Summary

Organoidi come modello per le malattie infettive: cultura dei diritti dell'uomo e organoidi stomaco murini e microiniezione dell'Helicobacter Pylori

Published: November 12, 2015
doi:

Summary

Stem cell derived cultures harbor tremendous potential to model infectious diseases. Here, the culture of mouse and human gastric organoids derived from adult stem cells is described. The organoids are microinjected with the gastric pathogen Helicobacter pylori.

Abstract

Recently infection biologists have employed stem cell derived cultures to answer the need for new and better models to study host-pathogen interactions. Three cellular sources have been used: Embryonic stem cells (ESC), induced pluripotent stem cells (iPSC) or adult stem cells. Here, culture of mouse and human gastric organoids derived from adult stem cells is described and used for infection with the gastric pathogen Helicobacter pylori. Human gastric glands are isolated from resection material, seeded in a basement matrix and embedded in medium containing growth factors epidermal growth factor (EGF), R-spondin, Noggin, Wnt, fibroblast growth factor (FGF) 10, gastrin and transforming growth factor (TGF) beta inhibitor. In these conditions, gastric glands grow into 3-dimensional organoids containing 4 lineages of the stomach. The organoids expand indefinitely and can be frozen and thawed similarly as cell lines. For infection studies, bacteria are microinjected into the lumen of the organoids. Infected organoids are processed for imaging. The described methods can be adapted to other organoids and infections with other bacteria, viruses or parasites. This allows the study of infection-induced changes in primary cells.

Introduction

Lo studio di patogeni si basa su adeguati sistemi modello per imitare l'infezione in vivo. Per alcuni agenti infettivi, adeguati sistemi modello sono carenti, mentre alcuni dei sistemi utilizzati sono ben lungi dall'essere ottimale. Un esempio è il batterio gastrico Helicobacter pylori (H. pylori), che è causalmente legata alla sviluppo del cancro gastrico. Tuttavia, in assenza di un sistema di coltura cellulare più adatto, molti studi che mirano ad analizzare i meccanismi molecolari alla base dello sviluppo del cancro linee cellulari tumorali uso, che rappresentano il punto finale della cascata cancerose. , Cellule non trasformate primarie sarebbe un modello migliore per questi studi. Tuttavia, le cellule primarie sono disponibili solo da un piccolo numero di donatori e non possono essere coltivate su periodi di tempo più lunghi. Negli ultimi anni, la ricerca sulle cellule staminali ha compiuto progressi significativi per fornire nuove fonti per colture cellulari primarie per lo studio della biologia infezione.

Culture disono state utilizzate tre fonti di cellule staminali: le cellule staminali embrionali (ESC), le cellule indotte pluripotenti staminali (IPSC) o le cellule staminali adulte. Essi sono stati utilizzati per modellare infezioni da virus, come il citomegalovirus 1,2 o virus dell'epatite C 3 – 7, parassiti, come Plasmodium falciparum 8 o 9 Toxoplasma gondii, e batteri, quali Bacterioides thetaiotaomicron 10 o Salmonella enterica 11. Più di recente, diversi approcci sono stati pubblicati per modellare l'infezione da H. pylori con organoidi derivati ​​da ESC o iPS cellule 12, le cellule staminali adulte del mouse 21,22 o umani cellule staminali adulte di 13 – 15.

Lo sviluppo delle colture organoide da cellule staminali adulte origine da uno studio, in cui le cellule staminali singole isolate da murino epitelio intestinale sono state seminate in una matrice 3-dimensionale eincorporato in mezzo che imitava l'ambiente delle cellule staminali intestinali che contengono FEG mitogeno, R-spondina per migliorare la segnalazione Wnt e Noggin di inibire la proteina morfogenetica dell'osso (BMP) di segnalazione 16. In particolare queste colture non richiedono co-coltura con cellule mesenchimali. In queste condizioni, le cellule staminali proliferano e formano piccole strutture con domini ospitare cellule delle cripte intestinali, e domini che contengono le cellule del villi intestinali. I organoidi quindi auto-organizzano per simulare la situazione in vivo. Oggi, le cellule staminali adulte da molte murino e tessuti umani possono essere coltivate in vitro e auto-organizzarsi in organoidi che ricordano la loro controparte in vivo, come l'intestino tenue e colon 17, stomaco 13,18, fegato 19,20, pancreas e 21 22 della prostata.

