Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Standardizing immunophenotyping for the Human Immunology Project

A Corrigendum to this article was published on 18 May 2012

Key Points

  • Standardized immunophenotyping assays are a requisite for accomplishing the proposed Human Immunology Project, which involves the comprehensive elucidation of the metrics of healthy versus diseased or perturbed human immune systems.

  • The variables inherent in flow cytometry immunophenotyping are largely known, and include reagent choice, sample handling, instrument setup and data analysis; strategies to mitigate each of these variables are available.

  • Several groups, including the Human Immunophenotyping Consortium, are standardizing reagent panels for flow cytometry.

  • Together with the adoption of such standard panels, an infrastructure for aggregating and mining results will be needed.

  • Availability of such panels and the data-mining infrastructure should result in more rapid biomarker discovery for immunologically relevant diseases.

Abstract

The heterogeneity in the healthy human immune system, and the immunological changes that portend various diseases, have been only partially described. Their comprehensive elucidation has been termed the 'Human Immunology Project'. The accurate measurement of variations in the human immune system requires precise and standardized assays to distinguish true biological changes from technical artefacts. Thus, to be successful, the Human Immunology Project will require standardized assays for immunophenotyping humans in health and disease. A major tool in this effort is flow cytometry, which remains highly variable with regard to sample handling, reagents, instrument setup and data analysis. In this Review, we outline the current state of standardization of flow cytometry assays and summarize the steps that are required to enable the Human Immunology Project.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: A typical flow cytometry experiment.
Figure 2: Identification of immune cell subsets by eight-colour antibody staining.
Figure 3: The importance of antibody choice.

Similar content being viewed by others

References

  1. Perez, O. D. et al. Multiparameter analysis of intracellular phosphoepitopes in immunophenotyped cell populations by flow cytometry. Curr. Protoc. Cytom. 32, 6.20.1–6.20.22 (2005).

    Google Scholar 

  2. Maecker, H. T. in Flow Cytometry Protocols (eds Hawley, T. S. & Hawley, R. G.) 95–107 (Humana Press, Totowa, New Jersey, 2004).

    Book  Google Scholar 

  3. Parish, C. R., Glidden, M. H., Quah, B. J. & Warren, H. S. Use of the intracellular fluorescent dye CFSE to monitor lymphocyte migration and proliferation. Curr. Protoc. Immunol. 84, 4.9.1–4.9.13 (2009).

  4. Betts, M. R. & Koup, R. A. Detection of T-cell degranulation: CD107a and b. Methods Cell Biol. 75, 497–512 (2004).

    Article  CAS  Google Scholar 

  5. Chattopadhyay, P. K., Yu, J. & Roederer, M. A live-cell assay to detect antigen-specific CD4+ T cells with diverse cytokine profiles. Nature Med. 11, 1113–1117 (2005).

    Article  CAS  Google Scholar 

  6. Frentsch, M. et al. Direct access to CD4+ T cells specific for defined antigens according to CD154 expression. Nature Med. 11, 1118–1124 (2005).

    Article  CAS  Google Scholar 

  7. Altman, J. D. et al. Phenotypic analysis of antigen-specific T lymphocytes. Science 274, 94–96 (1996).

    Article  CAS  Google Scholar 

  8. Prussin, C. & Metcalfe, D. D. Detection of intracytoplasmic cytokine using flow cytometry and directly conjugated anti-cytokine antibodies. J. Immunol. Methods 188, 117–128 (1995).

    Article  CAS  Google Scholar 

  9. Picker, L. J. et al. Direct demonstration of cytokine synthesis heterogeneity among human memory/effector T cells by flow cytometry. Blood 86, 1408–1419 (1995).

    CAS  PubMed  Google Scholar 

  10. Migueles, S. A. et al. HIV-specific CD8+ T cell proliferation is coupled to perforin expression and is maintained in nonprogressors. Nature Immunol. 3, 1061–1068 (2002).

    Article  CAS  Google Scholar 

  11. Betts, M. R. et al. HIV nonprogressors preferentially maintain highly functional HIV-specific CD8+ T-cells. Blood 107, 4781–4789 (2006).

    Article  CAS  Google Scholar 

  12. Fahey, J. L. et al. Immune pathogenesis of AIDS and related syndromes. Ann. Inst. Pasteur Immunol. 138, 245–252 (1987).

    Article  CAS  Google Scholar 

  13. Lifson, J. D., Finch, S. L., Sasaki, D. T. & Engleman, E. G. Variables affecting T-lymphocyte subsets in a volunteer blood donor population. Clin. Immunol. Immunopathol. 36, 151–160 (1985).

