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:

Humanized mice in translational biomedical research

Key Points

  • There is a growing need for animal models to carry out in vivo studies of human biological systems without putting individuals at risk. Severely immunodeficient mice engrafted with human cells and tissues, known as 'humanized' mice, facilitate progress in studies of human haematopoiesis, immunity, gene therapy, infectious diseases, cancer and regenerative medicine.

  • Advances in the generation of humanized mice have depended on a systematic progression of genetic modifications in immunodeficient mouse hosts and on improvements in engraftment techniques.

  • Mice homozygous for the severe combined immunodeficiency (scid) gene mutation or for targeted mutations at the recombination-activating gene 1 (Rag1) or Rag2 loci, accompanied by a targeted mutation at the interleukin-2 receptor γ-chain (Il2rg) locus, support greatly increased engraftment and function of human haematopoietic stem cells (HSCs) and peripheral-blood mononuclear cells (PBMCs) compared with previous immunodeficient mouse models.

  • The development of immunodeficient mice that are humanized by engraftment of human lymphoid tissues, HSCs and PBMCs provides an opportunity to carry out translational research on human immunity and autoimmune diseases. These models are also being used to study the biology of human pathogens responsible for AIDS and several other infectious diseases.

  • Humanized mice are being increasingly used as hosts for human malignant cells in studies of carcinogenesis, tumour metastasis and cancer therapy. The phenotypic and functional characterization of human tumour stem cells is also being advanced through the study of humanized mice.

  • The potential for new advances in our understanding of human immunology and other areas of human biology that is supported by studies in humanized mice remains promising. Additional genetic and technological modifications will accelerate progress towards the development of a functional human immune system in mice.

Abstract

The culmination of decades of research on humanized mice is leading to advances in our understanding of human haematopoiesis, innate and adaptive immunity, autoimmunity, infectious diseases, cancer biology and regenerative medicine. In this Review, we discuss the development of these new generations of humanized mice, how they will facilitate translational research in several biomedical disciplines and approaches to overcome the remaining limitations of these models.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: A schematic diagram of the development of various immunodeficient mouse models that are used as hosts for human cells and tissues.
Figure 2: The use of severe combined immunodeficient mice in regenerative medicine.

Similar content being viewed by others

References

  1. Bosma, G. C., Custer, R. P. & Bosma, M. J. A severe combined immunodeficiency mutation in the mouse. Nature 301, 527–530 (1983).

    Article  CAS  PubMed  Google Scholar 

  2. Mosier, D. E., Gulizia, R. J., Baird, S. M. & Wilson, D. B. Transfer of a functional human immune system to mice with severe combined immunodeficiency. Nature 335, 256–259 (1988). The original description of the use of CB17- scid mice as hosts for human PBMCs, known as the Hu-PBL-SCID model.

    Article  CAS  PubMed  Google Scholar 

  3. McCune, J. M. et al. The SCID-hu mouse: murine model for the analysis of human hematolymphoid differentiation and function. Science 241, 1632–1639 (1988). The original description of the use of CB17- scid mice as hosts for human haematopoietic fetal liver and thymus, resulting in the development of the SCID-hu model. This model is still used, in particular for testing HIV therapeutics.

    Article  CAS  PubMed  Google Scholar 

  4. Lapidot, T. et al. Cytokine stimulation of multi-lineage hematopoiesis from immature human cells engrafted in SCID mice. Science 255, 1137–1141 (1992). The original description of the use of CB17- scid mice as hosts for human HSCs and the development of the Hu-SRC-SCID model.

    Article  CAS  PubMed  Google Scholar 

  5. Greiner, D. L., Hesselton, R. A. & Shultz, L. D. SCID mouse models of human stem cell engraftment. Stem Cells 16, 166–177 (1998).

    Article  CAS  PubMed  Google Scholar 

  6. Fulop, G. M. & Phillips, R. A. The scid mutation in mice causes a general defect in DNA repair. Nature 347, 479–482 (1990).

    Article  CAS  PubMed  Google Scholar 

  7. Mombaerts, P. et al. RAG-1-deficient mice have no mature B and T lymphocytes. Cell 68, 869–877 (1992).

    Article  CAS  PubMed  Google Scholar 

  8. Shinkai, Y. et al. RAG-2-deficient mice lack mature lymphocytes owing to inability to initiate V(D)J rearrangement. Cell 68, 855–867 (1992).

