roles in multidrug resistance. However, the underlying biology of fusion-positive cells, as well as how clonal interactions between fusion-negative and positive populations influences proliferative fitness and therapeutic response remains unknown. Using a panel of fusion-negative and positive HGSOC single-cell clones, we demonstrate that in addition to mediating drug resistance, ABCB1 fusion-positive cells display impaired proliferative capacity, elevated oxidative metabolism, altered actin cellular morphology and an extracellular matrix/inflammatory enriched transcriptional profile. The co-culture of fusion-negative and positive populations had no effect on cellular proliferation but markedly altered drug sensitivity to doxorubicin, paclitaxel and cisplatin. Finally, high-throughput screening of 2907 FDA-approved compounds revealed 36 agents that induce equal cytotoxicity in both pure and mixed ABCB1 fusion populations. Collectively, our findings have unraveled the underlying biology of ABCB1 fusion-positive cells beyond drug resistance and identified novel therapeutic agents that may significantly improve the prognosis of relapsed HGSOC patients.
Collapse
Affiliation(s)
- Kathleen I. Pishas
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; (K.I.P.); (A.P.); (T.H.); (J.A.B.); (L.K.S.); (C.E.S.); (N.R.); (M.O.W.); (D.C.); (I.C.); (D.D.L.B.)
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3010, Australia;
| | - Karla J. Cowley
- Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; (K.J.C.); (J.L.); (R.V.)
| | - Ahwan Pandey
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; (K.I.P.); (A.P.); (T.H.); (J.A.B.); (L.K.S.); (C.E.S.); (N.R.); (M.O.W.); (D.C.); (I.C.); (D.D.L.B.)
| | - Therese Hoang
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; (K.I.P.); (A.P.); (T.H.); (J.A.B.); (L.K.S.); (C.E.S.); (N.R.); (M.O.W.); (D.C.); (I.C.); (D.D.L.B.)
| | - Jessica A. Beach
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; (K.I.P.); (A.P.); (T.H.); (J.A.B.); (L.K.S.); (C.E.S.); (N.R.); (M.O.W.); (D.C.); (I.C.); (D.D.L.B.)
| | - Jennii Luu
- Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; (K.J.C.); (J.L.); (R.V.)
| | - Robert Vary
- Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; (K.J.C.); (J.L.); (R.V.)
| | - Lorey K. Smith
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; (K.I.P.); (A.P.); (T.H.); (J.A.B.); (L.K.S.); (C.E.S.); (N.R.); (M.O.W.); (D.C.); (I.C.); (D.D.L.B.)
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3010, Australia;
| | - Carolyn E. Shembrey
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; (K.I.P.); (A.P.); (T.H.); (J.A.B.); (L.K.S.); (C.E.S.); (N.R.); (M.O.W.); (D.C.); (I.C.); (D.D.L.B.)
- Department of Clinical Pathology, Faculty of Medicine, Dentistry, and Health Science, The University of Melbourne, Melbourne, VIC 3000, Australia
| | - Nineveh Rashoo
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; (K.I.P.); (A.P.); (T.H.); (J.A.B.); (L.K.S.); (C.E.S.); (N.R.); (M.O.W.); (D.C.); (I.C.); (D.D.L.B.)
| | - Madelynne O. White
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; (K.I.P.); (A.P.); (T.H.); (J.A.B.); (L.K.S.); (C.E.S.); (N.R.); (M.O.W.); (D.C.); (I.C.); (D.D.L.B.)
| | - Kaylene J. Simpson
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3010, Australia;
- Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; (K.J.C.); (J.L.); (R.V.)
| | - Andrea Bild
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA 91010, USA; (A.B.); (J.I.G.)
| | - Jason I. Griffiths
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA 91010, USA; (A.B.); (J.I.G.)
| | - Dane Cheasley
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; (K.I.P.); (A.P.); (T.H.); (J.A.B.); (L.K.S.); (C.E.S.); (N.R.); (M.O.W.); (D.C.); (I.C.); (D.D.L.B.)
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3010, Australia;
| | - Ian Campbell
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; (K.I.P.); (A.P.); (T.H.); (J.A.B.); (L.K.S.); (C.E.S.); (N.R.); (M.O.W.); (D.C.); (I.C.); (D.D.L.B.)
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3010, Australia;
| | - David D. L. Bowtell
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; (K.I.P.); (A.P.); (T.H.); (J.A.B.); (L.K.S.); (C.E.S.); (N.R.); (M.O.W.); (D.C.); (I.C.); (D.D.L.B.)
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3010, Australia;
| | - Elizabeth L. Christie
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; (K.I.P.); (A.P.); (T.H.); (J.A.B.); (L.K.S.); (C.E.S.); (N.R.); (M.O.W.); (D.C.); (I.C.); (D.D.L.B.)
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3010, Australia;
- Correspondence:
| |
Collapse
|
24
|
Głów D, Meyer S, García Roldán I, Akingunsade LM, Riecken K, Fehse B. LATE-a novel sensitive cell-based assay for the study of CRISPR/Cas9-related long-term adverse treatment effects. Mol Ther Methods Clin Dev 2021; 22:249-262. [PMID: 34485609 PMCID: PMC8399046 DOI: 10.1016/j.omtm.2021.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 07/23/2021] [Indexed: 11/26/2022]
Abstract
Since the introduction of clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9), genome editing has been broadly applied in basic research and applied biotechnology, whereas translation into clinical testing has raised safety concerns. Indeed, although frequencies and locations of off-target events have been widely addressed, little is known about their potential biological consequences in large-scale long-term settings. We have developed a long-term adverse treatment effect (LATE) in vitro assay that addresses potential toxicity of designer nucleases by assessing cell transformation events. In small-scale proof-of-principle experiments we reproducibly detected low-frequency (<0.5%) growth-promoting events in primary human newborn foreskin fibroblasts (NUFF cells) resulting from off-target cleavage in the TP53 gene. Importantly, the LATE assay detected not only off-target effects in TP53 not predicted by popular online tools but also growth-promoting mutations in other tumor suppressor genes, such as p21 and PLZF. It convincingly verified strongly reduced off-target activities of high fidelity compared with first-generation Cas9. Finally, the LATE assay was readily adapted to other cell types, namely clinically relevant human mesenchymal stromal cells (hMSCs) and retinal pigmented epithelial (RPE-1) cells. In conclusion, the LATE assay allows assessment of physiological adverse effects of the CRISPR/Cas system and might therefore be useful for preclinical safety studies.
Collapse
Affiliation(s)
- Dawid Głów
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Centre Hamburg-Eppendorf (UKE), Martinistr. 52, Hamburg 20246, Germany
| | - Simon Meyer
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Centre Hamburg-Eppendorf (UKE), Martinistr. 52, Hamburg 20246, Germany
| | - Irene García Roldán
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Centre Hamburg-Eppendorf (UKE), Martinistr. 52, Hamburg 20246, Germany
| | - Lara Marie Akingunsade
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Centre Hamburg-Eppendorf (UKE), Martinistr. 52, Hamburg 20246, Germany
| | - Kristoffer Riecken
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Centre Hamburg-Eppendorf (UKE), Martinistr. 52, Hamburg 20246, Germany
| | - Boris Fehse
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Centre Hamburg-Eppendorf (UKE), Martinistr. 52, Hamburg 20246, Germany
| |
Collapse
|
25
|
Ring SS, Cupovic J, Onder L, Lütge M, Perez-Shibayama C, Gil-Cruz C, Scandella E, De Martin A, Mörbe U, Hartmann F, Wenger R, Spiegl M, Besse A, Bonilla WV, Stemeseder F, Schmidt S, Orlinger KK, Krebs P, Ludewig B, Flatz L. Viral vector-mediated reprogramming of the fibroblastic tumor stroma sustains curative melanoma treatment. Nat Commun 2021; 12:4734. [PMID: 34354077 PMCID: PMC8342618 DOI: 10.1038/s41467-021-25057-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 07/19/2021] [Indexed: 11/13/2022] Open
Abstract
The tumor microenvironment (TME) is a complex amalgam of tumor cells, immune cells, endothelial cells and fibroblastic stromal cells (FSC). Cancer-associated fibroblasts are generally seen as tumor-promoting entity. However, it is conceivable that particular FSC populations within the TME contribute to immune-mediated tumor control. Here, we show that intratumoral treatment of mice with a recombinant lymphocytic choriomeningitis virus-based vaccine vector expressing a melanocyte differentiation antigen resulted in T cell-dependent long-term control of melanomas. Using single-cell RNA-seq analysis, we demonstrate that viral vector-mediated transduction reprogrammed and activated a Cxcl13-expressing FSC subset that show a pronounced immunostimulatory signature and increased expression of the inflammatory cytokine IL-33. Ablation of Il33 gene expression in Cxcl13-Cre-positive FSCs reduces the functionality of intratumoral T cells and unleashes tumor growth. Thus, reprogramming of FSCs by a self-antigen-expressing viral vector in the TME is critical for curative melanoma treatment by locally sustaining the activity of tumor-specific T cells. Lymphocytic choriomeningitis virus (LCMV)-based viral vectors have been shown to induce potent antitumor immune responses. Here the authors show that a LCMV-based vaccine vector remodels the tumor-associated fibroblastic stroma, sustaining CD8+ T cell activation and reducing tumor growth in a preclinical model of melanoma.
