Summary

Ex vivo Levende Imaging af lungemetastase og deres mikromiljø

Published: February 03, 2016
doi:

Summary

Vi beskriver en forholdsvis enkel fremgangsmåde til ex vivo billeddannelse af tumor celle-stroma interaktioner inden lunge metastase, udnytte fluorescerende reportere i mus. Brug af spinning-disk konfokal mikroskopi, denne teknik muliggør visualisering af levende celler i mindst 4 timer, og kan tilpasses til at studere andre inflammatoriske lungesygdomme.

Abstract

Metastase er en væsentlig årsag til cancer-relaterede sygelighed og dødelighed. Metastase er en flertrins proces, og på grund af sin kompleksitet, de nøjagtige cellulære og molekylære processer, der styrer metastatisk spredning og vækst er stadig undvigende. Live-imaging tillader visualisering af de dynamiske og rumlige interaktioner af celler og deres mikromiljø. Faste tumorer almindeligvis metastaserer til lungerne. Men den anatomiske placering af lungerne udgør en udfordring for intravital billeddannelse,. Denne protokol giver en forholdsvis enkel og hurtig metode til ex vivo levende billeddannelse af de dynamiske interaktioner mellem tumorceller og deres omgivende stroma inden lunge metastase. Under anvendelse af denne fremgangsmåde kan motilitet af cancerceller samt vekselvirkninger mellem cancerceller og stromale celler i deres mikromiljø visualiseres i realtid i flere timer. Ved at anvende transgene fluorescerende reporter mus, en fluorescerende cellelinie, injicerbar fluorescensmærketmolekyler og / eller antistoffer, flere komponenter af lungen mikromiljø kan visualiseres, såsom blodkar og immunceller. At afbilde forskellige celletyper, har en roterende skive konfokalt mikroskop, der tillader langvarig, kontinuerlig billeddannelse med hurtig, firefarvet billedregistrering blevet anvendt. Time-lapse film kompileret fra billeder indsamlet over flere positioner og fokalplaner viser samspillet mellem levende metastatisk og immunceller i mindst 4 timer. Denne teknik kan yderligere anvendes til at teste kemoterapi eller målrettet terapi. Desuden kunne denne fremgangsmåde tilpasses til studiet af andre lunge-relaterede patologier, der kan påvirke lunge mikromiljø.

Introduction

The deadliest aspect of cancer is metastasis, which accounts for more than 90% of cancer-related morbidity and mortality1. Metastasis is a multistep process and due to its complexity, the exact cellular and molecular mechanisms that govern metastatic dissemination and growth are still elusive. To metastasize, tumor cells in the primary tumor must detach from their neighboring cells and basement membrane, cross through the extracellular matrix, intravasate, travel via blood or lymphatic vessels, extravasate at the secondary site, and finally, survive and establish secondary tumors. In addition to the properties of the tumor cells, the contribution from the microenvironment, which includes the adjacent stroma along with the normal counterparts of the cancer cells, is crucial for the seeding and establishment of metastatic lesions2.

Traditional methods to study metastatic seeding and growth examine static states, as tissues are excised and sectioned for histology. These data only generate a snapshot of this highly dynamic process. Although some useful information can be gained from these studies, the complicated process by which tumor and stromal cells interact during metastatic formation cannot be adequately assessed by these methods. Furthermore, it is not possible to gain insights into tumor or stromal cell migration patterns, which are important in establishing a colony at the distant site. In order to effectively study the metastatic process, it is essential to visualize various interactions between cancer cells and their microenvironment in a continuous manner and at real time.