Qui forniamo un protocollo video al topo cultura o organoidi gastriche umane da cel staminali adultels e li microinject con H. pylori. Questo protocollo si basa sulle precedenti relazioni 13,18. Questo metodo può essere adattato per la coltura e infettare altre culture organoide come organoidi intestinali.

Protocol

1. Istituzione di gastrico organoide Cultura Nota: Questo protocollo può essere utilizzato per l'isolamento delle ghiandole gastriche da mouse o tessuti umani. Si consiglia di usare il tessuto di circa 1 cm². Tessuto umano può essere ottenuto da resezioni gastriche o biopsie. Preparazione del materiale Nota: La matrice seminterrato utilizzato è Matrigel. Mantenere la matrice seminterrato su ghiaccio in ogni momento. Conservare la matrice seminterrato a -20 ° C e sc…

Representative Results

Questo protocollo permette isolamento delle ghiandole gastriche (Figura 2). Ghiandole vengono seminate in matrice seminterrato, che solidifica come goccia all'interno di un pozzo, fornendo una struttura tridimensionale 3 ricco di laminina e collagene per consentire le ghiandole crescono in organoidi (Figura 3). Organoidi tipicamente iniziano come piccole cisti e entro 12-16 giorni, si espandono in sfere con un diametro di 50-300 micron (Figura 4). Alcuni organoidi r…

Discussion

This protocol describes the use of ever-expanding, untransformed primary organoids from adult stem cells for infection biology. Critical steps are i) the isolation of viable glands, ii) expansion of organoids and iii) the microinjection. Below are some suggestions for modifications, troubleshooting and technical considerations.

Compared to other isolation methods, which use vigorous shaking or pipetting to release glands and can be equally successful, the technique presented here has the adva…

Disclosures

The authors have nothing to disclose.

Acknowledgements

This work was supported by EU Marie Curie Fellowship (EU/300686-InfO) to S.B. and a Research Prize from the United European Gastroenterology Foundation to H.C. We would like to thank Harry Begthel, Jeroen Korving and the Hubrecht Imaging Center for technical assistance, Meritxell Huch for help with initial organoid culture and Yana Zavros for discussion.

Materials

Medium
HEPES Invitrogen 15630-056
Advanced DMEM/F12 Invitrogen 12634-028
Matrigel, GFR, phenol free BD 356231
GlutaMAX Invitrogen 35050-079 Stock concentration 200 mM, final concentration 2 mM
B27 Invitrogen 17504-044 Stock concentration 50 x, final concentration 1x
N-Acetylcysteine Sigma-Aldrich A9165-5G Stock concentration 500 mM, final concentration 1 mM
Murine recombinant EGF Invitrogen PMG8043 Stock concentration 500 µg/mL, final concentration 50 ng/mL
Human recombinant FGF10 Peprotech 100-26 Stock concentration 100 µg/mL, final concentration 200 ng/mL
TGFβi A-83-01 Tocris 2939 Stock concentration 500 µM, final concentration 2 µM 
Nicotinamide Sigma-Aldrich N0636 Stock concentration 1 M, final concentration 10 mM 
[Leu15]-Gastrin Sigma-Aldrich G9145 Stock concentration 100 µM, final concentration 1 nM
RHOKi Y-27632 Sigma-Aldrich Y0503 Stock concentration 10 mM, final concentration 10 µM
Wnt3A conditioned medium Stable cell line generated in the Clevers Lab. Final concentration 50%. Cells can be obtained from Hans Clevers.
R-spondin1 conditioned medium Stable cell line generated in the Kuo Lab. Final concentration 10%. Cell line can be obtained from Calvin Kuo, Stanford.
Noggin conditioned medium Stable cell line generated in the Clevers Lab. Final concentration 10%. Cells can be obtained from Hans Clevers.
R-spondin3 R&D 3500-RS/CF Alternative source for R-spondin. This has been tested on human intestine organoids (1 µg/mL), but not yet on gastric organoids.
Noggin Peprotech 120-10 Alternative source for noggin. This has been tested on human intestine organoids (100 ng/mL), but not yet on gastric organoids.
TrypLE express Life Technologies 12605036 Enzymatic dissociation solution 
CoolCell® Alcohol-Free Cell Freezing Containers biocision BCS-405
Recovery Cell Culture Freezing Medium Invitrogen 12648-010
Antibiotics
Primocin Invivogen ant-pm-1 An antibiotics composition agains bacteria and fungi. It is helpful after initiation of a culture. For long term culture you can switch to other antibiotics or none.
Penicillin/Streptomycin Invitrogen 15140-122 Stock concentration 10000/10000 U/mL, final concentration 100/100 U/mL. Can be used alternatively to Primocin in long term culture.
Other
Tweezers Neolabs 2-1033 Tweezers with fine tips are helpful for the removal of muscle layer from the tissue.
4 Well Multidishes Thermo Scientific 144444 You can use other Multidishes. These were particularly helpful for microinjections because they have a low outer rim and allow more mobility for the manipulator.
Micromanipulator Narishige M-152
Microinjector Narishige IM-5B
Stereomicroscope Leica MZ75
Workbench Clean Air Custom made to fit the stereomicroscope in ML2 condition
Cappillaries Harvard Apparatus GC100T-10 1 mm outer diameter, 0,78 mm inner diameter.
Micropipette Puller Sutter Instruments Flaming Brown Micropipette Puller
anti Cag A antibody Santa Cruz sc-25766