    Article  CAS  PubMed  Google Scholar 

  9. Shultz, L. D. et al. Multiple defects in innate and adaptive immunologic function in NOD/LtSz-scid mice. J. Immunol. 154, 180–191 (1995).

    CAS  PubMed  Google Scholar 

  10. Hesselton, R. M. et al. High levels of human peripheral blood mononuclear cell engraftment and enhanced susceptibility to HIV-1 infection in NOD/LtSz-scid/scid mice. J. Infect. Dis. 172, 974–982 (1995).

    Article  CAS  PubMed  Google Scholar 

  11. Christianson, S. W. et al. Role of natural killer cells on engraftment of human lymphoid cells and on metastasis of human T-lymphoblastoid leukemia cells in C57BL/6J-scid mice and in C57BL/6J-scid bg mice. Cell. Immunol. 171, 186–199 (1996).

    CAS  PubMed  Google Scholar 

  12. Lowry, P. A. et al. Improved engraftment of human cord blood stem cells in NOD/LtSz-scid/scid mice after irradiation or multiple-day injections into unirradiated recipients. Biol. Blood Marrow Transplant. 2, 15–23 (1996).

    CAS  PubMed  Google Scholar 

  13. Pflumio, F. et al. Phenotype and function of human hematopoietic cells engrafting immune-deficient CB17-severe combined immunodeficiency mice and nonobese diabetic-severe combined immunodeficiency mice after transplantation of human cord blood mononuclear cells. Blood 88, 3731–3740 (1996).

    CAS  PubMed  Google Scholar 

  14. Ito, M. et al. NOD/SCID/γcnull mouse: an excellent recipient mouse model for engraftment of human cells. Blood 100, 3175–3182 (2002). This paper describes the development of a human immune system in NOD/Shi- scid Il2rg−/− mice engrafted as adults with human CD34+ cord-blood HSCs.

    Article  CAS  PubMed  Google Scholar 

  15. Traggiai, E. et al. Development of a human adaptive immune system in cord blood cell-transplanted mice. Science 304, 104–107 (2004). This paper shows the development of a complete human immune system in BALB/c- Rag2−/−Il2rg−/− mice engrafted intrahepatically as newborns with human cord-blood CD34+ HSCs.

    Article  CAS  PubMed  Google Scholar 

  16. Shultz, L. et al. Human lymphoid and myeloid cell development in NOD/LtSz-scid IL2rgnull mice engrafted with mobilized human hematopoietic stem cells. J. Immunol. 174, 6477–6489 (2005). One of the first studies to show the development of a complete human immune system in NOD- scid Il2rg−/− mice engrafted with human mobilized peripheral-blood CD34+ stem cells.

    Article  CAS  PubMed  Google Scholar 

  17. Ishikawa, F. et al. Development of functional human blood and immune systems in NOD/SCID/IL2 receptor γ-chainnull mice. Blood 106, 1565–1573 (2005). The first paper to show that engraftment of newborn NOD/LtSz- scid Il2rg−/− mice with human CD34+ HSCs leads to the generation of a complete haematopoietic system, including red blood cells and platelets.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Sugamura, K. et al. The interleukin-2 receptor γ-chain: its role in the multiple cytokine receptor complexes and T cell development in XSCID. Annu. Rev. Immunol. 14, 179–205 (1996).

    Article  CAS  PubMed  Google Scholar 

  19. Cao, X. et al. Defective lymphoid development in mice lacking expression of the common cytokine receptor γ-chain. Immunity 2, 223–238 (1995).

    Article  CAS  PubMed  Google Scholar 

  20. DiSanto, J. P., Muller, W., Guy-Grand, D., Fischer, A. & Rajewsky, K. Lymphoid development in mice with a targeted deletion of the interleukin 2 receptor γ-chain. Proc. Natl Acad. Sci. USA 92, 377–381 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Ohbo, K. et al. Modulation of hematopoiesis in mice with a truncated mutant of the interleukin-2 receptor γ-chain. Blood 87, 956–967 (1996).