Collapse
Affiliation(s)
- Sandra S Ring
- Institute of Immunobiology, Kantonsspital St.Gallen, St.Gallen, Switzerland
| | - Jovana Cupovic
- Institute of Immunobiology, Kantonsspital St.Gallen, St.Gallen, Switzerland.,Max Planck Institute of Immunology and Epigenetics, Freiburg, Germany
| | - Lucas Onder
- Institute of Immunobiology, Kantonsspital St.Gallen, St.Gallen, Switzerland
| | - Mechthild Lütge
- Institute of Immunobiology, Kantonsspital St.Gallen, St.Gallen, Switzerland
| | | | - Cristina Gil-Cruz
- Institute of Immunobiology, Kantonsspital St.Gallen, St.Gallen, Switzerland
| | - Elke Scandella
- Institute of Immunobiology, Kantonsspital St.Gallen, St.Gallen, Switzerland
| | - Angelina De Martin
- Institute of Immunobiology, Kantonsspital St.Gallen, St.Gallen, Switzerland
| | - Urs Mörbe
- Institute of Immunobiology, Kantonsspital St.Gallen, St.Gallen, Switzerland
| | - Fabienne Hartmann
- Institute of Immunobiology, Kantonsspital St.Gallen, St.Gallen, Switzerland
| | - Robert Wenger
- Department of Plastic Reconstructive Surgery, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Matthias Spiegl
- Department of Plastic Reconstructive Surgery, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Andrej Besse
- Department of Medical Oncology and Hematology, Kantonsspital St.Gallen, St.Gallen, Switzerland
| | - Weldy V Bonilla
- Division of Experimental Virology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | | | | | | | - Philippe Krebs
- Institute of Pathology, University of Berne, Berne, Switzerland
| | - Burkhard Ludewig
- Institute of Immunobiology, Kantonsspital St.Gallen, St.Gallen, Switzerland. .,Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland.
| | - Lukas Flatz
- Institute of Immunobiology, Kantonsspital St.Gallen, St.Gallen, Switzerland. .,Department of Dermatology, Kantonsspital St. Gallen, St. Gallen, Switzerland. .,Department of Dermatology, University Hospital Zurich, Zurich, Switzerland.
| |
Collapse
|
26
|
Lewis SM, Asselin-Labat ML, Nguyen Q, Berthelet J, Tan X, Wimmer VC, Merino D, Rogers KL, Naik SH. Spatial omics and multiplexed imaging to explore cancer biology. Nat Methods 2021; 18:997-1012. [PMID: 34341583 DOI: 10.1038/s41592-021-01203-6] [Citation(s) in RCA: 298] [Impact Index Per Article: 74.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 06/04/2021] [Indexed: 01/19/2023]
Abstract
Understanding intratumoral heterogeneity-the molecular variation among cells within a tumor-promises to address outstanding questions in cancer biology and improve the diagnosis and treatment of specific cancer subtypes. Single-cell analyses, especially RNA sequencing and other genomics modalities, have been transformative in revealing novel biomarkers and molecular regulators associated with tumor growth, metastasis and drug resistance. However, these approaches fail to provide a complete picture of tumor biology, as information on cellular location within the tumor microenvironment is lost. New technologies leveraging multiplexed fluorescence, DNA, RNA and isotope labeling enable the detection of tens to thousands of cancer subclones or molecular biomarkers within their native spatial context. The expeditious growth in these techniques, along with methods for multiomics data integration, promises to yield a more comprehensive understanding of cell-to-cell variation within and between individual tumors. Here we provide the current state and future perspectives on the spatial technologies expected to drive the next generation of research and diagnostic and therapeutic strategies for cancer.
Collapse
Affiliation(s)
- Sabrina M Lewis
- Advanced Technology and Biology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Victoria, Australia
| | - Marie-Liesse Asselin-Labat
- Department of Medical Biology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Victoria, Australia.,Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Quan Nguyen
- Division of Genetics and Genomics, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Jean Berthelet
- Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia.,School of Cancer Medicine, La Trobe University, Bundoora, Victoria, Australia
| | - Xiao Tan
- Division of Genetics and Genomics, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Verena C Wimmer
- Advanced Technology and Biology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Victoria, Australia
| | - Delphine Merino
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Victoria, Australia.,Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia.,School of Cancer Medicine, La Trobe University, Bundoora, Victoria, Australia
| | - Kelly L Rogers
- Advanced Technology and Biology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia. .,Department of Medical Biology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Victoria, Australia.
| | - Shalin H Naik
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia. .,Department of Medical Biology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Victoria, Australia.
| |
Collapse
|
27
|
Berthelet J, Wimmer VC, Whitfield HJ, Serrano A, Boudier T, Mangiola S, Merdas M, El-Saafin F, Baloyan D, Wilcox J, Wilcox S, Parslow AC, Papenfuss AT, Yeo B, Ernst M, Pal B, Anderson RL, Davis MJ, Rogers KL, Hollande F, Merino D. The site of breast cancer metastases dictates their clonal composition and reversible transcriptomic profile. SCIENCE ADVANCES 2021; 7:eabf4408. [PMID: 34233875 PMCID: PMC8262813 DOI: 10.1126/sciadv.abf4408] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 05/25/2021] [Indexed: 05/03/2023]
Abstract
Intratumoral heterogeneity is a driver of breast cancer progression, but the nature of the clonal interactive network involved in this process remains unclear. Here, we optimized the use of optical barcoding to visualize and characterize 31 cancer subclones in vivo. By mapping the clonal composition of thousands of metastases in two clinically relevant sites, the lungs and liver, we found that metastases were highly polyclonal in lungs but not in the liver. Furthermore, the transcriptome of the subclones varied according to their metastatic niche. We also identified a reversible niche-driven signature that was conserved in lung and liver metastases collected during patient autopsies. Among this signature, we found that the tumor necrosis factor-α pathway was up-regulated in lung compared to liver metastases, and inhibition of this pathway affected metastasis diversity. These results highlight that the cellular and molecular heterogeneity observed in metastases is largely dictated by the tumor microenvironment.
Collapse
Affiliation(s)
- Jean Berthelet
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia.
- School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
| | - Verena C Wimmer
- Advanced Technology and Biology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, Faculty of Medicine, Dentistry, and Health Science, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Holly J Whitfield
- Department of Medical Biology, Faculty of Medicine, Dentistry, and Health Science, The University of Melbourne, Parkville, VIC 3010, Australia
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Antonin Serrano
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia
- Department of Medical Biology, Faculty of Medicine, Dentistry, and Health Science, The University of Melbourne, Parkville, VIC 3010, Australia
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Thomas Boudier
- Advanced Technology and Biology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, Faculty of Medicine, Dentistry, and Health Science, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Stefano Mangiola
- Department of Medical Biology, Faculty of Medicine, Dentistry, and Health Science, The University of Melbourne, Parkville, VIC 3010, Australia
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Michal Merdas
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
| | - Farrah El-Saafin
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
| | - David Baloyan
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
| | - Jordan Wilcox
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
| | - Steven Wilcox
- Advanced Technology and Biology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, Faculty of Medicine, Dentistry, and Health Science, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Adam C Parslow
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
| | - Anthony T Papenfuss
- Department of Medical Biology, Faculty of Medicine, Dentistry, and Health Science, The University of Melbourne, Parkville, VIC 3010, Australia
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3000, Australia
| | - Belinda Yeo
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
- Austin Health, Heidelberg, VIC 3084, Australia
| | - Matthias Ernst
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
| | - Bhupinder Pal
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
| | - Robin L Anderson
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3000, Australia
| | - Melissa J Davis
- Department of Medical Biology, Faculty of Medicine, Dentistry, and Health Science, The University of Melbourne, Parkville, VIC 3010, Australia
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Clinical Pathology, Faculty of Medicine, Dentistry, and Health Science, The University of Melbourne, Melbourne, VIC 3000, Australia
| | - Kelly L Rogers
- Advanced Technology and Biology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, Faculty of Medicine, Dentistry, and Health Science, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Frédéric Hollande
- Department of Clinical Pathology, Faculty of Medicine, Dentistry, and Health Science, The University of Melbourne, Melbourne, VIC 3000, Australia
- University of Melbourne Centre for Cancer Research, Victorian Comprehensive Cancer Centre, Melbourne, VIC 3000, Australia
| | - Delphine Merino
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia.
- School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
- Department of Medical Biology, Faculty of Medicine, Dentistry, and Health Science, The University of Melbourne, Parkville, VIC 3010, Australia
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| |
Collapse
|
28
|
Nikulin S, Zakharova G, Poloznikov A, Raigorodskaya M, Wicklein D, Schumacher U, Nersisyan S, Bergquist J, Bakalkin G, Astakhova L, Tonevitsky A. Effect of the Expression of ELOVL5 and IGFBP6 Genes on the Metastatic Potential of Breast Cancer Cells. Front Genet 2021; 12:662843. [PMID: 34149804 PMCID: PMC8206645 DOI: 10.3389/fgene.2021.662843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 04/20/2021] [Indexed: 12/09/2022] Open
Abstract
Breast cancer (BC) is the leading cause of death from malignant neoplasms among women worldwide, and metastatic BC presents the biggest problems for treatment. Previously, it was shown that lower expression of ELOVL5 and IGFBP6 genes is associated with a higher risk of the formation of distant metastases in BC. In this work, we studied the change in phenotypical traits, as well as in the transcriptomic and proteomic profiles of BC cells as a result of the stable knockdown of ELOVL5 and IGFBP6 genes. The knockdown of ELOVL5 and IGFBP6 genes was found to lead to a strong increase in the expression of the matrix metalloproteinase (MMP) MMP1. These results were in good agreement with the correlation analysis of gene expression in tumor samples from patients and were additionally confirmed by zymography. The knockdown of ELOVL5 and IGFBP6 genes was also discovered to change the expression of a group of genes involved in the formation of intercellular contacts. In particular, the expression of the CDH11 gene was markedly reduced, which also complies with the correlation analysis. The spheroid formation assay showed that intercellular adhesion decreased as a result of the knockdown of the ELOVL5 and IGFBP6 genes. Thus, the obtained data indicate that malignant breast tumors with reduced expression of the ELOVL5 and IGFBP6 genes can metastasize with a higher probability due to a more efficient invasion of tumor cells.