References

  1. Aiuto, L., et al. Human Induced Pluripotent Stem Cell-Derived Models to Investigate Human Cytomegalovirus Infection in Neural Cells. PLoS ONE. 7 (11), e49700 (2012).
  2. Penkert, R. R., Kalejta, R. F. Human Embryonic Stem Cell Lines Model Experimental Human Cytomegalovirus Latency. mBio. 4 (3), e00298-13-e00298-13 (2013).
  3. Roelandt, P., et al. Human pluripotent stem cell-derived hepatocytes support complete replication of hepatitis C virus. J Hepatol. 57 (2), 246-251 (2012).
  4. Schwartz, R. E., Trehan, K., et al. Modeling hepatitis C virus infection using human induced pluripotent stem cells. Proc Natl Acad Sci USA. 109 (7), 2544-2548 (2012).
  5. Shlomai, A., et al. Modeling host interactions with hepatitis B virus using primary and induced pluripotent stem cell-derived hepatocellular systems. Proc Natl Acad Sci USA. 111 (33), 12193-12198 (2014).
  6. Wu, X., et al. Productive Hepatitis C Virus Infection of Stem Cell-Derived Hepatocytes Reveals a Critical Transition to Viral Permissiveness during Differentiation. PLoS Pathogens. 8 (4), e1002617 (2012).
  7. Yoshida, T., et al. Use of human hepatocyte-like cells derived from induced pluripotent stem cells as a model for hepatocytes in hepatitis C virus infection. Biochem Biophys Res Commun. 416 (1-2), 119-124 (2011).
  8. Ng, S., et al. Human iPSC-Derived Hepatocyte-like Cells Support Plasmodium Liver-Stage Infection In Vitro. Stem Cell Report. 4 (2), (2015).
  9. Klotz, C., Aebischer, T., Seeber, F. Stem cell-derived cell cultures and organoids for protozoan parasite propagation and studying host-parasite interaction. Int J Med Microbiol. 302 (4-5), 203-209 (2012).
  10. Engevik, M. A., et al. Loss of NHE3 alters gut microbiota composition and influences Bacteroides thetaiotaomicron growth. AJP: GI. 305 (10), G697-G711 (2013).
  11. Wilson, S. S., Tocchi, A., Holly, M. K., Parks, W. C., Smith, J. G. A small intestinal organoid model of non-invasive enteric pathogen-epithelial cell interactions. Mucosal Immunol. 8 (2), 352-361 (2015).
  12. McCracken, K. W., et al. Modelling human development and disease in pluripotent stem-cell-derived gastric organoids. Nature. 516 (7531), 400-404 (2014).
  13. Bartfeld, S., et al. In Vitro Expansion of Human Gastric Epithelial Stem Cells and Their Responses to Bacterial Infection. Gastroenterology. 148 (1), (2014).
  14. Schlaermann, P., Toelle, B., et al. A novel human gastric primary cell culture system for modelling Helicobacter pylori infection in vitro. Gut. , (2014).
  15. Bertaux-Skeirik, N., et al. CD44 Plays a Functional Role in Helicobacter pylori-induced Epithelial Cell Proliferation. PLOS Pathogens. 11 (2), e1004663 (2015).
  16. Sato, T., et al. Single Lgr5 stem cells build crypt villus structures in vitro without a mesenchymal niche. Nature. 459 (7244), 262-265 (2009).
  17. Sato, T., et al. Long-term Expansion of Epithelial Organoids From Human Colon, Adenoma, Adenocarcinoma, and Barrett’s Epithelium. Gastroenterology. 141 (5), 1762-1772 (2011).
