JoVE Logo

Sign In

A subscription to JoVE is required to view this content. Sign in or start your free trial.

In This Article

  • Summary
  • Abstract
  • Introduction
  • Protocol
  • Results
  • Discussion
  • Disclosures
  • Acknowledgements
  • Materials
  • References
  • Reprints and Permissions

Summary

We describe a method for using multiparameter flow cytometry to detect mitochondrial reactive oxygen species (ROS) in murine healthy hematopoietic stem and progenitor cells (HSPCs) and leukemia cells from a mouse model of acute myeloid leukemia (AML) driven by MLL-AF9.

Abstract

We present a flow cytometric approach for analyzing mitochondrial ROS in various live bone marrow (BM)-derived stem and progenitor cell populations from healthy mice as well as mice with AML driven by MLL-AF9. Specifically, we describe a two-step cell staining process, whereby healthy or leukemia BM cells are first stained with a fluorogenic dye that detects mitochondrial superoxides, followed by staining with fluorochrome-linked monoclonal antibodies that are used to distinguish various healthy and malignant hematopoietic progenitor populations. We also provide a strategy for acquiring and analyzing the samples by flow cytometry. The entire protocol can be carried out in a timeframe as short as 3-4 h. We also highlight the key variables to consider as well as the advantages and limitations of monitoring ROS production in the mitochondrial compartment of live hematopoietic and leukemia stem and progenitor subpopulations using fluorogenic dyes by flow cytometry. Furthermore, we present data that mitochondrial ROS abundance varies among distinct healthy HSPC sub-populations and leukemia progenitors and discuss the possible applications of this technique in hematologic research.

Introduction

Reactive Oxygen Species (ROS) are highly reactive molecules derived from molecular oxygen. The most well-defined cellular location of ROS production is the mitochondria, where electrons that pass through the electron transport chain (ETC) during oxidative phosphorylation (OXPHOS) are absorbed by molecular oxygen leading to the formation of a specific type of ROS called superoxides1. Through the actions of a series of enzymes, called superoxide dismutases or SODs, superoxides are converted into hydrogen peroxides, which are subsequently neutralized into water by enzymes such as catalase or glutathione peroxidases (GPX). Perturbations in ROS-regulatory mechanisms can lead to the excess production of ROS, often referred to as oxidative stress, which have harmful and potentially lethal cellular consequences such as macromolecule damage (i.e., DNA, protein, lipids). Moreover, oxidative stress is related to several pathologies, such as diabetes, inflammatory diseases, aging and tumors2,3,4. To maintain redox homeostasis and prevent oxidative stress, cells possess a variety of ROS-regulating mechanisms5.

Physiological levels of certain ROS are necessary for proper embryonic and adult hematopoiesis6. However, excess ROS is associated with DNA damage, cellular differentiation and exhaustion of the hematopoietic stem and progenitor pool. There is also evidence that alterations in redox biology may differ between leukemia and healthy cells. For example, ROS levels tend to be higher in acute myeloid leukemia (AML) cells relative to their healthy counterparts and other studies have suggested that leukemia stem cells maintain a low steady-state level of ROS for survival7,8. Importantly, strategies for therapeutically capitalizing on these redox differences have shown promise in several human cancer settings9,10. Therefore, assays that allow for the assessment of ROS levels in mouse models may improve our understanding of how these species contribute to cellular physiology and disease pathogenesis as well as potentially provide a platform for assessing the effectiveness of novel redox-targeting anti-cancer therapies.

Protocol

All of the animal procedures described in this protocol have been approved by the Institutional Animal Care and Use committee (IACUC) at Fox Chase Cancer Center.

NOTE: The protocol workflow is divided into 4 parts as presented in Figure 1 and the required reagents are listed in the Table of Materials.

1. Bone Marrow (BM) Isolation

NOTE: MLL-AF9 leukemia mice were generated as described previously11.