Collapse
Affiliation(s)
- Sergey Nikulin
- Faculty of Biology and Biotechnologies, National Research University Higher School of Economics, Moscow, Russia
| | | | - Andrey Poloznikov
- Faculty of Biology and Biotechnologies, National Research University Higher School of Economics, Moscow, Russia
- School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
| | - Maria Raigorodskaya
- Faculty of Biology and Biotechnologies, National Research University Higher School of Economics, Moscow, Russia
- Scientific Research Centre Bioclinicum, Moscow, Russia
| | - Daniel Wicklein
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Udo Schumacher
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stepan Nersisyan
- Faculty of Biology and Biotechnologies, National Research University Higher School of Economics, Moscow, Russia
| | - Jonas Bergquist
- Department of Chemistry – BMC, Uppsala University, Uppsala, Sweden
| | - Georgy Bakalkin
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Lidiia Astakhova
- Scientific Research Centre Bioclinicum, Moscow, Russia
- School of Life Sciences, Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - Alexander Tonevitsky
- Faculty of Biology and Biotechnologies, National Research University Higher School of Economics, Moscow, Russia
- Laboratory of Microfluidic Technologies for Biomedicine, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Moscow, Russia
| |
Collapse
|
29
|
Gonzalez Rajal A, Marzec KA, McCloy RA, Nobis M, Chin V, Hastings JF, Lai K, Kennerson M, Hughes WE, Vaghjiani V, Timpson P, Cain JE, Watkins DN, Croucher DR, Burgess A. A non-genetic, cell cycle-dependent mechanism of platinum resistance in lung adenocarcinoma. eLife 2021; 10:65234. [PMID: 33983115 PMCID: PMC8169122 DOI: 10.7554/elife.65234] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 05/11/2021] [Indexed: 12/15/2022] Open
Abstract
We previously used a pulse-based in vitro assay to unveil targetable signalling pathways associated with innate cisplatin resistance in lung adenocarcinoma (Hastings et al., 2020). Here, we advanced this model system and identified a non-genetic mechanism of resistance that drives recovery and regrowth in a subset of cells. Using RNAseq and a suite of biosensors to track single-cell fates both in vitro and in vivo, we identified that early S phase cells have a greater ability to maintain proliferative capacity, which correlated with reduced DNA damage over multiple generations. In contrast, cells in G1, late S or those treated with PARP/RAD51 inhibitors, maintained higher levels of DNA damage and underwent prolonged S/G2 phase arrest and senescence. Combined with our previous work, these data indicate that there is a non-genetic mechanism of resistance in human lung adenocarcinoma that is dependent on the cell cycle stage at the time of cisplatin exposure.
Collapse
Affiliation(s)
- Alvaro Gonzalez Rajal
- ANZAC Research Institute, Concord Hospital, Concord, Australia.,Garvan Institute of Medical Research, Sydney, Australia.,St Vincent's Hospital Clinical School, University of New South Wales, Sydney, Australia
| | - Kamila A Marzec
- ANZAC Research Institute, Concord Hospital, Concord, Australia
| | - Rachael A McCloy
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, Australia
| | - Max Nobis
- St Vincent's Hospital Clinical School, University of New South Wales, Sydney, Australia.,The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, Australia
| | - Venessa Chin
- St Vincent's Hospital Clinical School, University of New South Wales, Sydney, Australia.,The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, Australia.,St Vincent's Hospital Sydney, Darlinghurst, Australia
| | - Jordan F Hastings
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, Australia
| | - Kaitao Lai
- ANZAC Research Institute, Concord Hospital, Concord, Australia.,The University of Sydney Concord Clinical School, Faculty of Medicine and Health, Sydney, Australia
| | - Marina Kennerson
- ANZAC Research Institute, Concord Hospital, Concord, Australia.,The University of Sydney Concord Clinical School, Faculty of Medicine and Health, Sydney, Australia
| | - William E Hughes
- Garvan Institute of Medical Research, Sydney, Australia.,St Vincent's Hospital Clinical School, University of New South Wales, Sydney, Australia.,Children's Medical Research Institute, The University of Sydney, Westmead, Australia
| | | | - Paul Timpson
- St Vincent's Hospital Clinical School, University of New South Wales, Sydney, Australia.,The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, Australia
| | - Jason E Cain
- Hudson Institute of Medical Research, Clayton, Australia.,Department of Molecular and Translational Medicine, School of Medicine, Nursing and Health Sciences, Monash University, Clayton, Australia
| | - D Neil Watkins
- Research Institute in Oncology and Hematology, CancerCare Manitoba, Winnipeg, Canada.,Department of Internal Medicine, Rady Faculty of Health Science, University of Manitoba, Winnipeg, Canada
| | - David R Croucher
- St Vincent's Hospital Clinical School, University of New South Wales, Sydney, Australia.,The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, Australia
| | - Andrew Burgess
- ANZAC Research Institute, Concord Hospital, Concord, Australia
| |
Collapse
|
30
|
Gambera S, Patiño-Garcia A, Alfranca A, Garcia-Castro J. RGB-Marking to Identify Patterns of Selection and Neutral Evolution in Human Osteosarcoma Models. Cancers (Basel) 2021; 13:2003. [PMID: 33919355 PMCID: PMC8122697 DOI: 10.3390/cancers13092003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/05/2021] [Accepted: 04/19/2021] [Indexed: 12/29/2022] Open
Abstract
Osteosarcoma (OS) is a highly aggressive tumor characterized by malignant cells producing pathologic bone; the disease presents a natural tendency to metastasize. Genetic studies indicate that the OS genome is extremely complex, presenting signs of macro-evolution, and linear and branched patterns of clonal development. However, those studies were based on the phylogenetic reconstruction of next-generation sequencing (NGS) data, which present important limitations. Thus, testing clonal evolution in experimental models could be useful for validating this hypothesis. In the present study, lentiviral LeGO-vectors were employed to generate colorimetric red, green, blue (RGB)-marking in murine, canine, and human OS. With this strategy, we studied tumor heterogeneity and the clonal dynamics occurring in vivo in immunodeficient NOD.Cg-Prkdcscid-Il2rgtm1Wjl/SzJ (NSG) mice. Based on colorimetric label, tumor clonal composition was analyzed by confocal microscopy, flow cytometry, and different types of supervised and unsupervised clonal analyses. With this approach, we observed a consistent reduction in the clonal composition of RGB-marked tumors and identified evident clonal selection at the first passage in immunodeficient mice. Furthermore, we also demonstrated that OS could follow a neutral model of growth, where the disease is defined by the coexistence of different tumor sub-clones. Our study demonstrates the importance of rigorous testing of the selective forces in commonly used experimental models.
Collapse
Affiliation(s)
- Stefano Gambera
- Cellular Biotechnology Unit, Instituto de Salud Carlos III, 28220 Madrid, Spain; (S.G.); (A.A.)
| | - Ana Patiño-Garcia
- Department of Pediatrics, Laboratory of Advanced Therapies for Pediatric Solid Tumors, Solid Tumor Area, CIMA and Instituto de Investigación Sanitaria de Navarra, University Clinic of Navarra, IdiSNA, 31008 Pamplona, Spain;
| | - Arantzazu Alfranca
- Cellular Biotechnology Unit, Instituto de Salud Carlos III, 28220 Madrid, Spain; (S.G.); (A.A.)
- Immunology Department, Hospital Universitario de La Princesa, 28006 Madrid, Spain
| | - Javier Garcia-Castro
- Cellular Biotechnology Unit, Instituto de Salud Carlos III, 28220 Madrid, Spain; (S.G.); (A.A.)
| |
Collapse
|
31
|
Maire CL, Mohme M, Bockmayr M, Fita KD, Riecken K, Börnigen D, Alawi M, Failla A, Kolbe K, Zapf S, Holz M, Neumann K, Dührsen L, Lange T, Fehse B, Westphal M, Lamszus K. Glioma escape signature and clonal development under immune pressure. J Clin Invest 2021; 130:5257-5271. [PMID: 32603315 DOI: 10.1172/jci138760] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 06/24/2020] [Indexed: 12/21/2022] Open
Abstract
Immunotherapeutic strategies are increasingly important in neuro-oncology, and the elucidation of escape mechanisms that lead to treatment resistance is crucial. We investigated the impact of immune pressure on the clonal dynamics and immune escape signature by comparing glioma growth in immunocompetent versus immunodeficient mice. Glioma-bearing WT and Pd-1-/- mice survived significantly longer than immunodeficient Pfp-/- Rag2-/- mice. While tumors in Pfp-/- Rag2-/- mice were highly polyclonal, immunoedited tumors in WT and Pd-1-/- mice displayed reduced clonality with emergence of immune escape clones. Tumor cells in WT mice were distinguished by an IFN-γ-mediated response signature with upregulation of genes involved in immunosuppression. Tumor-infiltrating stromal cells, which include macrophages/microglia, contributed even more strongly to the immunosuppressive signature than the actual tumor cells. The identified murine immune escape signature was reflected in human patients and correlated with poor survival. In conclusion, immune pressure profoundly shapes the clonal composition and gene regulation in malignant gliomas.