The lung is a common site for metastases from solid tumors as breast, colorectal, pancreatic cancer, melanoma and sarcoma3. Intravital imaging was previously used to study cell-cell interaction in various primary tumor and metastatic models4,5. Methods of lung imaging in mice, including intravital imaging, lung section imaging, and an ex vivo pulmonary metastasis assay have been published6–9. Intravital imaging of mouse lungs utilizes a thoracic suction window to stabilize the lungs6. This method is used for time-lapse imaging of the lung microcirculation and alveolar spaces. The anatomical location of the lungs poses a challenge to intravital imaging. In order to access the lungs, the chest cavity must be opened which leads to loss of negative pressure and collapsed lungs. This method only allows the visualization of a small part of the lungs and is technically demanding; an unnecessary complication in studies that examine processes that are independent of blood flow. Moreover, this method also requires gating out movement caused by breathing. This is done either by collecting images between breaths or during post image acquisition analyses10. The alternative ex vivo lung section imaging provides stability and depth, and also prepares lung parenchyma for immunostaining7. However, the lengthy sectioning process leads to an extensive delay between the time of animal sacrifice and the start of the imaging session. Moreover, the process of sectioning a mouse lung causes considerable amount of cell death8, thus interfering with the quality and quantity of imaging samples and perhaps needlessly altering tumor-stroma interactions. In order to technically bridge between the methods of intravital imaging and lung section imaging, while exploiting the advantages of the two techniques, a relatively fast and easy method for ex vivo lung imaging was developed. This method was achieved by imaging of non-sectioned whole lung lobes. Using this method, the motility of cancer cells as well as interactions between cancer cells and stromal cells in their microenvironment can be visualized in real time for several hours.

Protocol

Alle beskrevne procedurer skal udføres i overensstemmelse med retningslinjer og regler for brug af hvirveldyr, herunder forudgående godkendelse af den lokale Institutional Animal Care og brug Udvalg (IACUC). 1. generation af lungemetastaser for Ex vivo Live-Imaging (Transgene eller haleveneinjektion) BEMÆRK: lungemetastaser kan genereres ved anvendelse af genetisk manipulerede musemodeller eller ved intravenøs (iv) injektion af cancerceller. <…

Representative Results

Brug spinning-disk konfokalmikroskopi, diverse mus modelsystemer og injectables, kan det metastatiske mikromiljø visualiseres og spores over tid. Ved hjælp af en MMTV-PyMT; ACTB-ECFP; c-fms-EGFP triple transgen musemodel, forskellige cellulære komponenter er fluorescensmærket (Figur 2A, Movie 1). Den typiske struktur af lungeparenkym kan visualiseres i FFP kanalen da alle celler udtrykker ECFP under β-actinpromotoren. Større / flercellede lungemetastaser let løst så dis…

Discussion

Dette håndskrift beskriver en detaljeret metode til ex vivo levende billeddannelse af lunge metastase i musemodeller af metastaser. Denne billedbehandling protokol giver en direkte visualisering af de dynamiske og rumlige tumor celle-stroma interaktioner inden lungerne mikromiljø. Det er en forholdsvis let og hurtig metode, der tillader pålidelig billeddannelse af lungemetastase i mindst 4 timer. Film erhvervet fra disse forsøg kan anvendes til at spore dynamiske processer som cellemotilitet og cellulære i…

Disclosures

The authors have nothing to disclose.

Acknowledgements

We thank Nguyen H. Nguyen for her technical help and Audrey O’Neill for support with the Zeiss Cell Observer spinning-disk confocal microscope. This work was supported by a Department of Defense postdoctoral fellowship (W81XWH-11-01-0139) and the Weizmann Institute of Science-National Postdoctoral Award Program for Advancing Women in Science (to V.P.).