  1. Recover mono-nuclear bone marrow cells, as described previously12,13,14,15, from wild type C57.Bl6 mice (which express the CD45.2 congenic marker) as well as from C57.Bl6-SJL mice ( which express the CD45.1 congenic marker) that have been transplanted with MLL-AF9-expressing leukemia cells (CD45.2+).
    NOTE: BM can be recovered from mice either by crushing12,13 or by flushing bones14,15. For the experiments presented here, BM was recovered from both healthy and leukemia mice via flushing.

2. Mitochondrial ROS Fluorogenic Dye Staining

  1. Once mono-nuclear bone marrow cells have been recovered from healthy and/or leukemia mice, stain an aliquot of cells with Trypan Blue and count using a hemocytometer to determine the starting number of total BM cells.
  2. Centrifuge the cells at 300 x g for 5 min. Aspirate the supernatant and resuspend the pellet in F-PBS (PBS supplemented with 2% fetal bovine serum and a 1% Penicillin/Streptomycin cocktail) to a concentration of 2 x 106 cells/mL.
  3. Aliquot 200 µL of cell suspension per tube into 9 single-color control tubes labeled as follows:
    No stain, B220-Cy5-PE, cKit-Cy7-APC, Sca1-PacBlue, CD150-APC (for healthy HSPCs only), CD45.2-APC (for leukemia cells only), CD34-FITC, Mitochondrial ROS dye and Live/dead cell stain.
    NOTE: (Optional) A positive control for the induction of mitochondrial ROS can be prepared by treating 2 x 105 cells in 200 µL with 20 µM of Menadione Sodium Bisulfite (MSB) for 1 h at 37 ˚C in a 5% CO2 incubator. A second control to reverse the MSB-mediated induction of mitochondrial ROS can be prepared by treating 2 x 105 cells in 200 µL with 20 µM of MSB plus 100 µM N-acetyl-L-cysteine (NAC) for 1 h at 37 ˚C in a 5% CO2 incubator.
  4. Aliquot the remaining cells in a tube (experimental tube) and centrifuge at 300 x g for 5 min.
  5. Resuspend the cells in F-PBS with a live/dead cell stain according to the manufacturer’s instructions. Incubate on ice for 30 min. Be sure to add live/dead stain to the single-color control tube.
  6. Add 1.0 mL of room temperature (RT) F-PBS to both single-color and experimental tubes stained with the live/dead dye. Centrifuge 5 min at 300 x g at RT.
  7. Resuspend 50 µg of the mitochondrial ROS dye in 13 µL of dimethyl sulfoxide (DMSO) to obtain a 5 mM stock solution.
  8. Dilute mitochondrial ROS dye to a final concentration of 5 µM in RT F-PBS with or without Verapamil (50 µM).
  9. Aspirate off the wash of the live/dead cell stain. Add 200 µL of mitochondrial ROS dye stain containing Verapamil to each experimental tube as well as the mitochondrial ROS dye single-color control tube.
  10. Vortex to mix and incubate for 10 min at 37 ˚C in the dark.
  11. Add 1.0 mL of RT F-PBS to the mitochondrial ROS-stained single-color control and experimental tubes. Centrifuge 5 min at 300 x g at RT.
  12. Aspirate off the supernatant and wash the cells with an additional 1.0 mL of RT F-PBS. Centrifuge 5 min at 300 x g at RT.

3. Lineage Antibody Staining

  1. Prepare the antibody cocktails listed in Table 1.
    NOTE: These antibodies cocktails have been optimized previously14,15,16.
  2. Aspirate the supernatant from the final mitochondrial ROS dye wash of the experimental tubes containing healthy BM and add 200 µL of antibody cocktail #1 to each tube. Vortex to mix. Also prepare the single-color control tubes. Incubate for 60 min on ice in the dark.
  3. Aspirate the supernatant from the final mitochondrial ROS dye wash of the experimental tubes containing leukemia BM and add 200 µL of the antibody cocktail #2 to each tube. Vortex to mix. Incubate for 60 min on ice in the dark.
  4. Wash with 1.0 mL of cold F-PBS and centrifuge 5 min at 300 x g at RT.
  5. Resuspend cells in 500 µL of cold F-PBS and filter the cells in a flow cytometer tube using a 40 µm filter to exclude aggregates.