Collapse
Affiliation(s)
| | | | - Michael Bockmayr
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Institute of Pathology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt University Berlin and Berlin Institute of Health, Berlin, Germany
| | | | - Kristoffer Riecken
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation
| | | | | | | | | | | | | | | | | | - Tobias Lange
- Institutes of Anatomy, Experimental Morphology and Pathology, University Cancer Center Hamburg, Hamburg, Germany
| | - Boris Fehse
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation
| | | | | |
Collapse
|
32
|
Liang Y, Walczak P. Long term intravital single cell tracking under multiphoton microscopy. J Neurosci Methods 2020; 349:109042. [PMID: 33340557 DOI: 10.1016/j.jneumeth.2020.109042] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/07/2020] [Accepted: 12/11/2020] [Indexed: 12/13/2022]
Abstract
Visualizing and tracking cells over time in a living organism has been a much-coveted dream before the invention of intravital microscopy. The opaque nature of tissue was a major hurdle that was remedied by the multiphoton microscopy. With the advancement of optical imaging and fluorescent labeling tools, intravital high resolution imaging has become increasingly accessible over the past few years. Long-term intravital tracking of single cells (LIST) under multiphoton microscopy provides a unique opportunity to gain insight into the longitudinal changes in the morphology, migration, or function of cells or subcellular structures. It is particularly suitable for studying slow-evolving cellular and molecular events during normal development or disease progression, without losing the opportunity of catching fast events such as calcium signals. Here, we review the application of LIST under 2-photon microscopy in various fields of neurobiology and discuss challenges and new directions in labeling and imaging methods for LIST. Overall, this review provides an overview of current applications of LIST in mammals, which is an emerging field that will contribute to a better understanding of essential molecular and cellular events in health and disease.
Collapse
Affiliation(s)
- Yajie Liang
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Piotr Walczak
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
33
|
The art of lineage tracing: From worm to human. Prog Neurobiol 2020; 199:101966. [PMID: 33249090 DOI: 10.1016/j.pneurobio.2020.101966] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 11/03/2020] [Accepted: 11/22/2020] [Indexed: 12/20/2022]
Abstract
Reconstructing the genealogy of every cell that makes up an organism remains a long-standing challenge in developmental biology. Besides its relevance for understanding the mechanisms underlying normal and pathological development, resolving the lineage origin of cell types will be crucial to create these types on-demand. Multiple strategies have been deployed towards the problem of lineage tracing, ranging from direct observation to sophisticated genetic approaches. Here we discuss the achievements and limitations of past and current technology. Finally, we speculate about the future of lineage tracing and how to reach the next milestones in the field.
Collapse
|
34
|
A mouse model for peritoneal metastases of colorectal origin recapitulates patient heterogeneity. J Transl Med 2020; 100:1465-1474. [PMID: 32504005 DOI: 10.1038/s41374-020-0448-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 05/13/2020] [Accepted: 05/20/2020] [Indexed: 12/24/2022] Open
Abstract
The peritoneum is a common site of dissemination in patients with colorectal cancer. In order to identify high-risk patients and improve therapeutic strategies, a better understanding of the peritoneal dissemination process and the reasons behind the high heterogeneity that is observed between patients is required. We aimed to create a murine model to further elucidate the process of peritoneal dissemination and to provide an experimental platform for further studies. We developed an in vivo model to assess patterns of peritoneal dissemination of 15 colorectal cancer cell lines. Immune deficient mice were intraperitoneally injected with 10,000 human colorectal cancer cells. Ten weeks after injection, or earlier in case of severe discomfort, the mice were sacrificed followed by dissection including assessment of the outgrowth and localization of peritoneal metastases. Furthermore, using a color-based clonal tracing method, the clonal dynamics of peritoneal nodules were observed. The different cell lines showed great variation in the extent of peritoneal outgrowth, ranging from no outgrowth to localized or widespread outgrowth of cells. An association between KRAS pathway activation and the formation of peritoneal metastases was identified. Also, cell line specific tumor location preferences were observed, with similar patterns of outgrowth in anatomically related areas. Furthermore, different patterns regarding clonal dynamics were found, varying from monoclonal or polyclonal outgrowth to extensively dispersed polyclonal lesions. The established murine model recapitulates heterogeneity as observed in human peritoneal metastases, which makes it a suitable platform for future (intervention) studies.
Collapse
|
35
|
Pericoli G, Petrini S, Giorda E, Ferretti R, Ajmone-Cat MA, Court W, Conti LA, De Simone R, Bencivenga P, Palma A, Di Giannatale A, Jones C, Carai A, Mastronuzzi A, de Billy E, Locatelli F, Vinci M. Integration of Multiple Platforms for the Analysis of Multifluorescent Marking Technology Applied to Pediatric GBM and DIPG. Int J Mol Sci 2020; 21:E6763. [PMID: 32942636 PMCID: PMC7555235 DOI: 10.3390/ijms21186763] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/10/2020] [Accepted: 09/13/2020] [Indexed: 12/15/2022] Open
Abstract
The intratumor heterogeneity represents one of the most difficult challenges for the development of effective therapies to treat pediatric glioblastoma (pGBM) and diffuse intrinsic pontine glioma (DIPG). These brain tumors are composed of heterogeneous cell subpopulations that coexist and cooperate to build a functional network responsible for their aggressive phenotype. Understanding the cellular and molecular mechanisms sustaining such network will be crucial for the identification of new therapeutic strategies. To study more in-depth these mechanisms, we sought to apply the Multifluorescent Marking Technology. We generated multifluorescent pGBM and DIPG bulk cell lines randomly expressing six different fluorescent proteins and from which we derived stable optical barcoded single cell-derived clones. In this study, we focused on the application of the Multifluorescent Marking Technology in 2D and 3D in vitro/ex vivo culture systems. We discuss how we integrated different multimodal fluorescence analysis platforms, identifying their strengths and limitations, to establish the tools that will enable further studies on the intratumor heterogeneity and interclonal interactions in pGBM and DIPG.
Collapse
Affiliation(s)
- Giulia Pericoli
- Department of Onco-Hematology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital—IRCCS, 00146 Rome, Italy; (G.P.); (R.F.); (A.D.G.); (A.M.); (E.d.B.); (F.L.)
| | - Stefania Petrini
- Confocal Microscopy Core Facility, Research Center, Bambino Gesù Children’s Hospital—IRCCS, 00146 Rome, Italy; (S.P.); (L.A.C.)
| | - Ezio Giorda
- FACs Core Facility, Bambino Gesù Children’s Hospital—IRCCS, 00146 Rome, Italy;
| | - Roberta Ferretti
- Department of Onco-Hematology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital—IRCCS, 00146 Rome, Italy; (G.P.); (R.F.); (A.D.G.); (A.M.); (E.d.B.); (F.L.)
| | - Maria Antonietta Ajmone-Cat
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.A.A.-C.); (R.D.S.)
| | - Will Court
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, Sutton SM2 5NG, UK;
| | - Libenzio Adrian Conti
- Confocal Microscopy Core Facility, Research Center, Bambino Gesù Children’s Hospital—IRCCS, 00146 Rome, Italy; (S.P.); (L.A.C.)
| | - Roberta De Simone
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.A.A.-C.); (R.D.S.)
| | - Paola Bencivenga
- Research Laboratories, Bambino Gesù Children’s Hospital—IRCCS, 00146 Rome, Italy; (P.B.); (A.P.)
| | - Alessia Palma
- Research Laboratories, Bambino Gesù Children’s Hospital—IRCCS, 00146 Rome, Italy; (P.B.); (A.P.)
| | - Angela Di Giannatale
- Department of Onco-Hematology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital—IRCCS, 00146 Rome, Italy; (G.P.); (R.F.); (A.D.G.); (A.M.); (E.d.B.); (F.L.)
| | - Chris Jones
- Division of Molecular Pathology, Institute of Cancer Research, Sutton SM2 5NG, UK;
| | - Andrea Carai
- Department of Neuroscience and Neurorehabilitation, Bambino Gesù Children’s Hospital—IRCCS, 00165 Rome, Italy;
| | - Angela Mastronuzzi
- Department of Onco-Hematology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital—IRCCS, 00146 Rome, Italy; (G.P.); (R.F.); (A.D.G.); (A.M.); (E.d.B.); (F.L.)
| | - Emmanuel de Billy
- Department of Onco-Hematology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital—IRCCS, 00146 Rome, Italy; (G.P.); (R.F.); (A.D.G.); (A.M.); (E.d.B.); (F.L.)
| | - Franco Locatelli
- Department of Onco-Hematology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital—IRCCS, 00146 Rome, Italy; (G.P.); (R.F.); (A.D.G.); (A.M.); (E.d.B.); (F.L.)
| | - Maria Vinci
- Department of Onco-Hematology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital—IRCCS, 00146 Rome, Italy; (G.P.); (R.F.); (A.D.G.); (A.M.); (E.d.B.); (F.L.)
| |
Collapse
|
36
|
Bramlett C, Jiang D, Nogalska A, Eerdeng J, Contreras J, Lu R. Clonal tracking using embedded viral barcoding and high-throughput sequencing. Nat Protoc 2020; 15:1436-1458. [PMID: 32132718 PMCID: PMC7427513 DOI: 10.1038/s41596-019-0290-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 12/21/2019] [Indexed: 11/09/2022]
Abstract
Embedded viral barcoding in combination with high-throughput sequencing is a powerful technology with which to track single-cell clones. It can provide clonal-level insights into cellular proliferation, development, differentiation, migration, and treatment efficacy. Here, we present a detailed protocol for a viral barcoding procedure that includes the creation of barcode libraries, the viral delivery of barcodes, the recovery of barcodes, and the computational analysis of barcode sequencing data. The entire procedure can be completed within a few weeks. This barcoding method requires cells to be susceptible to viral transduction. It provides high sensitivity and throughput, and enables precise quantification of cellular progeny. It is cost efficient and does not require any advanced skills. It can also be easily adapted to many types of applications, including both in vitro and in vivo experiments.
