Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

State-of-the-art analytical methods of viral infections in human lung organoids

  • Morris Baumgardt ,

    Contributed equally to this work with: Morris Baumgardt, Maren Hülsemann

    Roles Conceptualization, Methodology, Visualization, Writing – original draft, Writing – review & editing

    Affiliation Department of Infectious Diseases and Respiratory Medicine, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany

  • Maren Hülsemann ,

    Contributed equally to this work with: Morris Baumgardt, Maren Hülsemann

    Roles Conceptualization, Writing – original draft, Writing – review & editing

    Affiliation Berlin Institute of Health at Charité (BIH), BIH QUEST Center for Responsible Research, Berlin, Germany

  • Anna Löwa,

    Roles Methodology, Writing – review & editing

    Affiliation Department of Infectious Diseases and Respiratory Medicine, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany

  • Diana Fatykhova,

    Roles Methodology, Visualization

    Affiliation Department of Infectious Diseases and Respiratory Medicine, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany

  • Karen Hoffmann,

    Roles Methodology

    Affiliation Department of Infectious Diseases and Respiratory Medicine, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany

  • Mirjana Kessler,

    Roles Methodology

    Affiliations Department of Infectious Diseases and Respiratory Medicine, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany, Department of Gynecology and Obstetrics, University Hospital, LMU, Munich, Germany

  • Maren Mieth,

    Roles Methodology

    Affiliation Department of Infectious Diseases and Respiratory Medicine, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany

  • Katharina Hellwig,

    Roles Methodology, Visualization

    Affiliation Department of Infectious Diseases and Respiratory Medicine, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany

  • Doris Frey,

    Roles Methodology

    Affiliation Department of Infectious Diseases and Respiratory Medicine, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany

  • Alina Langenhagen,

    Roles Methodology

    Affiliation Department of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany

  • Anne Voss,

    Roles Methodology

    Affiliation Department of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany

  • Benedikt Obermayer,

    Roles Methodology

    Affiliation Core Unit Bioinformatics, Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany

  • Emanuel Wyler,

    Roles Methodology

    Affiliation Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC) and IRI Life Sciences, Institute for Biology, Humboldt Universität zu Berlin, Berlin, Germany

  • Simon Dökel,

    Roles Methodology

    Affiliation Department of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany

  • Achim D. Gruber,

    Roles Methodology

    Affiliation Department of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany

  • Ulf Tölch,

    Roles Methodology

    Affiliation Berlin Institute of Health at Charité (BIH), BIH QUEST Center for Responsible Research, Berlin, Germany

  • Stefan Hippenstiel,

    Roles Funding acquisition, Supervision, Writing – review & editing

    Affiliation Department of Infectious Diseases and Respiratory Medicine, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany

  • Andreas C. Hocke,

    Roles Funding acquisition, Supervision, Visualization, Writing – review & editing

    Affiliation Department of Infectious Diseases and Respiratory Medicine, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany

  •  [ ... ],
  • Katja Hönzke

    Roles Funding acquisition, Methodology, Visualization, Writing – review & editing

    katja.hoenzke@charite.de

    Affiliation Department of Infectious Diseases and Respiratory Medicine, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany

  • [ view all ]
  • [ view less ]

Correction

3 Nov 2023: Baumgardt M, Hülsemann M, Löwa A, Fatykhova D, Hoffmann K, et al. (2023) Correction: State-of-the-art analytical methods of viral infections in human lung organoids. PLOS ONE 18(11): e0294216. https://doi.org/10.1371/journal.pone.0294216 View correction