4. Flow Cytometry Acquisition and Analysis

NOTE: Several hematopoietic stem and progenitor subsets are rare, such as long-term hematopoietic stem cells. Thus, ideally 3-5 million events should be collected for each experimental tube during flow cytometry acquisition for sufficient analysis of mitochondrial ROS in the various HSPC subsets.

  1. Use the no-stain control tube to set the forward (FSC-A) and side (SSC-A) scatter plots based on the size and complexity of the cell population analyzed.
  2. Use the no-stain and single-color control tubes to compensate the flow cytometer.
  3. Gate out extraneous debris from the forward and side scatter plot (Figure 2A,B, first panel from the left).
  4. Gate out doublets using a double discriminator such as the forward discriminator (Figure 2A,B, second panel from the left).
  5. Follow the gating strategy proposed in Figure 2A,B to select live cells, lineage low cells and the various HSPC and leukemia subsets.
  6. For each population of interest, analyze the median fluorescence intensity (MFI) of the TRPE channel (x-axis) in a histogram plot to evaluate differences in the mitochondrial ROS signal (Figure 3A-C, left panels). Levels of mitochondrial ROS can be evaluated at the single-cell level by comparing mitochondrial ROS staining versus specific lineage markers in a scatter plot.

Results

Presented is a method for analyzing ROS in the mitochondria of multiple healthy and MLL-AF9-expressing leukemia progenitor populations. Figure 1 displays a schematic view of the protocol workflow, which consists of 4 major steps: 1) BM isolation from mice; 2) Staining BM cells with a fluorogenic dye that recognizes mitochondrial ROS, particularly superoxides; 3) Surface marker antibody staining to delineate various healthy and leukemia hematopoietic populations; and 4) Flow cytometry acquisi...

Discussion

Fluorogenic dyes that have been developed for the detection of ROS are frequently evaluated in fixed cells by microscopy or in live cells by flow cytometry22. Flow cytometric evaluation of mitochondrial ROS in BM cells using mitochondrial ROS fluorogenic dyes has two major advantages: 1) It is a fast and simple technique that is suitable for live cell analysis and 2) it allows for distinguishing and analyzing rare populations at the single-cell level in the BM using surface marker staining. The st...

Disclosures

The authors have nothing to disclose.

Acknowledgements

This work was supported by The Fox Chase Cancer Center Board of Directors (DDM), the American Society of Hematology Scholar Award (SMS), American Cancer Society RSG (SMS) and the Department of Defense (Award#: W81XWH-18-1-0472).

Materials

NameCompanyCatalog NumberComments
Heat inactivated FBSVWR Seradigm LIFE SCIENCE97068-085Media
Penicillin StreptomycinCorning30-002-CIMedia
PBSFisher ScientificBP399-20Buffer
15 mL conical tubeBD falcon352096Tissue Culture Supplies
50 mL conical tubeBD falcon352098Tissue Culture Supplies
40 μm cell strainersFisher Scientific22-363-547Tissue Culture Supplies
RBC Lysis BufferFisher Scientific50-112-9751Tissue Culture Supplies
Menadione sodium BisulfiteSigma aldrichM5750Pro-oxidant
NACSigma aldrichA7250Anti-oxidant
CD3 PE-Cy5 clone 145-2c11Biolegend100310Antibody
CD4 PE-Cy5 clone RM4-5eBioscience15-0041-81Antibody
CD8 PE-Cy5 clone 53-6.7eBioscience15-0081-81Antibody
CD19 PE-Cy5 clone 6D5Biolegend115510Antibody
B220 PE-Cy5 clone RA3-6B2Biolegend103210Antibody
Gr1 PE-Cy5 clone RB6-8C5Biolegend108410Antibody
Ter119 PE-Cy5 clone Ter-119Biolegend116210Antibody
CD48 PE-Cy5 clone HM48-1Biolegend103420Antibody
CD117  APC-Cy7 clone 2B8Biolegend105825Antibody
Sca1 peacific Blue clone D7Biolegend108120Antibody
CD150 APC clone TC15-12F12.2Biolegend115909Antibody
CD34 FITC clone RAM34BD Bioscience553733Antibody
CD45.2 APC clone 104Biolegend1098313Antibody
MitoSOX RedThermoFisher ScientificM36008Dye
Mitotracker GreenThermoFisher ScientificM7514Dye
Live/dead Yellow DyeThermoFisher ScientificL34967Dye