Collapse
Affiliation(s)
- Charles Bramlett
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, Los Angeles, CA, USA
| | - Du Jiang
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, Los Angeles, CA, USA
| | - Anna Nogalska
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, Los Angeles, CA, USA
| | - Jiya Eerdeng
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, Los Angeles, CA, USA
| | - Jorge Contreras
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, Los Angeles, CA, USA
| | - Rong Lu
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
37
|
Genetic barcoding reveals clonal dominance in iPSC-derived mesenchymal stromal cells. Stem Cell Res Ther 2020; 11:105. [PMID: 32138773 PMCID: PMC7059393 DOI: 10.1186/s13287-020-01619-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 02/10/2020] [Accepted: 02/24/2020] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND The use of mesenchymal stromal cells (MSCs) for research and clinical application is hampered by cellular heterogeneity and replicative senescence. Generation of MSC-like cells from induced pluripotent stem cells (iPSCs) may circumvent these limitations, and such iPSC-derived MSCs (iMSCs) are already tested in clinical trials. So far, a comparison of MSCs and iMSCs was particularly addressed in bulk culture. Despite the high hopes in cellular therapy, only little is known how the composition of different subclones changes in these cell preparations during culture expansion. METHODS In this study, we used multicolor lentiviral genetic barcoding for the marking of individual cells within cell preparations. Based on this, we could track the clonal composition of syngenic MSCs, iPSCs, and iMSCs during culture expansion. Furthermore, we analyzed DNA methylation patterns at senescence-associated genomic regions by barcoded bisulfite amplicon sequencing. The proliferation and differentiation capacities of individual subclones within MSCs and iMSCs were investigated with limiting dilution assays. RESULTS Overall, the clonal composition of primary MSCs and iPSCs gradually declined during expansion. In contrast, iMSCs became oligoclonal early during differentiation, indicating that they were derived from few individual iPSCs. This dominant clonal outgrowth of iMSCs was not associated with changes in chromosomal copy number variation. Furthermore, clonal dynamics were not clearly reflected by stochastically acquired DNA methylation patterns. Limiting dilution assays revealed that iMSCs are heterogeneous in colony formation and in vitro differentiation potential, while this was even more pronounced in primary MSCs. CONCLUSIONS Our results indicate that the subclonal diversity of MSCs and iPSCs declines gradually during in vitro culture, whereas derivation of iMSCs may stem from few individual iPSCs. Differentiation regimen needs to be further optimized to achieve homogeneous differentiation of iPSCs towards iMSCs.
Collapse
|
38
|
Möller J, Rosenberg M, Riecken K, Pörtner R, Zeng AP, Jandt U. Quantification of the dynamics of population heterogeneities in CHO cultures with stably integrated fluorescent markers. Anal Bioanal Chem 2020; 412:2065-2080. [PMID: 32130440 PMCID: PMC7072063 DOI: 10.1007/s00216-020-02401-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 12/20/2019] [Accepted: 01/09/2020] [Indexed: 12/13/2022]
Abstract
Cell population heterogeneities and their changes in mammalian cell culture processes are still not well characterized. In this study, the formation and dynamics of cell population heterogeneities were investigated with flow cytometry and stably integrated fluorescent markers based on the lentiviral gene ontology (LeGO) vector system. To achieve this, antibody-producing CHO cells were transduced with different LeGO vectors to stably express single or multiple fluorescent proteins. This enables the tracking of the transduced populations and is discussed in two case studies from the field of bioprocess engineering: In case study I, cells were co-transduced to express red, green, and blue fluorescent proteins and the development of sub-populations and expression heterogeneities were investigated in high passage cultivations (total 130 days). The formation of a fast-growing and more productive population was observed with a simultaneous increase in cell density and product titer. In case study II, different preculture growth phases and their influence on the population dynamics were investigated in mixed batch cultures with flow cytometry (offline and automated). Four cell line derivatives, each expressing a different fluorescent protein, were generated and cultivated for different time intervals, corresponding to different growth phases. Mixed cultures were inoculated from them, and changes in the composition of the cell populations were observed during the first 48 h of cultivation with reduced process productivity. In summary, we showed how the dynamics of population heterogeneities can be characterized. This represents a novel approach to investigate the dynamics of cell population heterogeneities under near-physiological conditions with changing productivity in mammalian cell culture processes.
Collapse
Affiliation(s)
- Johannes Möller
- Hamburg University of Technology, Bioprocess and Biosystems Engineering, Denickestr. 15, 21073, Hamburg, Germany.
| | - Marcel Rosenberg
- Hamburg University of Technology, Bioprocess and Biosystems Engineering, Denickestr. 15, 21073, Hamburg, Germany
| | - Kristoffer Riecken
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Centre (UMC) Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Ralf Pörtner
- Hamburg University of Technology, Bioprocess and Biosystems Engineering, Denickestr. 15, 21073, Hamburg, Germany
| | - An-Ping Zeng
- Hamburg University of Technology, Bioprocess and Biosystems Engineering, Denickestr. 15, 21073, Hamburg, Germany
| | - Uwe Jandt
- Hamburg University of Technology, Bioprocess and Biosystems Engineering, Denickestr. 15, 21073, Hamburg, Germany
| |
Collapse
|
39
|
Roh V, Abramowski P, Hiou-Feige A, Cornils K, Rivals JP, Zougman A, Aranyossy T, Thielecke L, Truan Z, Mermod M, Monnier Y, Prassolov V, Glauche I, Nowrouzi A, Abdollahi A, Fehse B, Simon C, Tolstonog GV. Cellular Barcoding Identifies Clonal Substitution as a Hallmark of Local Recurrence in a Surgical Model of Head and Neck Squamous Cell Carcinoma. Cell Rep 2019; 25:2208-2222.e7. [PMID: 30463016 DOI: 10.1016/j.celrep.2018.10.090] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 09/04/2018] [Accepted: 10/24/2018] [Indexed: 01/04/2023] Open
Abstract
Local recurrence after surgery for head and neck squamous cell carcinoma (HNSCC) remains a common event associated with a dismal prognosis. Improving this outcome requires a better understanding of cancer cell populations that expand from postsurgical minimal residual disease (MRD). Therefore, we assessed clonal dynamics in a surgical model of barcoded HNSCC growing in the submental region of immunodeficient mice. Clonal substitution and massive reduction of clonal heterogeneity emerged as hallmarks of local recurrence, as the clones dominating in less heterogeneous recurrences were scarce in their matched primary tumors. These lineages were selected by their ability to persist after surgery and competitively expand from MRD. Clones enriched in recurrences exhibited both private and shared genetic features and likely originated from ancestors shared with clones dominating in primary tumors. They demonstrated high invasiveness and epithelial-to-mesenchymal transition, eventually providing an attractive target for obtaining better local control for these tumors.
Collapse
Affiliation(s)
- Vincent Roh
- Department of Otolaryngology - Head and Neck Surgery, University Hospital of Lausanne, Lausanne, Switzerland
| | - Pierre Abramowski
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Agnès Hiou-Feige
- Department of Otolaryngology - Head and Neck Surgery, University Hospital of Lausanne, Lausanne, Switzerland
| | - Kerstin Cornils
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Jean-Paul Rivals
- Department of Otolaryngology - Head and Neck Surgery, University Hospital of Lausanne, Lausanne, Switzerland
| | - Alexandre Zougman
- Clinical and Biomedical Proteomics Group, Cancer Research UK Centre, Leeds Institute of Cancer and Pathology, St. James's University Hospital, Leeds, UK
| | - Tim Aranyossy
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Lars Thielecke
- Institute for Medical Informatics and Biometry, Faculty of Medicine Carl Gustave Carus, Technische Universität Dresden, Dresden, Germany
| | - Zinnia Truan
- Department of Otolaryngology - Head and Neck Surgery, University Hospital of Lausanne, Lausanne, Switzerland
| | - Maxime Mermod
- Department of Otolaryngology - Head and Neck Surgery, University Hospital of Lausanne, Lausanne, Switzerland
| | - Yan Monnier
- Department of Otolaryngology - Head and Neck Surgery, University Hospital of Lausanne, Lausanne, Switzerland
| | - Vladimir Prassolov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia
| | - Ingmar Glauche
- Institute for Medical Informatics and Biometry, Faculty of Medicine Carl Gustave Carus, Technische Universität Dresden, Dresden, Germany
| | - Ali Nowrouzi
- German Cancer Consortium (DKTK), Translational Radiation Oncology, German Cancer Research Center (DKFZ), Core Center Heidelberg, Heidelberg, Germany; Division of Molecular and Translational Radiation Oncology, Heidelberg University Hospital (UKHD) and DKFZ, Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Research in Oncology (NCRO), Heidelberg, Germany
| | - Amir Abdollahi
- German Cancer Consortium (DKTK), Translational Radiation Oncology, German Cancer Research Center (DKFZ), Core Center Heidelberg, Heidelberg, Germany; Division of Molecular and Translational Radiation Oncology, Heidelberg University Hospital (UKHD) and DKFZ, Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Research in Oncology (NCRO), Heidelberg, Germany
| | - Boris Fehse
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.