Materials

MMTV-PyMT/FVB mice Jackson Laboratory 2374 Female mice
ACTB-ECFP/FVB mice UCSF Werb lab Female mice
c-fms-EGFP/FVB mice UCSF Werb lab Female mice
FVB mice Jackson Laboratory 1800 Female mice
GFP+ VO-PyMT cells UCSF Werb lab
70,000 kDa Dextran, rhodamine-conjugated Invitrogen D1818 Dilute to  4mg/ml in 1 x PBS and store at -20  °C. Use 0.4 mg per animal. 
10,000 kDa Dextran, Alexa Fluor 647 conjugated Invitrogen D22914 Dilute to  4mg/ml in 1 x PBS and store at -20  °C. Use 0.4 mg per animal. 
Anti-mouse Gr-1 antibody Alexa Fluor 647 UCSF Monoclonal antibody core Stock 1mg/ml. Use 7 ug per animal.
Anesthetic Anesthesia approved by IACUC, used for anesthesia and/or euthanesia
1X PBS UCSF cell culture facility
PBS, USP sterile  Amresco INC K813-500ML Ultra pure grade for i.v. injection
Styrofoam platform Will be used as dissection board
Fine scissors sharp  Fine Science Tools 14060-11
Forceps Roboz Surgical Store RS-5135
Hot bead sterilizer Fine Science Tools 18000-45 Turn ON 30min before use
Air UCSF
Oxygen UCSF
Carbon dioxide UCSF
1 mL syringe without needle  BD 309659
27 G x 1/2 needle   BD 305109 for i.v. injection
20 G x 1 needle, short bevel   BD 305178
Low-melting-temperature agarose  Lonza 50111 To make 10 ml of solution, weigh 0.2 g of agarose, add to 10 ml 1 x PBS, and heat to dissolve. Agarose will solidify at room temperature, so maintain in a 37 °C water bath until used for inflation.
RPMI-1640 medium without phenol red Life Technologies 11835-030
24 well Imaging plate  E&K scientific EK-42892
Glass cover slides, 15 mm  Fisher Scientific 22-031-144
Digital CO2 and temperature controller Okolab DGTCO2BX http://www.oko-lab.com
Climate chamber Okolab http://www.oko-lab.com
Cell Observer spinning disk confocal microscope Zeiss
Zen software Zeiss
Inverted microscope Carl Zeiss Inc Zeiss Axiovert 200M
ICCD camera Stanford Photonics XR-Mega-10EX S-30
Spinning disk confocal scan-head Yokogawa Corporation CSU-10b
Imaris Bitplane
mManager Vale lab, UCSF Open-source software