Abstract

Human-based organ models can provide strong predictive value to investigate the tropism, virulence, and replication kinetics of viral pathogens. Currently, such models have received widespread attention in the study of SARS-CoV-2 causing the COVID-19 pandemic. Applicable to a large set of organoid models and viruses, we provide a step-by-step work instruction for the infection of human alveolar-like organoids with SARS-CoV-2 in this protocol collection. We also prepared a detailed description on state-of-the-art methodologies to assess the infection impact and the analysis of relevant host factors in organoids. This protocol collection consists of five different sets of protocols. Set 1 describes the protein extraction from human alveolar-like organoids and the determination of protein expression of angiotensin-converting enzyme 2 (ACE2), transmembrane serine protease 2 (TMPRSS2) and FURIN as exemplary host factors of SARS-CoV-2. Set 2 provides detailed guidance on the extraction of RNA from human alveolar-like organoids and the subsequent qPCR to quantify the expression level of ACE2, TMPRSS2, and FURIN as host factors of SARS-CoV-2 on the mRNA level. Protocol set 3 contains an in-depth explanation on how to infect human alveolar-like organoids with SARS-CoV-2 and how to quantify the viral replication by plaque assay and viral E gene-based RT-qPCR. Set 4 provides a step-by-step protocol for the isolation of single cells from infected human alveolar-like organoids for further processing in single-cell RNA sequencing or flow cytometry. Set 5 presents a detailed protocol on how to perform the fixation of human alveolar-like organoids and guides through all steps of immunohistochemistry and in situ hybridization to visualize SARS-CoV-2 and its host factors. The infection and all subsequent analytical methods have been successfully validated by biological replications with human alveolar-like organoids based on material from different donors.

Introduction

The current outbreak of the COVID-19 pandemic caused by the severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) has once again highlighted the need for appropriate and rapidly available model systems with high predictive value towards clinical translation [1]. The SARS-CoV-2 exposure of most animal models either did not lead to infection, or only partly reflect relevant human aspects of disease [24]. Hence, human-based organ models provide promising opportunities for the rapid study of pathogens and their impact on the human system [510]. To investigate the effects of a viral pathogen on our respiratory system, human alveolar-like organoids serve as an excellent tool to gain insights on virulence mechanisms and viral tropisms [11]. However, standard infection methods used in classical 2D cell culture systems are not transferable to 3D human alveolar-like organoids [12].

Therefore, we are providing a detailed protocol collection on the infection of human alveolar-like organoids as well as their preparation for associated readouts. The protocol collection includes an in-depth instruction on the handling and preparation of human alveolar-like organoids for infection with SARS-CoV-2 under biosafety level 3 conditions, followed by the measurement of viral replication by plaque assay and viral quantitative reverse transcription PCR (RT-qPCR) [13].

This protocol collection also provides the thorough work instructions to determine the expression of host factors in 3D organ models on the protein and mRNA level. Exemplarily, the expression of ACE2, TMPRSS2, and FURIN as key host factors of SARS-CoV-2 is described by Western blotting and real-time quantitative PCR (qPCR) [14, 15]. Novel technologies such as single-cell RNA sequencing (scRNA-seq) have tremendous impact on current COVID-19 research. Applied on human alveolar-like organoids, scRNA-seq can increase our understanding on the pathophysiological signature, and the cell-mediated immune response after SARS-CoV-2 infection. Therefore, this protocol collection includes a detailed description of single cell preparation from human alveolar-like organoids for further processing in single-cell technologies. Imaging techniques such as immunohistochemistry and in situ hybridization allow a detailed analysis of viral distribution, tissue damage and host factor location in the human alveolar-like organoids. We provide elaborate instructions on sample preparation and staining procedures to provide sections for high resolution imaging [16].

Given the urgency of the pandemic, researchers worldwide need to coordinate their activities and support collaborative efforts in COVID-19 research. Thereby, the distribution of transparent and detailed protocols is an essential tool towards high reproducibility rates and scientific rigor [1719].

Materials and methods

The protocol collection described in this peer-reviewed article is published on protocols.io (doi.org/10.17504/protocols.io.5jyl89pw6v2w/v2) and is attached to this article as S1S6 Files for printing. Informed written consent was obtained from all volunteers and the study was approved by the Charité Ethics Committee (project 451, EA2/079/13).

Expected results

The presented protocols enabled us to screen mature human alveolar-like organoids [11] for three important currently known host factors of SARS-CoV-2.