References

  1. Dröse, S., Brandt, U. Molecular mechanisms of superoxide production by the mitochondrial respiratory chain. Advances in experimental medicine and biology. 748, 145-169 (2012).
  2. Gerber, P. A., Rutter, G. A. The Role of Oxidative Stress and Hypoxia in Pancreatic Beta-Cell Dysfunction in Diabetes Mellitus. Antioxidant & Redox Signaling. 26 (10), 501-518 (2017).
  3. Höhn, A., et al. Happily (n)ever after: Aging in the context of oxidative stress, proteostasis loss and cellular senescence. Redox Biology. 11, 482-501 (2017).
  4. Reuter, S., Gupta, S. C., Chaturvedi, M. M., Aggarwal, B. B. Oxidative stress, inflammation, and cancer: how are they linked. Free Radical Biology & Medicine. 49 (11), 1603-1616 (2010).
  5. Lee, B. W. L., Ghode, P., Ong, D. S. T. Redox regulation of cell state and fate. Redox Biology. 2213-2317 (18), 30899 (2018).
  6. Harris, J. M., et al. Glucose metabolism impacts the spatiotemporal onset and magnitude of HSC induction in vivo. Blood. 121, 2483-2493 (2013).
  7. Hole, P. S., Darley, R. L., Tonks, A. Do reactive oxygen species play a role in myeloid leukemias. Blood. 117, 5816-5826 (2011).
  8. Lagadinou, E. D., et al. BCL-2 inhibition targets oxidative phosphorylation and selectively eradicates quiescent human leukemia stem cells. Cell Stem Cell. 12 (3), 329-341 (2013).
  9. Di Marcantonio, D., et al. Protein Kinase C Epsilon Is a Key Regulator of Mitochondrial Redox Homeostasis in Acute Myeloid Leukemia. Clinical Cancer Research. 24 (3), 608-618 (2018).
  10. Glasauer, A., Chandel, N. S. Targeting antioxidants for cancer therapy. Biochemical Pharmacology. 92 (1), 90-101 (2014).
  11. Krivtsov, A. V., et al. Transformation from committed progenitor to leukaemia stem cell initiated by MLL-AF9. Nature. 442 (7104), 818-822 (2006).
  12. Frascoli, M., Proietti, M., Grassi, F. Phenotypic analysis and isolation of murine hematopoietic stem cells and lineage-committed progenitors. Journal of Visualized Experiments. (65), (2012).
  13. Lo Celso, C., Scadden, D. T. Isolation and transplantation of hematopoietic stem cells (HSCs). Journal of Visualized Experiments. (157), (2007).
  14. Kalaitzidis, D., et al. mTOR complex 1 plays critical roles in hematopoiesis and Pten-loss-evoked leukemogenesis. Cell Stem Cell. 11 (3), 429-439 (2012).
  15. Sykes, S. M., et al. AKT/FOXO signaling enforces reversible differentiation blockade in myeloid leukemias. Cell. 146 (5), 697-708 (2011).
  16. Kalaitzidis, D., Neel, B. G. Flow-cytometric phosphoprotein analysis reveals agonist and temporal differences in responses of murine hematopoietic stem/progenitor cells. PLoS One. 3 (11), 3776 (2008).
  17. Kiel, M. J., Yilmaz, O. H., Iwashita, T., Yilmaz, O. H., Terhorst, C., Morrison, S. J. SLAM family receptors distinguish hematopoietic stem and progenitor cells and reveal endothelial niches for stem cells. Cell. 121 (7), 1109-1121 (2005).