    Article  CAS  Google Scholar 

  14. Kestens, L. et al. Expression of activation antigens, HLA-DR and CD38, on CD8 lymphocytes during HIV-1 infection. AIDS 6, 793–797 (1992).

    Article  CAS  Google Scholar 

  15. Duque, R. E. Flow cytometric analysis of lymphomas and acute leukemias. Ann. NY Acad. Sci. 677, 309–325 (1993).

    Article  CAS  Google Scholar 

  16. Posnett, D. N., Edinger, J. W., Manavalan, J. S., Irwin, C. & Marodon, G. Differentiation of human CD8 T cells: implications for vitro persistence of CD8+ CD28 cytotoxic effector clones. Int. Immunol. 11, 229–241 (1999).

    Article  CAS  Google Scholar 

  17. Khan, N. et al. Cytomegalovirus seropositivity drives the CD8 T cell repertoire toward greater clonality in healthy elderly individuals. J. Immunol. 169, 1984–1992 (2002).

    Article  CAS  Google Scholar 

  18. Maecker, H. T. et al. A model for harmonizing flow cytometry in clinical trials. Nature Immunol. 11, 975–978 (2010). This perspective details the ways in which flow cytometry phenotyping assays can vary in the clinical trial setting, and advocates ways to standardize them.

    Article  CAS  Google Scholar 

  19. Suni, M. A. et al. Performance of plate-based cytokine flow cytometry with automated data analysis. BMC Immunol. 4, 9 (2003).

    Article  Google Scholar 

  20. Maecker, H. T. et al. Standardization of cytokine flow cytometry assays. BMC Immunol. 6, 13 (2005). Using multi-site data on shared samples, this paper shows the benefits of centralized and/or automated gating and preconfigured lyophilized reagents.

    Article  Google Scholar 

  21. Dunne, J. F. & Maecker, H. T. Automation of cytokine flow cytometry assays. JALA 9, 5–9 (2004).

    CAS  Google Scholar 

  22. Gratama, J. W. et al. Analysis of variation in results of flow cytometric lymphocyte immunophenotyping in a multicenter study. Cytometry 30, 166–177 (1997).

    Article  CAS  Google Scholar 

  23. Beisvag, V. et al. Contributions of the EMERALD project to assessing and improving microarray data quality. Biotechniques 50, 27–31 (2011).

    Article  CAS  Google Scholar 

  24. Devonshire, A. S., Elaswarapu, R. & Foy, C. A. Evaluation of external RNA controls for the standardisation of gene expression biomarker measurements. BMC Genomics 11, 662 (2010).

    Article  CAS  Google Scholar 

  25. Calza, S. & Pawitan, Y. Normalization of gene-expression microarray data. Methods Mol. Biol. 673, 37–52 (2010).

    Article  CAS  Google Scholar 

  26. Rogers, S. & Cambrosio, A. Making a new technology work: the standardization and regulation of microarrays. Yale J. Biol. Med. 80, 165–178 (2007).

    CAS  PubMed  Google Scholar 

  27. Ball, C. A. et al. Standards for microarray data. Science 298, 539 (2002).

    Article  CAS  Google Scholar 

  28. Patterson, T. A. et al. Performance comparison of one-color and two-color platforms within the MicroArray Quality Control (MAQC) project. Nature Biotech. 24, 1140–1150 (2006).

    Article  CAS  Google Scholar 

  29. Shi, L. et al. The MicroArray Quality Control (MAQC) project shows inter- and intraplatform reproducibility of gene expression measurements. Nature Biotech. 24, 1151–1161 (2006). A seminal paper demonstrating that the major commercial microarray platforms can generate similar and reproducible data.

    Article  CAS  Google Scholar 

  30. Davis, M. M. A prescription for human immunology. Immunity 29, 835–838 (2008). In this article, Mark Davis lays the foundation for why we need a 'Human Immunology Project'.

    Article  CAS  Google Scholar 

  31. Sallusto, F., Lenig, D., Forster, R., Lipp, M. & Lanzavecchia, A. Two subsets of memory T lymphocytes with distinct homing potentials and effector functions. Nature 401, 708–712 (1999).

    Article  CAS  Google Scholar 

  32. Tussey, L., Speller, S., Gallimore, A. & Vessey, R. Functionally distinct CD8+ memory T cell subsets in persistent EBV infection are differentiated by migratory receptor expression. Eur. J. Immunol. 30, 1823–1829 (2000).

    Article  CAS  Google Scholar 

  33. Appay, V. et al. Memory CD8+ T cells vary in differentiation phenotype in different persistent virus infections. Nature Med. 8, 379–385 (2002).