We analyzed ACE2, TMPRSS2 and FURIN in human alveolar-like organoids (n = 3) as exemplary host factors by Western blot and used Calu-3 cells as positive control (Fig 1). All three host factors were expressed in the alveolar-like organoids. The TMPRSS2 blot showed two characteristic bands described as full-length TMPRSS2 (54kDa) and the cleaved form (25kDa). The mRNA expression level of SARS-CoV-2 host factors ACE2, TMPRSS2, and FURIN were analyzed by qPCR in human alveolar-like organoids (n = 4) (Fig 2). The figure shows the basic expression of the host factors by using Ct values and two different reference genes (GAPDH, β-ACTIN).

thumbnail
Fig 1. Analysis of SARS-CoV-2 host factor expression by Western blot.

Representative Western blots for ACE2, TMPRSS2, and FURIN of protein lysates from human alveolar-like organoids of three different donors (P8-P10). Calu-3 cells served as positive control.

https://doi.org/10.1371/journal.pone.0276115.g001

thumbnail
Fig 2. Analysis of ACE2, TMPRSS2, and FURIN expression in human alveolar-like organoids.

Expression of ACE2, TMPRSS2, FURIN and GAPDH and β-ACTIN as housekeeping genes measured by qPCR on bulk RNA of human alveolar-like organoids. Shown are Ct values of four different donors (P4-P7) and two technical replicates.

https://doi.org/10.1371/journal.pone.0276115.g002

The successful infection of human alveolar-like organoids with SARS-CoV-2 was shown by the increasing number of infectious particles over the time course of 96 hours for seven different donors via plaque assay (Fig 3A). Additionally, the number of viral genomic copies was quantified in the cell culture supernatant via viral E gene-based RT-qPCR, as described in the protocol collection (Fig 3B). The viral RNA that is produced indicates whether viral replication occurs and virions are formed. However, it is not possible to say whether these viral particles are complete and infectious. Therefore, it is of particular importance to also generate a replication curve via the plaque assay approach, which shows the number of infectious particles over time. Especially when it comes to drug screening, the indication of viral RNA is not sufficient. In that sense, the two values are not comparable, but together they give a good indication of the overall replication dynamics.

thumbnail
Fig 3. Replication kinetics of SARS-CoV-2-infected human alveolar-like organoids.

Human alveolar-like organoids were infected with SARS-CoV-2 (MOI = 1) and viral replication was assessed by plaque assay (A) and via viral E gene-based RT-qPCR (B). Data of seven (plaque assay) and four (RT-qPCR) biological replicates are shown as individual data points. The mean is visualized by a horizontal black line.

https://doi.org/10.1371/journal.pone.0276115.g003

After enzymatic digestion of human alveolar-like organoids into single cell suspension, scRNA-seq can be performed to allow dissection of gene expression at single-cell resolution (Fig 4). In this exemplary case, scRNA-seq data from human alveolar-like organoids of six donors were annotated according to Travaglini et al. [20] and the following cell clusters were identified: AT2, basal and secretory cells (Fig 4A). The markers used for cell type identification are shown in Fig 4D and 4E. The used protocol generates organoid cultures of the alveolar epithelium from bipotent progenitors excluding at least in part bronchial cells. AT1 cells were not found in the model, the mechanism underlying their differentiation is not completely understood and needs further elucidation. Fig 4B shows the specific cell cycle phases. Most of the cells are in the G1-phase which is believed to serve as a window of opportunity to initiate cell differentiation [21]. The UMAP in Fig 4C shows the different donors combined in this study. ScRNA-seq enables an in-depth analysis of the effect of SARS-CoV-2 infection on the different cell types and allows validation of their host factor expression levels as shown in Fig 4F for ACE2, TMPRSS2, and FURIN. These expression patterns are consistent with those already observed at the protein and mRNA levels (Figs 1 and 2). All data are available at: https://ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE197949 and https://ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE198864.

thumbnail
Fig 4. Single-cell RNA sequencing displays the proportion of three different cell types and SARS-CoV-2 host factors present in uninfected human alveolar-like organoids.