References

  1. Chaffer, C. L., & Weinberg, R. A. A perspective on cancer cell metastasis. Science. 331 (6024), 1559–1564 (2011).
  2. Plaks, V., Kong, N., & Werb, Z. The cancer stem cell niche: how essential is the niche in regulating stemness of tumor cells? Cell stem cell. 16 (3), 225–238 (2015).
  3. Nguyen, D. X., Bos, P. D., & Massague, J. Metastasis: from dissemination to organ-specific colonization. Nat Rev Cancer. 9 (4), 274–284 (2009).
  4. Egeblad, M., Ewald, A. J., et al. Visualizing stromal cell dynamics in different tumor microenvironments by spinning disk confocal microscopy. Dis Model Mech. 1 (2-3), 155–167; discussion 165 (2008).
  5. Ellenbroek, S. I. J., & van Rheenen, J. Imaging hallmarks of cancer in living mice. Nat Rev Cancer. 14 (6), 406–418 (2014).
  6. Looney, M. R., Thornton, E. E., Sen, D., Lamm, W. J., Glenny, R. W., & Krummel, M. F. Stabilized imaging of immune surveillance in the mouse lung. Nat Methods. 8 (1), 91–96 (2011).
  7. Thornton, E. E., Krummel, M. F., & Looney, M. R. Live imaging of the lung. Cur Protoc Cytom. Chapter 12 (2012).
  8. Thornton, E. E., Looney, M. R., et al. Spatiotemporally separated antigen uptake by alveolar dendritic cells and airway presentation to T cells in the lung. J Exp Med. 209 (6), 1183–1199 (2012).
  9. Mendoza, A., Hong, S.-H., et al. Modeling metastasis biology and therapy in real time in the mouse lung. J Clin Invest. 120 (8), 2979–2988 (2010).
  10. Lelkes, E., Headley, M. B., Thornton, E. E., Looney, M. R., & Krummel, M. F. The spatiotemporal cellular dynamics of lung immunity. Trends Immunol. 35 (8), 379–386 (2014).
  11. Guy, C. T., Cardiff, R. D., & Muller, W. J. Induction of mammary tumors by expression of polyomavirus middle T oncogene: a transgenic mouse model for metastatic disease. Mol Cell Biol. 12 (3), 954–961 (1992).
  12. Hadjantonakis, A.-K., Macmaster, S., & Nagy, A. Embryonic stem cells and mice expressing different GFP variants for multiple non-invasive reporter usage within a single animal. BMC Biotechnol. 2 (2002).
  13. Casbon, A.-J., Reynaud, D., et al. Invasive breast cancer reprograms early myeloid differentiation in the bone marrow to generate immunosuppressive neutrophils. Proc Natl Acad Sci USA. 112 (6), E566–575 (2015).
  14. Donovan, J., & Brown, P. Parenteral injections. Curr Protoc Immunol. Chapter 1 (2006).
  15. Halpern, J., Lynch, C. C., et al. The application of a murine bone bioreactor as a model of tumor: bone interaction. Clin Exp Metastas. 23 (7-8), 345–356 (2006).
  16. Sasmono, R. T., Oceandy, D., et al. A macrophage colony-stimulating factor receptor-green fluorescent protein transgene is expressed throughout the mononuclear phagocyte system of the mouse. Blood. 101 (3), 1155–1163 (2003).
  17. Ewald, A. J., Werb, Z., & Egeblad, M. Dynamic, long-term in vivo imaging of tumor-stroma interactions in mouse models of breast cancer using spinning-disk confocal microscopy. Cold Spring Harb Protoc. 2011 (2) (2011).
  18. Bonnans, C., Lohela, M., & Werb, Z. Real-time imaging of myeloid cells dynamics in ApcMin/+ intestinal tumors by spinning disk confocal microscopy. J Vis Exp. (92) (2014).
  19. Nakasone, E. S., Askautrud, H. A., et al. Imaging tumor-stroma interactions during chemotherapy reveals contributions of the microenvironment to resistance. Cancer cell. 21 (4) (2012).
  20. Cheng, N., & Lambert, D. L. Mammary transplantation of stromal cells and carcinoma cells in C57BL/6J mice. J Vis Exp. (54) (2011).
  21. Al-Mehdi, A. B., Tozawa, K., Fisher, A. B., Shientag, L., Lee, A., & Muschel, R. J. Intravascular origin of metastasis from the proliferation of endothelium-attached tumor cells: a new model for metastasis. Nat Med. 6 (1), 100–102 (2000).
  22. Wong, C. W., Song, C., et al. Intravascular location of breast cancer cells after spontaneous metastasis to the lung. Am J Pathol. 161 (3), 749–753 (2002).
  23. Liang, C.-C., Park, A. Y., & Guan, J.-L. In vitro scratch assay: a convenient and inexpensive method for analysis of cell migration in vitro. Nat protoc. 2 (2), 329–333 (2007).
  24. Nelson, K., Bobba, C., Ghadiali, S., Hayes, D. J., Black, S. M., & Whitson, B. A. Animal models of ex vivo lung perfusion as a platform for transplantation research. World J Exp Med. 4 (2) (2014).
  25. Magness, S. T., Bataller, R., Yang, L., & Brenner, D. A. A dual reporter gene transgenic mouse demonstrates heterogeneity in hepatic fibrogenic cell populations. Hepatology. 40 (5), 1151–1159 (2004).
  26. Motoike, T., Loughna, S., et al. Universal GFP reporter for the study of vascular development. Genesis. 28 (2) (2000).
  27. Srivastava, M. K., Andersson, A., et al. Myeloid suppressor cells and immune modulation in lung cancer. Immunotherapy. 4 (3) (2012).
  28. Craig, A., Mai, J., Cai, S., & Jeyaseelan, S. Neutrophil recruitment to the lungs during bacterial pneumonia. Infect Immun. 77 (2), 568–575 (2009).
  29. Kreisel, D., Nava, R. G., et al. In vivo two-photon imaging reveals monocyte-dependent neutrophil extravasation during pulmonary inflammation. Proc Natl Acad Sci USA. 107 (42), 18073–18078 (2010).
check_url/53741?article_type=t

Play Video

Cite This Article
van den Bijgaart, R. J., Kong, N., Maynard, C., Plaks, V. Ex vivo Live Imaging of Lung Metastasis and Their Microenvironment. J. Vis. Exp. (108), e53741, doi:10.3791/53741 (2016).

View Video