| | - Christian Simon
- Department of Otolaryngology - Head and Neck Surgery, University Hospital of Lausanne, Lausanne, Switzerland.
| | - Genrich V Tolstonog
- Department of Otolaryngology - Head and Neck Surgery, University Hospital of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
40
|
Möller J, Bhat K, Riecken K, Pörtner R, Zeng AP, Jandt U. Process-induced cell cycle oscillations in CHO cultures: Online monitoring and model-based investigation. Biotechnol Bioeng 2019; 116:2931-2943. [PMID: 31342512 DOI: 10.1002/bit.27124] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 07/05/2019] [Accepted: 07/19/2019] [Indexed: 01/04/2023]
Abstract
The influence of process strategies on the dynamics of cell population heterogeneities in mammalian cell culture is still not well understood. We recently found that the progression of cells through the cell cycle causes metabolic regulations with variable productivities in antibody-producing Chimese hamster ovary (CHO) cells. On the other hand, it is so far unknown how bulk cultivation conditions, for example, variable nutrient concentrations depending on process strategies, can influence cell cycle-derived population dynamics. In this study, process-induced cell cycle synchronization was assessed in repeated-batch and fed-batch cultures. An automated flow cytometry set-up was developed to measure the cell cycle distribution online, using antibody-producing CHO DP-12 cells transduced with the cell cycle-specific fluorescent ubiquitination-based cell cycle indicator (FUCCI) system. On the basis of the population-resolved model, feeding-induced partial self-synchronization was predicted and the results were evaluated experimentally. In the repeated-batch culture, stable cell cycle oscillations were confirmed with an oscillating G1 phase distribution between 41% and 72%. Furthermore, oscillations of the cell cycle distribution were simulated and determined in a (bolus) fed-batch process with up to 25 × 1 0 6 cells/ml. The cell cycle synchronization arose with pulse feeding only and ceased with continuous feeding. Both simulated and observed oscillations occurred at higher frequencies than those observable based on regular (e.g., daily) sample analysis, thus demonstrating the need for high-frequency online cell cycle analysis. In summary, we showed how experimental methods combined with simulations enable the improved assessment of the effects of process strategies on the dynamics of cell cycle-dependent population heterogeneities. This provides a novel approach to understand cell cycle regulations, control cell population dynamics, avoid inadvertently induced oscillations of cell cycle distributions and thus to improve process stability and efficiency.
Collapse
Affiliation(s)
- Johannes Möller
- Bioprocess and Biosystems Engineering, Hamburg University of Technology, Hamburg, Germany
| | - Krathika Bhat
- Bioprocess and Biosystems Engineering, Hamburg University of Technology, Hamburg, Germany
| | - Kristoffer Riecken
- Department of Stem Cell Transplantation, Research Department Cell and Gene Therapy, University Medical Centre (UMC) Hamburg-Eppendorf, Hamburg, Germany
| | - Ralf Pörtner
- Bioprocess and Biosystems Engineering, Hamburg University of Technology, Hamburg, Germany
| | - An-Ping Zeng
- Bioprocess and Biosystems Engineering, Hamburg University of Technology, Hamburg, Germany
| | - Uwe Jandt
- Bioprocess and Biosystems Engineering, Hamburg University of Technology, Hamburg, Germany
| |
Collapse
|
41
|
van der Heijden M, Miedema DM, Waclaw B, Veenstra VL, Lecca MC, Nijman LE, van Dijk E, van Neerven SM, Lodestijn SC, Lenos KJ, de Groot NE, Prasetyanti PR, Arricibita Varea A, Winton DJ, Medema JP, Morrissey E, Ylstra B, Nowak MA, Bijlsma MF, Vermeulen L. Spatiotemporal regulation of clonogenicity in colorectal cancer xenografts. Proc Natl Acad Sci U S A 2019; 116:6140-6145. [PMID: 30850544 PMCID: PMC6442578 DOI: 10.1073/pnas.1813417116] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Cancer evolution is predominantly studied by focusing on differences in the genetic characteristics of malignant cells within tumors. However, the spatiotemporal dynamics of clonal outgrowth that underlie evolutionary trajectories remain largely unresolved. Here, we sought to unravel the clonal dynamics of colorectal cancer (CRC) expansion in space and time by using a color-based clonal tracing method. This method involves lentiviral red-green-blue (RGB) marking of cell populations, which enabled us to track individual cells and their clonal outgrowth during tumor initiation and growth in a xenograft model. We found that clonal expansion largely depends on the location of a clone, as small clones reside in the center and large clones mostly drive tumor growth at the border. These dynamics are recapitulated in a computational model, which confirms that the clone position within a tumor rather than cell-intrinsic features, is crucial for clonal outgrowth. We also found that no significant clonal loss occurs during tumor growth and clonal dispersal is limited in most models. Our results imply that, in addition to molecular features of clones such as (epi-)genetic differences between cells, clone location and the geometry of tumor growth are crucial for clonal expansion. Our findings suggest that either microenvironmental signals on the tumor border or differences in physical properties within the tumor, are major contributors to explain heterogeneous clonal expansion. Thus, this study provides further insights into the dynamics of solid tumor growth and progression, as well as the origins of tumor cell heterogeneity in a relevant model system.
Collapse
Affiliation(s)
- Maartje van der Heijden
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam Gastroenterology & Metabolism, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Daniël M Miedema
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam Gastroenterology & Metabolism, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Bartlomiej Waclaw
- School of Physics and Astronomy, The University of Edinburgh, EH9 3FD Edinburgh, United Kingdom
| | - Veronique L Veenstra
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam Gastroenterology & Metabolism, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Maria C Lecca
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam Gastroenterology & Metabolism, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Lisanne E Nijman
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam Gastroenterology & Metabolism, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Erik van Dijk
- Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Sanne M van Neerven
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam Gastroenterology & Metabolism, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Sophie C Lodestijn
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam Gastroenterology & Metabolism, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Kristiaan J Lenos
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam Gastroenterology & Metabolism, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Nina E de Groot
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam Gastroenterology & Metabolism, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Pramudita R Prasetyanti
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam Gastroenterology & Metabolism, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Andrea Arricibita Varea
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam Gastroenterology & Metabolism, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Douglas J Winton
- Cancer Research UK, Cambridge Institute, University of Cambridge, CB2 0RE Cambridge, United Kingdom
| | - Jan Paul Medema
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam Gastroenterology & Metabolism, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Edward Morrissey
- Medical Research Council Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, OX3 9DS Oxford, United Kingdom
| | - Bauke Ylstra
- Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Martin A Nowak
- Program for Evolutionary Dynamics, Harvard University, Cambridge, MA 02138
| | - Maarten F Bijlsma
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam Gastroenterology & Metabolism, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Louis Vermeulen
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam Gastroenterology & Metabolism, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands;
| |
Collapse
|
42
|
Giersch K, Bhadra OD, Volz T, Allweiss L, Riecken K, Fehse B, Lohse AW, Petersen J, Sureau C, Urban S, Dandri M, Lütgehetmann M. Hepatitis delta virus persists during liver regeneration and is amplified through cell division both in vitro and in vivo. Gut 2019; 68:150-157. [PMID: 29217749 DOI: 10.1136/gutjnl-2017-314713] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 11/17/2017] [Accepted: 11/20/2017] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Hepatitis delta virus (HDV) was shown to persist for weeks in the absence of HBV and for months after liver transplantation, demonstrating the ability of HDV to persevere in quiescent hepatocytes. The aim of the study was to evaluate the impact of cell proliferation on HDV persistence in vitro and in vivo. DESIGN Genetically labelled human sodium taurocholate cotransporting polypeptide (hNTCP)-transduced human hepatoma(HepG2) cells were infected with HBV/HDV and passaged every 7 days for 100 days in the presence of the entry inhibitor Myrcludex-B. In vivo, cell proliferation was triggered by transplanting primary human hepatocytes (PHHs) isolated from HBV/HDV-infected humanised mice into naïve recipients. Virological parameters were measured by quantitative real time polymerase chain reaction (qRT-PCR). Hepatitis delta antigen (HDAg), hepatitis B core antigen (HBcAg) and cell proliferation were determined by immunofluorescence. RESULTS Despite 15 in vitro cell passages and block of viral spreading by Myrcludex-B, clonal cell expansion permitted amplification of HDV infection. In vivo, expansion of PHHs isolated from HBV/HDV-infected humanised mice was confirmed 3 days, 2, 4 and 8 weeks after transplantation. While HBV markers rapidly dropped in proliferating PHHs, HDAg-positive hepatocytes were observed among dividing cells at all time points. Notably, HDAg-positive cells appeared in clusters, indicating that HDV was transmitted to daughter cells during liver regeneration even in the absence of de novo infection. CONCLUSION This study demonstrates that HDV persists during liver regeneration by transmitting HDV RNA to dividing cells even in the absence of HBV coinfection. The strong persistence capacities of HDV may also explain why HDV clearance is difficult to achieve in HBV/HDV chronically infected patients.
Collapse
Affiliation(s)
- Katja Giersch
- I. Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Oliver D Bhadra
- I. Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tassilo Volz
- I. Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lena Allweiss
- I. Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kristoffer Riecken
- Department of Stem Cell transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Boris Fehse
- Department of Stem Cell transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ansgar W Lohse
- I. Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,German Center for Infection Research (DZIF), Hamburg-Lübeck-Borstel and Heidelberg Partner sites, Germany
| | - Joerg Petersen
- IFI Institute for Interdisciplinary Medicine, Asklepios Clinic St. Georg, Hamburg, Germany
| | - Camille Sureau
- Laboratoirede Virologie Moleculaire, INTS, Centre National de la Recherche Scientifique, Paris, France
| | - Stephan Urban
- German Center for Infection Research (DZIF), Hamburg-Lübeck-Borstel and Heidelberg Partner sites, Germany.,Department of Infectious Diseases, Molecular Virology, University Hospital Heidelberg, Heidelberg, Germany
| | - Maura Dandri
- I. Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,German Center for Infection Research (DZIF), Hamburg-Lübeck-Borstel and Heidelberg Partner sites, Germany
| | - Marc Lütgehetmann
- Institute of Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
43
|
R SK, Kumar SKA, Vijayakrishna K, Sivaramakrishna A, Brahmmananda Rao CVS, Sivaraman N, Sahoo SK. Development of the Smartphone-Assisted Colorimetric Detection of Thorium by Using New Schiff’s Base and Its Applications to Real Time Samples. Inorg Chem 2018; 57:15270-15279. [DOI: 10.1021/acs.inorgchem.8b02564] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Selva Kumar R
- Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| | - S. K. Ashok Kumar
- Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| | - Kari Vijayakrishna
- Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| | - Akella Sivaramakrishna
- Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| | - C. V. S. Brahmmananda Rao
- Indira Gandhi Centre for Atomic Research, Homi Bhabha National Institute, Kalpakkam 603102, Tamil Nadu, India
| | - N. Sivaraman
- Indira Gandhi Centre for Atomic Research, Homi Bhabha National Institute, Kalpakkam 603102, Tamil Nadu, India
| | - Suban K. Sahoo
- Department of Applied Chemistry, S. V. National Institute Technology, Surat 395007, Gujarat, India
| |
Collapse
|
44
|
Zubiaur P, Saiz-Rodríguez M, Koller D, Ovejero-Benito MC, Wojnicz A, Abad-Santos F. How to make P-glycoprotein (ABCB1, MDR1) harbor mutations and measure its expression and activity in cell cultures? Pharmacogenomics 2018; 19:1285-1297. [PMID: 30334473 DOI: 10.2217/pgs-2018-0101] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Several polymorphisms have been identified in ABCB1, the gene encoding for the P-glycoprotein. This transporter alters the pharmacokinetics or effectiveness of drugs by excreting them from cells where it is expressed (e.g., blood-brain barrier, intestine or tumors). No consensus has been reached regarding the functional consequences of these polymorphisms in the transporter's function. The aim of this review was to describe a methodology that allows the assessment of P-gp function when harboring polymorphisms. We describe how to obtain cell lines with high expression levels of the transporter with polymorphisms and several tactics to measure its expression and activity. This methodology may help elucidate the contribution of polymorphisms in ABCB1 to drug pharmacokinetics, effectiveness and safety or to cancer chemotherapy failure.
Collapse
Affiliation(s)
- Pablo Zubiaur
- Clinical Pharmacology Department, Hospital Universitario de la Princesa, Instituto Teófilo Hernando, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria la Princesa (IP), Madrid, Spain
| | - Miriam Saiz-Rodríguez
- Clinical Pharmacology Department, Hospital Universitario de la Princesa, Instituto Teófilo Hernando, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria la Princesa (IP), Madrid, Spain
| | - Dora Koller
- Clinical Pharmacology Department, Hospital Universitario de la Princesa, Instituto Teófilo Hernando, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria la Princesa (IP), Madrid, Spain
| | - María Carmen Ovejero-Benito
- Clinical Pharmacology Department, Hospital Universitario de la Princesa, Instituto Teófilo Hernando, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria la Princesa (IP), Madrid, Spain
| | - Aneta Wojnicz
- Clinical Pharmacology Department, Hospital Universitario de la Princesa, Instituto Teófilo Hernando, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria la Princesa (IP), Madrid, Spain
| | - Francisco Abad-Santos
- Clinical Pharmacology Department, Hospital Universitario de la Princesa, Instituto Teófilo Hernando, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria la Princesa (IP), Madrid, Spain.,UICEC Hospital Universitario de la Princesa, Plataforma SCReN (Spanish Clinical Reseach Network), Instituto de Investigación Sanitaria la Princesa (IP), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
45
|
Gambera S, Abarrategi A, González-Camacho F, Morales-Molina Á, Roma J, Alfranca A, García-Castro J. Clonal dynamics in osteosarcoma defined by RGB marking. Nat Commun 2018; 9:3994. [PMID: 30266933 PMCID: PMC6162235 DOI: 10.1038/s41467-018-06401-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Accepted: 09/03/2018] [Indexed: 02/06/2023] Open
Abstract
Osteosarcoma is a type of bone tumour characterized by considerable levels of phenotypic heterogeneity, aneuploidy, and a high mutational rate. The life expectancy of osteosarcoma patients has not changed during the last three decades and thus much remains to be learned about the disease biology. Here, we employ a RGB-based single-cell tracking system to study the clonal dynamics occurring in a de novo-induced murine osteosarcoma model. We show that osteosarcoma cells present initial polyclonal dynamics, followed by clonal dominance associated with adaptation to the microenvironment. Interestingly, the dominant clones are composed of subclones with a similar tumour generation potential when they are re-implanted in mice. Moreover, individual spontaneous metastases are clonal or oligoclonal, but they have a different cellular origin than the dominant clones present in primary tumours. In summary, we present evidence that osteosarcomagenesis can follow a neutral evolution model, in which different cancer clones coexist and propagate simultaneously.
Collapse
Affiliation(s)
- Stefano Gambera
- Cellular Biotechnology Unit, Instituto de Salud Carlos III (ISCIII), Madrid, 28029, Spain
| | - Ander Abarrategi
- Cellular Biotechnology Unit, Instituto de Salud Carlos III (ISCIII), Madrid, 28029, Spain
- Haematopoietic Stem Cell Lab, The Francis Crick Institute, London, NW1 1AT, UK
| | | | - Álvaro Morales-Molina
- Cellular Biotechnology Unit, Instituto de Salud Carlos III (ISCIII), Madrid, 28029, Spain
| | - Josep Roma
- Laboratory of Translational Research in Child and Adolescent Cancer, Vall d'Hebron Hospital, Barcelona, 08035, Spain
| | - Arantzazu Alfranca
- Cellular Biotechnology Unit, Instituto de Salud Carlos III (ISCIII), Madrid, 28029, Spain
- Immunology Department, Hospital Universitario de La Princesa, Madrid, 28006, Spain
| | - Javier García-Castro
- Cellular Biotechnology Unit, Instituto de Salud Carlos III (ISCIII), Madrid, 28029, Spain.
| |
Collapse
|
46
|
Aranyossy T, Thielecke L, Glauche I, Fehse B, Cornils K. Genetic Barcodes Facilitate Competitive Clonal Analyses In Vivo. Hum Gene Ther 2018; 28:926-937. [PMID: 28847169 DOI: 10.1089/hum.2017.124] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Monitoring the fate of individual cell clones is an important task to better understand normal tissue regeneration, for example after hematopoietic stem cell (HSC) transplantation, but also cancerogenesis. Based on their integration into the host cell's genome, retroviral vectors are commonly used to stably mark target cells and their progeny. The development of genetic barcoding techniques has opened new possibilities to determine clonal composition and dynamics in great detail. A modular genetic barcode was recently introduced consisting of 32 variable positions (BC32) with a customized backbone, and its advantages were demonstrated with regard to barcode calling and quantification. The study presented applied the BC32 system in a complex in vivo situation, namely to analyze clonal reconstitution dynamics for HSC grafts consisting of up to three cell populations with distinguishable barcodes using different alpha- and lentiviral vectors. In a competitive transplantation setup, it was possible to follow the differently marked cell populations within individual animals. This enabled the clonal contribution of the different BC32 constructs during reconstitution and long-term hematopoiesis in the peripheral blood and the spatial distribution in bone marrow and spleen to be identified. Thus, it was demonstrated that the system allows the output of individually marked cells to be tracked in vivo and their influence on clonal dynamics to be analyzed. Successful application of the BC32 system in a complex, competitive in vivo situation provided proof-of-principle that its high complexity and the large Hamming distance between individual barcodes, combined with the easy customization, facilitate efficient and precise quantification, even without prior knowledge of individual barcode sequences. Importantly, simultaneous high-sensitivity analyses of different cell populations in single animals may significantly reduce numbers of animals required to investigate specific scientific questions in accordance with RRR principles. It is concluded that this BC32 system will be excellently suited for various research applications in regenerative medicine and cancer biology.