    Article  CAS  Google Scholar 

  34. Lin, S. J., Chao, H. C., Yan, D. C. & Huang, Y. J. Expression of adhesion molecules on T lymphocytes in young children and infants — a comparative study using whole blood lysis or density gradient separation. Clin. Lab. Haematol. 24, 353–359 (2002).

    Article  Google Scholar 

  35. Weinberg, A. et al. Optimization and limitations of use of cryopreserved peripheral blood mononuclear cells for functional and phenotypic T-cell characterization. Clin. Vaccine Immunol. 16, 1176–1186 (2009). One of the best articles on the cryopreservation of PBMCs, its optimization and the caveats for use with T cell assays.

    Article  CAS  Google Scholar 

  36. Fritsch, R. D. et al. Stepwise differentiation of CD4 memory T cells defined by expression of CCR7 and CD27. J. Immunol. 175, 6489–6497 (2005).

    Article  CAS  Google Scholar 

  37. Pascual, V. et al. Analysis of somatic mutation in five B cell subsets of human tonsil. J. Exp. Med. 180, 329–339 (1994).

    Article  CAS  Google Scholar 

  38. Bohnhorst, J. O., Bjorgan, M. B., Thoen, J. E., Natvig, J. B. & Thompson, K. M. Bm1–Bm5 classification of peripheral blood B cells reveals circulating germinal center founder cells in healthy individuals and disturbance in the B cell subpopulations in patients with primary Sjogren's syndrome. J. Immunol. 167, 3610–3618 (2001).

    Article  CAS  Google Scholar 

  39. Agematsu, K. et al. B cell subpopulations separated by CD27 and crucial collaboration of CD27+ B cells and helper T cells in immunoglobulin production. Eur. J. Immunol. 27, 2073–2079 (1997).

    Article  CAS  Google Scholar 

  40. Cuss, A. K. et al. Expansion of functionally immature transitional B cells is associated with human-immunodeficient states characterized by impaired humoral immunity. J. Immunol. 176, 1506–1516 (2006).

    Article  CAS  Google Scholar 

  41. Rawstron, A. C. Immunophenotyping of plasma cells. Curr. Protoc. Cytom. 36, 6.23.1–6.23.14 (2006).

  42. Avery, D. T. et al. Increased expression of CD27 on activated human memory B cells correlates with their commitment to the plasma cell lineage. J. Immunol. 174, 4034–4042 (2005).

    Article  CAS  Google Scholar 

  43. Hamerman, J. A., Ogasawara, K. & Lanier, L. L. NK cells in innate immunity. Curr. Opin. Immunol. 17, 29–35 (2005).

    Article  CAS  Google Scholar 

  44. Beziat, V. et al. CD56brightCD16+ NK cells: a functional intermediate stage of NK cell differentiation. J. Immunol. 186, 6753–6761 (2011).

    Article  CAS  Google Scholar 

  45. Willmann, K. & Dunne, J. F. A flow cytometric immune function assay for human peripheral blood dendritic cells. J. Leukoc. Biol. 67, 536–544 (2000).

    Article  CAS  Google Scholar 

  46. Ju, X., Clark, G. & Hart, D. N. Review of human DC subtypes. Methods Mol. Biol. 595, 3–20 (2010).

    Article  Google Scholar 

  47. Dzionek, A. et al. BDCA-2, BDCA-3, and BDCA-4: three markers for distinct subsets of dendritic cells in human peripheral blood. J. Immunol. 165, 6037–6046 (2000).

    Article  CAS  Google Scholar 

  48. Ziegler-Heitbrock, H. W. et al. Small (CD14+/CD16+) monocytes and regular monocytes in human blood. Pathobiology 59, 127–130 (1991).

    Article  CAS  Google Scholar 

  49. Allers, K. et al. Migration patterns of nonspecifically activated versus nonactivated nonhuman primate T lymphocytes: preferential homing of activated autologous CD8+ T cells in the rectal mucosa. J. Immunother. 31, 334–344 (2008).

    Article  Google Scholar 

  50. Wrammert, J. et al. Rapid cloning of high-affinity human monoclonal antibodies against influenza virus. Nature 453, 667–671 (2008).

    Article  CAS  Google Scholar 

  51. Maecker, H. T., Frey, T., Nomura, L. E. & Trotter, J. Selecting fluorochrome conjugates for maximum sensitivity. Cytometry A 62, 169–173 (2004).

    Article  Google Scholar 

  52. Nomura, L., Maino, V. C. & Maecker, H. T. Standardization and optimization of multiparameter intracellular cytokine staining. Cytometry A 73, 984–991 (2008).