    CAS  PubMed  Google Scholar 

  22. Jacobs, H. et al. PIM1 reconstitutes thymus cellularity in interleukin-7- and common γ-chain-mutant mice and permits thymocyte maturation in Rag- but not CD3γ-deficient mice. J. Exp. Med. 190, 1059–1068 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Yahata, T. et al. A highly sensitive strategy for SCID-repopulating cell assay by direct injection of primitive human hematopoietic cells into NOD/SCID mice bone marrow. Blood 101, 2905–2913 (2003).

    Article  CAS  PubMed  Google Scholar 

  24. Gimeno, R. et al. Monitoring the effect of gene silencing by RNA interference in human CD34+ cells injected into newborn Rag2−/−γ c−/− mice: functional inactivation of p53 in developing T cells. Blood 104, 3886–3893 (2004).

    Article  CAS  PubMed  Google Scholar 

  25. Bryder, D., Rossi, D. J. & Weissman, I. L. Hematopoietic stem cells: the paradigmatic tissue-specific stem cell. Am. J. Pathol. 169, 338–346 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Zanjani, E. D., Almeida-Porada, G. & Flake, A. W. Retention and multilineage expression of human hematopoietic stem cells in human–sheep chimeras. Stem Cells 13, 101–111 (1995).

    Article  CAS  PubMed  Google Scholar 

  27. Clutterbuck, R. D. et al. Studies on the development of human acute myeloid leukaemia xenografts in immune-deprived mice: comparison with cells in short-term culture. Leuk. Res. 9, 1511–1518 (1985).

    Article  CAS  PubMed  Google Scholar 

  28. Ganick, D. J., Sarnwick, R. D., Shahidi, N. T. & Manning, D. D. Inability of intravenously injected monocellular suspensions of human bone marrow to establish in the nude mouse. Int. Arch. Allergy Appl. Immunol. 62, 330–333 (1980).

    Article  CAS  PubMed  Google Scholar 

  29. Shultz, L. D. et al. NOD/LtSz-Rag1null mice: an immunodeficient and radioresistant model for engraftment of human hematolymphoid cells, HIV infection, and adoptive transfer of NOD mouse diabetogenic T cells. J. Immunol. 164, 2496–2507 (2000).

    Article  CAS  PubMed  Google Scholar 

  30. Mazurier, F., Doedens, M., Gan, O. I. & Dick, J. E. Rapid myeloerythroid repopulation after intrafemoral transplantation of NOD-SCID mice reveals a new class of human stem cells. Nature Med. 9, 959–963 (2003).

    Article  CAS  PubMed  Google Scholar 

  31. Schoeberlein, A. et al. Engraftment kinetics of human cord blood and murine fetal liver stem cells following in utero transplantation into immunodeficient mice. Stem Cells Dev. 13, 677–684 (2004).

    Article  PubMed  Google Scholar 

  32. Glimm, H. et al. Previously undetected human hematopoietic cell populations with short- term repopulating activity selectively engraft NOD/SCID-β2 microglobulin-null mice. J. Clin. Invest. 107, 199–206 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Miyoshi, H., Smith, K. A., Mosier, D. E., Verma, I. M. & Torbett, B. E. Transduction of human CD34+ cells that mediate long-term engraftment of NOD/SCID mice by HIV vectors. Science 283, 682–686 (1999).

    Article  CAS  PubMed  Google Scholar 

  34. Cohen-Haguenauer, O. et al. In vivo repopulation ability of genetically corrected bone marrow cells from Fanconi anemia patients. Proc. Natl Acad. Sci. USA 103, 2340–2345 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Bleeker, W. K. et al. Dual mode of action of a human anti-epidermal growth factor receptor monoclonal antibody for cancer therapy. J. Immunol. 173, 4699–4707 (2004).

    Article  CAS  PubMed  Google Scholar 

  36. Kolar, G. R., Yokota, T., Rossi, M. I., Nath, S. K. & Capra, J. D. Human fetal, cord blood, and adult lymphocyte progenitors have similar potential for generating B cells with a diverse immunoglobulin repertoire. Blood 104, 2981–2987 (2004).

    Article  CAS  PubMed  Google Scholar 

  37. Yahata, T. et al. Functional human T lymphocyte development from cord blood CD34+ cells in nonobese diabetic/Shi-scid, IL-2 receptor-γ-null mice. J. Immunol. 169, 204–209 (2002).

    Article  CAS  PubMed  Google Scholar 

  38. Legrand, N., Weijer, K. & Spits, H. Experimental models to study development and function of the human immune system in vivo. J. Immunol. 176, 2053–2058 (2006).

    Article  CAS  PubMed  Google Scholar 

  39. Matsumura, T. et al. Functional CD5+ B cells develop predominantly in the spleen of NOD/SCID/γcnull (NOG) mice transplanted either with human umbilical cord blood, bone marrow, or mobilized peripheral blood CD34+ cells. Exp. Hematol. 31, 789–797 (2003).

    Article  PubMed  Google Scholar 

  40. Legrand, N. et al. Transient accumulation of human mature thymocytes and regulatory T cells with CD28 superagonist in 'human immune system' Rag2−/−γ c−/− mice. Blood 108, 238–245 (2006).

    Article  CAS  PubMed  Google Scholar 

  41. Banuelos, S. J. et al. Rejection of human islets and human HLA-A2.1-transgenic mouse islets by alloreactive human lymphocytes in immunodeficient NOD-scid and NOD-Rag1nullPrf1null mice. Clin. Immunol. 112, 273–283 (2004).

    Article  CAS  PubMed  Google Scholar 

  42. Camacho, R. E. et al. Intra-thymic/splenic engraftment of human T cells in HLA-DR1 transgenic NOD/scid mice. Cell. Immunol. 232, 86–95 (2004).

    Article  CAS  PubMed  Google Scholar 

  43. Bajenoff, M. et al. Stromal cell networks regulate lymphocyte entry, migration, and territoriality in lymph nodes. Immunity 25, 989–1001 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Suematsu, S. & Watanabe, T. Generation of a synthetic lymphoid tissue-like organoid in mice. Nature Biotechnol. 22, 1539–1545 (2004).

    Article  CAS  Google Scholar 

  45. Melkus, M. W. et al. Humanized mice mount specific adaptive and innate immune responses to EBV and TSST-1. Nature Med. 12, 1316–1322 (2006).

    Article  CAS  PubMed  Google Scholar 

  46. Ifversen, P. & Borrebaeck, C. A. SCID-hu-PBL: a model for making human antibodies? Semin. Immunol. 8, 243–248 (1996).

    Article  CAS  PubMed  Google Scholar 

  47. Murphy, W. J., Taub, D. D. & Longo, D. L. The huPBL-SCID mouse as a means to examine human immune function in vivo. Semin. Immunol. 8, 233–241 (1996).

    Article  CAS  PubMed  Google Scholar 

  48. Shultz, L. D. et al. NOD/LtSz-Rag1nullPfpnull mice: a new model system to increase levels of human peripheral leukocyte and hematopoietic stem cell engraftment. Transplantation 76, 1036–1042 (2003).

    Article  PubMed  Google Scholar 

  49. Christianson, S. W. et al. Enhanced human CD4+ T cell engraftment in β2-microglobulin-deficient NOD-scid mice. J. Immunol. 158, 3578–3586 (1997).

    CAS  PubMed  Google Scholar 

  50. Tary-Lehmann, M., Lehmann, P. V., Schols, D., Roncarolo, M. G. & Saxon, A. Anti-SCID mouse reactivity shapes the human CD4+ T cell repertoire in hu-PBL-SCID chimeras. J. Exp. Med. 180, 1817–1827 (1994).

    Article  CAS  PubMed  Google Scholar 

  51. Greiner, D. L. & Shultz, L. D. in NOD Mice and Related Strains: Research Applications in Diabetes, AIDS, Cancer and Other Diseases (eds Leiter, E. & Atkinson, M.) 173–203 (Landes Bioscience, Austin, 1998).

    Google Scholar 

  52. May, K. F. Jr et al. Anti-human CTLA-4 monoclonal antibody promotes T-cell expansion and immunity in a hu-PBL-SCID model: a new method for preclinical screening of costimulatory monoclonal antibodies. Blood 105, 1114–1120 (2005).

    Article  CAS  PubMed  Google Scholar 

  53. Murray, A. G. et al. Human T-cell-mediated destruction of allogeneic dermal microvessels in a severe combined immunodeficient mouse. Proc. Natl Acad. Sci. USA 91, 9146–9150 (1994).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Chang, S. K., Arendt, B. K., Darce, J. R., Wu, X. & Jelinek, D. F. A role for BLyS in the activation of innate immune cells. Blood 108, 2687–2694 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Woodland, R. T. & Schmidt, M. R. Homeostatic proliferation of B cells. Semin. Immunol. 17, 209–217 (2005).

    Article  CAS  PubMed  Google Scholar 

  56. Mosier, D. E. et al. EBV-induced human B cell lymphomas in hu-PBL-SCID mice. AIDS Res. Hum. Retroviruses 8, 735–740 (1992).

    Article  CAS  PubMed  Google Scholar 

  57. Wagar, E. J. et al. Regulation of human cell engraftment and development of EBV-related lymphoproliferative disorders in Hu-PBL-scid mice. J. Immunol. 165, 518–527 (2000).

    Article  CAS  PubMed  Google Scholar 

  58. van Rijn, R. S. et al. A new xenograft model for graft-versus-host disease by intravenous transfer of human peripheral blood mononuclear cells in Rag2−/−γ c−/− double-mutant mice. Blood 102, 2522–2531 (2003).

    Article  CAS  PubMed  Google Scholar 

  59. Jung, S. et al. In vivo depletion of CD11c+ dendritic cells abrogates priming of CD8+ T cells by exogenous cell-associated antigens. Immunity 17, 211–220 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Mosier, D. E. Human immunodeficiency virus infection of human cells transplanted to severe combined immunodeficient mice. Adv. Immunol. 63, 79–125 (1996).

    Article  CAS  PubMed  Google Scholar 

  61. McCune, J. et al. The SCID-hu mouse: a small animal model for HIV infection and pathogenesis. Annu. Rev. Immunol. 9, 399–429 (1991).

    Article  CAS  PubMed  Google Scholar 

  62. Davis, P. H. & Stanley, S. L. Jr Breaking the species barrier: use of SCID mouse–human chimeras for the study of human infectious diseases. Cel. Microbiol. 5, 849–860 (2003).

    Article  CAS  Google Scholar 

  63. Bente, D. A., Melkus, M. W., Garcia, J. V. & Rico-Hesse, R. Dengue fever in humanized NOD/SCID mice. J. Virol. 79, 13797–13799 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Islas-Ohlmayer, M. et al. Experimental infection of NOD/SCID mice reconstituted with human CD34+ cells with Epstein–Barr virus. J. Virol. 78, 13891–13900 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Kneteman, N. M. et al. Anti-HCV therapies in chimeric scid-Alb/uPA mice parallel outcomes in human clinical application. Hepatology 43, 1346–1353 (2006).

    Article  CAS  PubMed  Google Scholar 

  66. Nelson, F. K., Greiner, D. L., Shultz, L. D. & Rajan, T. V. The immunodeficient scid mouse as a model for human lymphatic filariasis. J. Exp. Med. 173, 659–663 (1991).

    Article  CAS  PubMed  Google Scholar 

  67. Guirado, E. et al. Passive serum therapy with polyclonal antibodies against Mycobacterium tuberculosis protects against post-chemotherapy relapse of tuberculosis infection in SCID mice. Microbes Infect. 8, 1252–1259 (2006).

    Article  CAS  PubMed  Google Scholar 

  68. Morosan, S. et al. Liver-stage development of Plasmodium falciparum, in a humanized mouse model. J. Infect. Dis. 193, 996–1004 (2006).

    Article  CAS  PubMed  Google Scholar 

  69. Moreno, A. et al. The course of infections and pathology in immunomodulated NOD/LtSz-SCID mice inoculated with Plasmodium falciparum laboratory lines and clinical isolates. Int. J. Parasitol. 36, 361–369 (2006).

    Article  CAS  PubMed  Google Scholar 

  70. Macchiarini, F., Manz, M. G., Palucka, A. K. & Shultz, L. D. Humanized mice: are we there yet? J. Exp. Med. 202, 1307–1311 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Lapenta, C. et al. Pertussis toxin B-oligomer inhibits HIV infection and replication in hu-PBL-SCID mice. Int. Immunol. 17, 469–475 (2005).

    Article  CAS  PubMed  Google Scholar 

  72. Mosier, D. E. Viral pathogenesis in hu-PBL-SCID mice. Semin. Immunol. 8, 255–262 (1996).

    Article  CAS  PubMed  Google Scholar 

  73. Ichiyama, K. et al. A duodenally absorbable CXC-chemokine receptor 4 antagonist, KRH-1636, exhibits a potent and selective anti-HIV-1 activity. Proc. Natl Acad. Sci. USA 100, 4185–4190 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Baenziger, S. et al. Disseminated and sustained HIV infection in CD34+ cord blood cell-transplanted Rag2−/−γc−/− mice. Proc. Natl Acad. Sci. USA 103, 15951–15956 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Berges, B. K., Wheat, W. H., Palmer, B. E., Connick, E. & Akkina, R. HIV-1 infection and CD4 T cell depletion in the humanized Rag2−/−γ c−/− (RAG-hu) mouse model. Retrovirology 3, 76 (2006).

    Article  PubMed  PubMed Central  Google Scholar 

  76. Watanabe, S. et al. Hematopoietic stem cell-engrafted NOD/SCID/Il2rγnull mice develop human lymphoid system and induce long-lasting HIV-1 infection with specific humoral immune responses. Blood 109, 212–218 (2007).

    Article  CAS  PubMed  Google Scholar 

  77. Petersen, J. S. et al. Transfer of type 1 (insulin-dependent) diabetes mellitus associated autoimmunity to mice with severe combined immunodeficiency (SCID). Diabetologia 36, 510–515 (1993).

    Article  CAS  PubMed  Google Scholar 

  78. Martin, A. et al. Characteristics of long-term human thyroid peroxidase autoantibody secretion in scid mice transplanted with lymphocytes from patients with autoimmune thyroiditis. Int. Arch. Allergy Immunol. 98, 317–323 (1992).

    Article  CAS  PubMed  Google Scholar 

  79. Davis, L. S. et al. Inflammation, immune reactivity, and angiogenesis in a severe combined immunodeficiency model of rheumatoid arthritis. Am. J. Pathol. 160, 357–367 (2002).

    Article  PubMed  PubMed Central  Google Scholar 

  80. Tighe, H. et al. Autoantibody production by severe combined immunodeficient mice reconstituted with synovial cells from rheumatoid arthritis patients. Eur. J. Immunol. 20, 1843–1848 (1990).

    Article  CAS  PubMed  Google Scholar 

  81. Gregersen, J. W., Holmes, S. & Fugger, L. Humanized animal models for autoimmune diseases. Tissue Antigens 63, 383–394 (2004).

    Article  CAS  PubMed  Google Scholar 

  82. Friese, M. A., Jensen, L. T., Willcox, N. & Fugger, L. Humanized mouse models for organ-specific autoimmune diseases. Curr. Opin. Immunol. 18, 704–709 (2006).

    Article  CAS  PubMed  Google Scholar 

  83. Takaki, T. et al. HLA-A*0201-restricted T cells from humanized NOD mice recognize autoantigens of potential clinical relevance to type 1 diabetes. J. Immunol. 176, 3257–3265 (2006).

    Article  CAS  PubMed  Google Scholar 

  84. Wen, L., Chen, N. Y., Tang, J., Sherwin, R. & Wong, F. S. The regulatory role of DR4 in a spontaneous diabetes DQ8 transgenic model. J. Clin. Invest. 107, 871–880 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Marron, M. P., Graser, R. T., Chapman, H. D. & Serreze, D. V. Functional evidence for the mediation of diabetogenic T cell responses by HLA-A2.1 MHC class I molecules through transgenic expression in NOD mice. Proc. Natl Acad. Sci. USA 99, 13753–13758 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Ellmerich, S. et al. High incidence of spontaneous disease in an HLA-DR15 and TCR transgenic multiple sclerosis model. J. Immunol. 174, 1938–1946 (2005).

    Article  CAS  PubMed  Google Scholar 

  87. Mallone, R. et al. Differential recognition and activation thresholds in human autoreactive GAD-specific T cells. Diabetes 53, 971–977 (2004).

    Article  CAS  PubMed  Google Scholar 

  88. Kent, S. C. et al. Expanded T cells from pancreatic lymph nodes of type 1 diabetic subjects recognize an insulin epitope. Nature 435, 224–228 (2005).

    Article  CAS  PubMed  Google Scholar 

  89. Holst, J., Vignali, K. M., Burton, A. R. & Vignali, D. A. Rapid analysis of T-cell selection in vivo using T cell-receptor retrogenic mice. Nature Methods 3, 191–197 (2006).