Collapse
Affiliation(s)
- Tim Aranyossy
- 1 Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf , Hamburg, Germany
| | - Lars Thielecke
- 2 Institute for Medical Informatics and Biometry, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Ingmar Glauche
- 2 Institute for Medical Informatics and Biometry, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Boris Fehse
- 1 Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf , Hamburg, Germany
| | - Kerstin Cornils
- 1 Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf , Hamburg, Germany .,3 Department of Pediatric Hematology and Oncology, Division Pediatric Stem Cell Transplantation and Immunology, University Medical Center Hamburg-Eppendorf , Hamburg, Germany .,4 Research Institute Children's Cancer Center Hamburg, Hamburg, Germany
| |
Collapse
|
47
|
Robust imaging and gene delivery to study human lymphoblastoid cell lines. J Hum Genet 2018; 63:945-955. [DOI: 10.1038/s10038-018-0483-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 05/23/2018] [Accepted: 05/30/2018] [Indexed: 12/20/2022]
|
48
|
Brenière-Letuffe D, Domke-Shibamiya A, Hansen A, Eschenhagen T, Fehse B, Riecken K, Stenzig J. Clonal dynamics studied in cultured induced pluripotent stem cells reveal major growth imbalances within a few weeks. Stem Cell Res Ther 2018; 9:165. [PMID: 29914569 PMCID: PMC6006556 DOI: 10.1186/s13287-018-0893-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 04/06/2018] [Accepted: 05/02/2018] [Indexed: 12/19/2022] Open
Abstract
Background Human induced pluripotent stem (iPS) cells have revolutionised research and spark hopes for future tissue replacement therapies. To obtain high cell numbers, iPS cells can be expanded indefinitely. However, as long-term expansion can compromise cell integrity and quality, we set out to assess potential reduction of clonal diversity by inherent growth imbalances. Methods Using red, green, blue marking as a lentiviral multi-colour clonal cell tracking technology, we marked three different iPS cell lines as well as three other cell lines, assigning a unique fluorescent colour to each cell at one point in culture. Subsequently, we followed the sub-clonal distribution over time by flow cytometry and fluorescence microscopy analysis in regular intervals. Results In three human iPS cell lines as well as primary human fibroblasts and two widely used human cell lines as controls (K562 and HEK 293 T), we observed a marked reduction in sub-clonal diversity over time of culture (weeks). After 38 passages, all iPS cultures consisted of less than 10 residual clones. Karyotype and function, the latter assessed by cardiomyocyte differentiation and tissue engineering, did not reveal obvious differences. Conclusions Our results argue for a quick selection of sub-clones with a growth advantage and flag a normally invisible and potentially undesired behaviour of cultured iPS cells, especially when using long-term cultured iPS cells for experiments or even in-vivo applications. Electronic supplementary material The online version of this article (10.1186/s13287-018-0893-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- David Brenière-Letuffe
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Aya Domke-Shibamiya
- Core Facility Stem Cells, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Arne Hansen
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Thomas Eschenhagen
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Boris Fehse
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Kristoffer Riecken
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany.
| | - Justus Stenzig
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany. .,DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany.
| |
Collapse
|
49
|
Nuclear accumulation of SHIP1 mutants derived from AML patients leads to increased proliferation of leukemic cells. Cell Signal 2018; 49:87-94. [PMID: 29852247 DOI: 10.1016/j.cellsig.2018.05.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 05/18/2018] [Accepted: 05/18/2018] [Indexed: 11/22/2022]
Abstract
The inositol 5-phosphatase SHIP1 acts as negative regulator of intracellular signaling in myeloid cells and is a tumor suppressor in myeloid leukemogenesis. After relocalization from the cytoplasm to the plasma membrane SHIP1 terminates PI3-kinase mediated signaling processes. Furthermore, SHIP1 is also found in distinct puncta in the cell nucleus and nuclear SHIP1 has a pro-proliferative function. Here we report the identification of five nuclear export signals (NESs) which regulate together with the two known nuclear localization signals (NLSs) the nucleocytoplasmic shuttling of SHIP1. Mutation of NLSs reduced the nuclear import and mutation of NESs decreased the nuclear export of SHIP1 in the acute myeloid leukemia (AML) cell line UKE-1. Interestingly, four SHIP1 mutants (K210R, N508D, V684E, Q1153L) derived from AML patients showed a nuclear accumulation after expression in UKE-1 cells. In addition, overexpression of the AML patient-derived mutation N508D caused an increased proliferation rate of UKE-1 cells in comparison to wild type SHIP1. Furthermore, we identified serine and tyrosine phosphorylation as a molecular mechanism for the regulation of nucleocytoplasmic shuttling of SHIP1 where tyrosine phosphorylation of distinct residues i.e. Y864, Y914, Y1021 reduces nuclear localization, whereas serine phosphorylation at S933 enhances nuclear localization of SHIP1. In summary, our data further implicate nuclear SHIP1 in cellular signaling and suggest that enhanced accumulation of SHIP1 mutants in the nucleus may be a contributory factor of abnormally high proliferation of AML cells.
Collapse
|
50
|
Behrenbruch C, Shembrey C, Paquet-Fifield S, Mølck C, Cho HJ, Michael M, Thomson BNJ, Heriot AG, Hollande F. Surgical stress response and promotion of metastasis in colorectal cancer: a complex and heterogeneous process. Clin Exp Metastasis 2018; 35:333-345. [PMID: 29335811 DOI: 10.1007/s10585-018-9873-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 01/06/2018] [Indexed: 12/12/2022]
Abstract
Surgery remains the curative treatment modality for colorectal cancer in all stages, including stage IV with resectable liver metastasis. There is emerging evidence that the stress response caused by surgery as well as other perioperative therapies such as anesthesia and analgesia may promote growth of pre-existing micro-metastasis or potentially initiate tumor dissemination. Therapeutically targeting the perioperative period may therefore reduce the effect that surgical treatments have in promoting metastases, for example by combining β-adrenergic receptor antagonists and cyclooxygenase-2 (COX-2) inhibitors in the perioperative setting. In this paper, we highlight some of the mechanisms that may underlie surgery-related metastatic development in colorectal cancer. These include direct tumor spillage at the time of surgery, suppression of the anti-tumor immune response, direct stimulatory effects on tumor cells, and activation of the coagulation system. We summarize in more detail results that support a role for catecholamines as major drivers of the pro-metastatic effect induced by the surgical stress response, predominantly through activation of β-adrenergic signaling. Additionally, we argue that an improved understanding of surgical stress-induced dissemination, and more specifically whether it impacts on the level and nature of heterogeneity within residual tumor cells, would contribute to the successful clinical targeting of this process. Finally, we provide a proof-of-concept demonstration that ex-vivo analyses of colorectal cancer patient-derived samples using RGB-labeling technology can provide important insights into the heterogeneous sensitivity of tumor cells to stress signals. This suggests that intra-tumor heterogeneity is likely to influence the efficacy of perioperative β-adrenergic receptor and COX-2 inhibition, and that ex-vivo characterization of heterogeneous stress response in tumor samples can synergize with other models to optimize perioperative treatments and further improve outcome in colorectal and other solid cancers.
Collapse
Affiliation(s)
- Corina Behrenbruch
- Department of Pathology, University of Melbourne Centre for Cancer Research, The University of Melbourne, Victorian Comprehensive Cancer Centre, Level 10, 305 Grattan Street, Melbourne, VIC, 3000, Australia
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Victorian Comprehensive Cancer Centre, 305 Grattan St, Melbourne, 3000, Australia
| | - Carolyn Shembrey
- Department of Pathology, University of Melbourne Centre for Cancer Research, The University of Melbourne, Victorian Comprehensive Cancer Centre, Level 10, 305 Grattan Street, Melbourne, VIC, 3000, Australia
| | - Sophie Paquet-Fifield
- Department of Pathology, University of Melbourne Centre for Cancer Research, The University of Melbourne, Victorian Comprehensive Cancer Centre, Level 10, 305 Grattan Street, Melbourne, VIC, 3000, Australia
| | - Christina Mølck
- Department of Pathology, University of Melbourne Centre for Cancer Research, The University of Melbourne, Victorian Comprehensive Cancer Centre, Level 10, 305 Grattan Street, Melbourne, VIC, 3000, Australia
| | - Hyun-Jung Cho
- Biological Optical Microscopy Platform, The University of Melbourne, Medical Building, Grattan Street, Parkville, 3010, Australia
| | - Michael Michael
- Division of Cancer Medicine, Peter MacCallum Cancer Centre, Victorian Comprehensive Cancer Centre, 305 Grattan St, Melbourne, 3000, Australia
| | - Benjamin N J Thomson
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Victorian Comprehensive Cancer Centre, 305 Grattan St, Melbourne, 3000, Australia
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, 300 Grattan St, Parkville, 3000, Australia
| | - Alexander G Heriot
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Victorian Comprehensive Cancer Centre, 305 Grattan St, Melbourne, 3000, Australia
| | - Frédéric Hollande
- Department of Pathology, University of Melbourne Centre for Cancer Research, The University of Melbourne, Victorian Comprehensive Cancer Centre, Level 10, 305 Grattan Street, Melbourne, VIC, 3000, Australia.
| |
Collapse
|