    Article  Google Scholar 

  53. Belouski, S. S. et al. Utility of lyophilized PMA and ionomycin to stimulate lymphocytes in whole blood for immunological assays. Cytometry B Clin. Cytom. 78, 59–64 (2009).

    Google Scholar 

  54. Inokuma, M. et al. Functional T cell responses to tumor antigens in breast cancer patients have a distinct phenotype and cytokine signature. J. Immunol. 179, 2627–2633 (2007).

    Article  CAS  Google Scholar 

  55. Maecker, H. T. & Trotter, J. Flow cytometry controls, instrument setup, and the determination of positivity. Cytometry A 69, 1037–1042 (2006).

    Article  Google Scholar 

  56. Jaimes, M. C. et al. Quality assurance of intracellular cytokine staining assays: Analysis of multiple rounds of proficiency testing. J. Immunol. Methods 363, 143–157 (2010).

    Article  Google Scholar 

  57. Aghaeepour, N. et al. Early immunologic correlates of HIV protection can be identified from computational analysis of complex multivariate T-cell flow cytometry assays. Bioinformatics (in the press).

  58. Hultin, L. E. et al. Comparison of interlaboratory variation in absolute T-cell counts by single-platform and optimized dual-platform methods. Cytometry B Clin. Cytom. 78, 194–200 (2010).

    PubMed  PubMed Central  Google Scholar 

  59. Bandura, D. R. et al. Mass cytometry: technique for real time single cell multitarget immunoassay based on inductively coupled plasma time-of-flight mass spectrometry. Anal. Chem. 81, 6813–6822 (2009).

    Article  CAS  Google Scholar 

  60. Ornatsky, O. et al. Highly multiparametric analysis by mass cytometry. J. Immunol. Methods 361, 1–20 (2010).

    Article  CAS  Google Scholar 

  61. Bendall, S. C. et al. Single-cell mass cytometry of differential immune and drug responses across a human hematopoietic continuum. Science 332, 687–696 (2011). An impressive demonstration of the power of mass cytometry for highly multidimensional analyses of haematopoietic cells.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The authors thank all the contributors to the HIPC/FITMaN immunophenotyping panel, in particular S. Heck, F. Nestle, A. Biancotto, S. Gupta, M. Malipatlolla, L. Devine, R. Montgomery and D. Hafler. The mention of any commercial products in this manuscript does not imply endorsement by the US Government or the US National Institutes of Health.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Holden T. Maecker.

Ethics declarations

Competing interests

Holden T. Maecker is a stockholder of BD Biosciences, which is a manufacturer of flow cytometers and reagents.

Related links

Related links

FURTHER INFORMATION

Holden T. Maecker's homepage

Clinical Trials in Organ Transplantation

FlowCAP

Spring 2011 FOCIS newsletter

Glossary

Immune phenotypes

Measurable aspects of the immune system, such as the proportions of various cell subsets or measures of cellular immune function.

Gates

Sequential filters that are applied to a set of flow cytometry data to focus the analysis on particular cell subsets of interest.

Human Immunology Project

The comprehensive mapping of immune phenotypes in healthy and diseased human populations.

Density gradient separation

The isolation of mononuclear cells from blood or other sources by centrifugation over a density gradient, which usually consists of a carbohydrate polymer solution (Ficoll).

Cryopreservation

The processing and storage of cells at sub-zero temperatures under conditions that preserve their viability for later assays.

Lineage cocktail

A mixture of antibodies specific for various lineage-specific markers, such as CD3 (for T cells), CD14 (for monocytes) and CD19 and CD20 (for B cells).

Antibody capture beads

Microparticles conjugated with immunoglobulin-specific antibodies. These beads can be stained with fluorescent antibodies to create single-colour controls for flow cytometry instrument setup and compensation.

Phosphoepitope flow cytometry

(Also known as 'phospho-flow'). A technique that uses antibodies specific for phosphorylated versions of proteins to analyse cell signalling by flow cytometry.

Mass cytometry

Flow cytometry using antibodies tagged with heavy metal ions, which are detected by mass spectrometry (as opposed to classical flow cytometry, which uses antibodies tagged with fluorophores and optical detection).

Optical spillover

The presence of signals from fluorescent antibody staining in multiple detectors of a cytometer, resulting in a loss of resolution sensitivity.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Maecker, H., McCoy, J. & Nussenblatt, R. Standardizing immunophenotyping for the Human Immunology Project. Nat Rev Immunol 12, 191–200 (2012). https://doi.org/10.1038/nri3158

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nri3158

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing