Summary

Organoids som model for infektionssygdomme: Kultur i humane og murine mave Organoids og Mikroinjektion af 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

Undersøgelsen af patogener beror på passende modelsystemer for at efterligne in vivo-infektion. For nogle smitstoffer, er tilstrækkelige modelsystemer mangler, mens nogle af de anvendte systemer er langt fra optimal. Et eksempel er den gastriske bakterien Helicobacter pylori (H. pylori), som er kausalt relateret til udviklingen af mavekræft. Men i mangel af en mere egnet cellekultur-system, at mange undersøgelser, der sigter analysere de molekylære mekanismer bag udviklingen af ​​kræft brug cancer cellelinjer, som repræsenterer endepunkt af kræft kaskade. Primære, ikke-transformerede celler vil være en bedre model for disse undersøgelser. Imidlertid er kun tilgængelige fra et lille antal donorer primære celler og kan ikke dyrkes over længere perioder. I de senere år har forskning i stamceller gjort betydelige fremskridt at tilvejebringe nye kilder til primære cellekulturer til studiet af infektionsbiologi.

Kulturer fratre stamceller kilder er blevet anvendt: Embryonale stamceller (ESC), inducerede pluripotente stamceller (IPSC) eller voksne stamceller. De er blevet brugt til at modellere infektioner med vira, såsom cytomegalovirus 1,2 eller hepatitis C-virus 3-7, parasitter, såsom Plasmodium falciparum 8 eller Toxoplasma 9, og bakterier, såsom Bacterioides thetaiotaomicron 10 eller Salmonella enterica 11. Senest har flere tilgange blevet offentliggjort at modellere infektion med H. pylori med organoids afledt af ESC eller iPS celler 12, mus voksne stamceller 21,22 eller humane voksne stamceller 13 – 15.

Udviklingen af ​​organoide kulturer fra voksne stamceller stammer fra en undersøgelse, hvor enkelt stamceller isoleret fra murine intestinale epithel blev podet i en 3-dimensionel matrix ogindlejret i medium, der efterlignede miljøet i de intestinale celler, der indeholder stamceller EGF som mitogen, R-spondin kan forbedre Wnt signalering og Noggin at inhibere knoglemorfogenetisk protein (BMP) signalering 16. Især disse kulturer kræver ikke co-kultur med mesenchymale celler. Under disse omstændigheder, stamcellerne prolifererer og danner små strukturer med domæner huser celler af tarmens krypter, og domæner, der indeholder cellerne i tarmen villus. De organoids således selvorganiserende at efterligne in vivo-situationen. I dag kan voksne stamceller fra mange murine og humane væv dyrkes in vitro og selv-organisere sig i organoids der ligner deres in vivo-modstykke, såsom tyndtarmen og tyktarmen 17, mave 13,18, lever 19,20, bugspytkirtel 21 og prostata 22.

Her giver vi en video-protokol til kultur mus eller humane gastriske organoids fra voksne stamceller cells og microinject dem med H. pylori. Denne protokol er baseret på tidligere rapporter 13,18. Denne metode kan tilpasses til dyrkning og inficere andre organoide kulturer såsom tarm organoids.

Protocol

1. Etablering af gastrisk organoide Kultur Bemærk: Denne protokol kan anvendes til isolering af gastriske kirtler fra mus eller humant væv. Det tilrådes at bruge væv på ca. 1 cm. Humant væv kan fås fra gastriske resektioner eller biopsier. Fremstilling af materiale Bemærk: Den anvendte kælderen matrix er Matrigel. Hold kælderen matrix på is på alle tidspunkter. Opbevar kælderen matrix ved -20 ° C og optøs på is før anvendelse. Basal medium henviser til Adva…

Representative Results

Denne protokol tillader isolering af gastriske kirtler (figur 2). Kirtler podes i kælderen matrix, der størkner som drop i en brønd, hvilket giver en 3-dimensionel ramme rig på laminin og kollagen at tillade kirtler vokse ind organoids (figur 3). Organoids starter typisk som små cyster og inden 12-16 dage, udvider de sig til kugler med en diameter på 50-300 um (figur 4). Nogle organoids vil forblive cystisk, nogle vil udvikle små buddings. Sidstnævnte er normalt…

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…

Divulgazioni

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.
Altro
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

Riferimenti

  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/it/53359?article_type=t

Play Video

Citazione di questo articolo
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