UMAP embedding of human alveolar-like organoids (n = 6; P1-P3 and P10-P12) shows AT2, basal, and secretory cells present in the organoids (A). Cell cycle phase of each cell in either G2M, S or G1 phase (B) and individual donor composition (C) is displayed. The marker genes needed for cell type identification are shown (D and E) as well as the expression of the SARS-CoV-2 host factors ACE2, TMPRSS2, and FURIN (F).

https://doi.org/10.1371/journal.pone.0276115.g004

In order to visualize the expression patterns of host factors and the virus, we utilized immunohistochemistry (n = 3) and in situ hybridization (n = 3) and acquired high-resolution imaging data showing viral particles that have migrated into human alveolar-like organoids as well as ACE2 expression (Fig 5). In situ hybridization also shows viral particles present in alveolar-like organoids and ACE2 expression (Fig 5). In context of this protocol collection, we present exemplary data, however, experiments should be performed on at least three biological replicates of human alveolar-like organoids derived from different donors.

thumbnail
Fig 5. Spectral imaging of mock- and SARS-CoV-2-infected and immunostained human alveolar-like organoids and in situ hybridization.

(A) Representative immunostainings for exemplary mock- (left panel) and SARS-CoV-2-infected (right column) human alveolar-like organoids. Shown are immunostainings for SARS-CoV-2 (N-protein, red), ACE2 (green) and via in situ hybridization visualized ACE2 mRNA expression (red dots) 24 h post infection (MOI = 1). Arrows indicate either cells positive for SARS-CoV-2 (red arrows) or areas of particularly high ACE2 expression (protein: green arrows, mRNA: red dotted arrows). Cell nuclei are visualized by DAPI stain (blue). Scale bars = 20 μm. (B) In situ hybridization of human alveolar-like organoids shows SARS-CoV-2 mRNA expression (left column). DapB and EF1a served as negative respectively positive control. Red arrows indicate cells positive for SARS-CoV-2. Scale bars = 20 μm.

https://doi.org/10.1371/journal.pone.0276115.g005

Supporting information

S1 File. Collection: State-of-the-art analytical methods of viral infections in human lung organoids.

https://doi.org/10.1371/journal.pone.0276115.s001

(PDF)

S2 File. Step-by-step protocol: Protein extraction and western blot of human lung organoids.

https://doi.org/10.1371/journal.pone.0276115.s002

(PDF)

S3 File. Step-by-step protocol: RNA Extraction and RT-qPCR of human lung organoids.

https://doi.org/10.1371/journal.pone.0276115.s003

(PDF)

S4 File. Step-by-step protocol: SARS-CoV-2 infection and viral replication of human lung organoids.

https://doi.org/10.1371/journal.pone.0276115.s004

(PDF)

S5 File. Step-by-step protocol: Single cell isolation of human lung organoids.

https://doi.org/10.1371/journal.pone.0276115.s005

(PDF)

S6 File. Step-by-step protocol: Fixation, immunohistochemistry and in situ hybridization of human lung organoids.

https://doi.org/10.1371/journal.pone.0276115.s006

(PDF)

Acknowledgments

This work is part of the doctoral thesis of M.B.

We thank Tracey Weissgerber and René Bernard for their guidance in the ’publish my protocol’ workshop at BIH QUEST Center.

References

  1. 1. Adhikary PP, Ul Ain Q, Hocke AC, Hedtrich S. COVID-19 highlights the model dilemma in biomedical research. Nat Rev Mater. 2021;6(5):374–6. pmid:33747552
  2. 2. Kang I, Smirnova L, Kuhn JH, Hogberg HT, Kleinstreuer NC, Hartung T. COVID-19—prime time for microphysiological systems, as illustrated for the brain. ALTEX. 2021;38(4):535–49. pmid:34698363
  3. 3. Munoz-Fontela C, Widerspick L, Albrecht RA, Beer M, Carroll MW, de Wit E, et al. Advances and gaps in SARS-CoV-2 infection models. PLoS Pathog. 2022;18(1):e1010161. pmid:35025969
  4. 4. Hoffmann K, Obermayer B, Honzke K, Fatykhova D, Demir Z, Lowa A, et al. Human alveolar progenitors generate dual lineage bronchioalveolar organoids. Commun Biol. 2022;5(1):875. pmid:36008580
  5. 5. Clevers H. Modeling Development and Disease with Organoids. Cell. 2016;165(7):1586–97. pmid:27315476
  6. 6. van der Vaart J, Lamers MM, Haagmans BL, Clevers H. Advancing lung organoids for COVID-19 research. Dis Model Mech. 2021;14(6). pmid:34219165
  7. 7. Dutta D, Clevers H. Organoid culture systems to study host-pathogen interactions. Curr Opin Immunol. 2017;48:15–22. pmid:28756233
  8. 8. Blutt SE, Estes MK. Organoid Models for Infectious Disease. Annu Rev Med. 2022;73:167–82. pmid:34644153
  9. 9. Mboko WP, Chhabra P, Valcarce MD, Costantini V, Vinje J. Advances in understanding of the innate immune response to human norovirus infection using organoid models. J Gen Virol. 2022;103(1). pmid:35077345
  10. 10. Su X, Yue P, Kong J, Xu X, Zhang Y, Cao W, et al. Human Brain Organoids as an In Vitro Model System of Viral Infectious Diseases. Front Immunol. 2021;12:792316. pmid:35087520
  11. 11. Youk J, Kim T, Evans KV, Jeong YI, Hur Y, Hong SP, et al. Three-Dimensional Human Alveolar Stem Cell Culture Models Reveal Infection Response to SARS-CoV-2. Cell Stem Cell. 2020;27(6):905–19 e10.
  12. 12. Sasaki M, Toba S, Itakura Y, Chambaro HM, Kishimoto M, Tabata K, et al. SARS-CoV-2 Bearing a Mutation at the S1/S2 Cleavage Site Exhibits Attenuated Virulence and Confers Protective Immunity. mBio. 2021;12(4):e0141521. pmid:34425707
  13. 13. Corman VM, Landt O, Kaiser M, Molenkamp R, Meijer A, Chu DK, et al. Detection of 2019 novel coronavirus (2019-nCoV) by real-time RT-PCR. Euro Surveill. 2020;25(3).
  14. 14. Hoffmann M, Kleine-Weber H, Schroeder S, Kruger N, Herrler T, Erichsen S, et al. SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor. Cell. 2020;181(2):271–80 e8. pmid:32142651
  15. 15. Honzke K, Obermayer B, Mache C, Fathykova D, Kessler M, Dokel S, et al. Human lungs show limited permissiveness for SARS-CoV-2 due to scarce ACE2 levels but virus-induced expansion of inflammatory macrophages. Eur Respir J. 2022. pmid:35728978
  16. 16. Hocke AC, Suttorp N, Hippenstiel S. Human lung ex vivo infection models. Cell Tissue Res. 2017;367(3):511–24. pmid:27999962
  17. 17. Dirnagl U, Kurreck C, Castanos-Velez E, Bernard R. Quality management for academic laboratories: burden or boon? Professional quality management could be very beneficial for academic research but needs to overcome specific caveats. EMBO Rep. 2018;19(11). pmid:30341068
  18. 18. Dowden H, Munro J. Trends in clinical success rates and therapeutic focus. Nat Rev Drug Discov. 2019;18(7):495–6. pmid:31267067
  19. 19. Hulsemann M, Wiebach J, Drude NI, Kniffert S, Behm L, Honzke K, et al. Introducing quality measures in an academic research consortium: Lessons and recommendation from implementing an ad hoc quality management system for organ model research: Lessons and recommendation from implementing an ad hoc quality management system for organ model research. EMBO Rep. 2022;23(8):e55095. pmid:35876564
  20. 20. Travaglini KJ, Nabhan AN, Penland L, Sinha R, Gillich A, Sit RV, et al. A molecular cell atlas of the human lung from single-cell RNA sequencing. Nature. 2020;587(7835):619–25. pmid:33208946
  21. 21. Dalton S. Linking the Cell Cycle to Cell Fate Decisions. Trends Cell Biol. 2015;25(10):592–600. pmid:26410405