  18. Mooney, C. J., Cunningham, A., Tsapogas, P., Toellner, K. M., Brown, G. Selective expression of flt3 within the mouse hematopoietic stem cell compartment. International Journal Molecular Sciences. 18 (5), (2017).
  19. Oguro, H., Ding, L., Morrison, S. I. SLAM family markers resolve functional distinct sub-populations of hematopoietic stem cells and multipotent progenitors. Cell Stem Cell. 13 (1), 102-116 (2013).
  20. Osawa, M., Hanada, K., Hamada, H., Nakauchi, H. Long-term lymphohematopoietic reconstitution by a single CD34-low/negative hematopoietic stem cell. Science. 273 (5272), 242-245 (1996).
  21. Morita, Y., Ema, H., Nakauchi, H. Heterogeneity and hierarchy within the most primitive hematopoietic stem cell compartment. Journal of Experimental Medicine. 207 (6), 1173-1178 (2010).
  22. Mukhopadhyay, P., Rajesh, M., Haskó, G., Hawkins, B. J., Madesh, M., Pacher, P. Simultaneous detection of apoptosis and mitochondrial superoxide production in live cells by flow cytometry and confocal microscopy. Nature Protocols. 2 (9), 2295-2301 (2007).
  23. Camargo, F. D., Chambers, S. M., Drew, E., McNagny, K. M., Goodell, M. A. Hematopoietic stem cells do not engraft with absolute efficiencies. Blood. 107 (2), 501-507 (2006).
  24. Morita, Y., Ema, H., Yamazaki, S., Nakauchi, H. Non-side-population hematopoietic stem cells in mouse bone marrow. Blood. 108 (8), 2850-2856 (2006).
  25. de Almeida, M. J., Luchsinger, L. L., Corrigan, D. J., Williams, L. J., Snoeck, H. W. Dye-Independent Methods Reveal Elevated Mitochondrial Mass in Hematopoietic Stem Cells. Cell Stem Cell. 21 (6), 725-729 (2017).
  26. Bonora, M., Ito, K., Morganti, C., Pinton, P., Ito, K. Membrane-potential compensation reveals mitochondrial volume expansion during HSC commitment. Experimental Hematology. 68, 30-37 (2018).
  27. Somervaille, T. C., Cleary, M. L. Identification and characterization of leukemia stem cells in murine MLL-AF9 acute myeloid leukemia. Cancer Cell. 10 (4), 257-268 (2006).
  28. Hao, X., et al. Metabolic Imaging Reveals a Unique Preference of Symmetric Cell Division and Homing of Leukemia-Initiating Cells in an Endosteal Niche. Cell Metabolism. 29 (4), 950-965 (2019).

Reprints and Permissions

Request permission to reuse the text or figures of this JoVE article

Request Permission

Explore More Articles

Flow CytometryReactive Oxygen SpeciesROSHematopoietic Stem CellsProgenitor CellsMitochondrial ROSAcute Myeloid LeukemiaAMLGene ManipulationCell MetabolismLive Cell AnalysisBone MarrowStaining ProtocolMononuclear CellsLeukemic Cells

This article has been published

Video Coming Soon

JoVE Logo

Privacy

Terms of Use

Policies

Research

Education

ABOUT JoVE

Copyright © 2025 MyJoVE Corporation. All rights reserved