  18. Barker, N., et al. Lgr5+ve Stem Cells Drive Self-Renewal in the Stomach and Build Long-Lived Gastric Units In Vitro. Cell Stem Cell. 6 (1), 25-36 (2010).
  19. Huch, M., et al. In vitro expansion of single Lgr5+ liver stem cells induced by Wnt-driven regeneration. Nature. 494 (7436), 247-250 (2013).
  20. Huch, M., et al. Long-Term Culture of Genome-Stable Bipotent Stem Cells from Adult Human Liver. Cell. 160 (1-2), 299-312 (2015).
  21. Boj, S. F., et al. Organoid Models of Human and Mouse Ductal Pancreatic Cancer. Cell. 160 (1-2), 324-338 (2015).
  22. Karthaus, W. R., et al. Identification of multipotent luminal progenitor cells in human prostate organoid cultures. Cell. 159 (1), 163-175 (2014).
  23. Bartfeld, S., et al. High-throughput and single-cell imaging of NF-kappaB oscillations using monoclonal cell lines. BMC cell. 11, 21 (2010).
  24. Blanchard, T. G., Nedrud, J. G. Laboratory Maintenance of Helicobacter Species. Curr Protoc Microbiol. , (2006).
  25. Van Es, J. H., de Geest, N., van de Born, M., Clevers, H., Hassan, B. A. Intestinal stem cells lacking the Math1 tumour suppressor are refractory to Notch inhibitors. Nat Commun. 1 (2), 1-5 (2010).
  26. Andersson-Rolf, A., Fink, J., Mustata, R. C., Koo, B. -. K. A Video Protocol of Retroviral Infection in Primary Intestinal Organoid Culture. J Vis Exp. (90), (2014).
  27. Stange, D. E., Koo, B. -. K., et al. Differentiated Troy+ Chief Cells Act as Reserve Stem Cells to Generate All Lineages of the Stomach Epithelium. Cell. 155 (2), 357-368 (2013).
  28. Van de Wetering, M., Sancho, E., et al. The beta-catenin/TCF-4 complex imposes a crypt progenitor phenotype on colorectal cancer cells. Cell. 111 (2), 241-250 (2002).
  29. Schumacher, M. A., Aihara, E., et al. The use of murine-derived fundic organoids in studies of gastric physiology. Journal Physiol. 593 (8), 1809-1827 (2015).
  30. Schwank, G., Andersson-Rolf, A., Koo, B. -. K., Sasaki, N., Clevers, H. Generation of BAC Transgenic Epithelial Organoids. PLoS ONE. 8 (10), e76871 (2013).
  31. Koo, B. -. K., et al. Controlled gene expression in primary Lgr5 organoid cultures. Nat Meth. 9 (1), 81-83 (2012).
  32. Schwank, G., et al. Functional repair of CFTR by CRISPR/Cas9 in intestinal stem cell organoids of cystic fibrosis patients. Cell Stem Cell. 13 (6), 653-658 (2013).
  33. Li, V. S. W., Ng, S. S., et al. Wnt Signaling through Inhibition of β-Catenin Degradation in an Intact Axin1 Complex. Cell. 149 (6), 1245-1256 (2012).
  34. Van de Wetering, M., et al. Prospective derivation of a ‘Living Organoid Biobank’ of colorectal cancer patients. Cell. 161 (4), 933-945 (2015).
check_url/53359?article_type=t

Play Video

Cite This Article
Bartfeld, S., Clevers, H. Organoids as Model for Infectious Diseases: Culture of Human and Murine Stomach Organoids and Microinjection of Helicobacter Pylori. J. Vis. Exp. (105), e53359, doi:10.3791/53359 (2015).

View Video