    Article  CAS  PubMed  Google Scholar 

  90. Fogh, J., Fogh, J. M. & Orfeo, T. One hundred and twenty-seven cultured human tumor cell lines producing tumors in nude mice. J. Natl Cancer Inst. 59, 221–226 (1977).

    Article  CAS  PubMed  Google Scholar 

  91. Hudson, W. A., Li, Q., Le, C. & Kersey, J. H. Xenotransplantation of human lymphoid malignancies is optimized in mice with multiple immunologic defects. Leukemia 12, 2029–2033 (1998).

    Article  CAS  PubMed  Google Scholar 

  92. Dick, J. E. & Lapidot, T. Biology of normal and acute myeloid leukemia stem cells. Int. J. Hematol. 82, 389–396 (2005).

    Article  CAS  PubMed  Google Scholar 

  93. Kawano, N. et al. Efficient engraftment of primary adult T-cell leukemia cells in newborn NOD/SCID/β2-microglobulinnull mice. Leukemia 19, 1384–1390 (2005).

    Article  CAS  PubMed  Google Scholar 

  94. Nakamura, Y. et al. Engraftment of NOD/SCID/γcnull mice with multilineage neoplastic cells from patients with juvenile myelomonocytic leukaemia. Br. J. Haematol. 130, 51–57 (2005).

    Article  PubMed  Google Scholar 

  95. Pardal, R., Clarke, M. F. & Morrison, S. J. Applying the principles of stem-cell biology to cancer. Nature Rev. Cancer 3, 895–902 (2003).

    Article  CAS  Google Scholar 

  96. Reya, T., Morrison, S. J., Clarke, M. F. & Weissman, I. L. Stem cells, cancer, and cancer stem cells. Nature 414, 105–111 (2001).

    Article  CAS  PubMed  Google Scholar 

  97. Bonnet, D. & Dick, J. E. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nature Med. 3, 730–737 (1997).

    Article  CAS  PubMed  Google Scholar 

  98. Pilarski, L. M. & Belch, A. R. Clonotypic myeloma cells able to xenograft myeloma to nonobese diabetic severe combined immunodeficient mice copurify with CD34+ hematopoietic progenitors. Clin. Cancer Res. 8, 3198–3204 (2002).

    PubMed  Google Scholar 

  99. Al-Hajj, M., Wicha, M. S., Benito-Hernandez, A., Morrison, S. J. & Clarke, M. F. Prospective identification of tumorigenic breast cancer cells. Proc. Natl Acad. Sci. USA 100, 3983–3988 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. O'Brien, C. A., Pollett, A., Gallinger, S. & Dick, J. E. A human colon cancer cell capable of initiating tumour growth in immunodeficient mice. Nature 445, 106–110 (2007).

    Article  CAS  PubMed  Google Scholar 

  101. Singh, S. K. et al. Identification of human brain tumour initiating cells. Nature 432, 396–401 (2004).

    Article  CAS  PubMed  Google Scholar 

  102. Yoshida, S. et al. Long-term engraftment and self-renewal of AML stem cells in the newborn NOD-scid/Il2rgnull immunodeficient mouse model. Blood 106, A1261 (2005).

    Google Scholar 

  103. Nomura, T. et al. Induction of cancer, actinic keratosis, and specific p53 mutations by UVB light in human skin maintained in severe combined immunodeficient mice. Cancer Res. 57, 2081–2084 (1997).

    CAS  PubMed  Google Scholar 

  104. Mitsiades, C. S. et al. Fluorescence imaging of multiple myeloma cells in a clinically relevant SCID/NOD in vivo model: biologic and clinical implications. Cancer Res. 63, 6689–6696 (2003).

    CAS  PubMed  Google Scholar 

  105. O'Reilly, M. S., Holmgren, L., Chen, C. & Folkman, J. Angiostatin induces and sustains dormancy of human primary tumors in mice. Nature Med. 2, 689–692 (1996).

    Article  CAS  PubMed  Google Scholar 

  106. Siegler, U. et al. Activated natural killer cells from patients with acute myeloid leukemia are cytotoxic against autologous leukemic blasts in NOD/SCID mice. Leukemia 19, 2215–2222 (2005).

    Article  CAS  PubMed  Google Scholar 

  107. Flavell, D. J. et al. The anti-CD20 antibody rituximab augments the immunospecific therapeutic effectiveness of an anti-CD19 immunotoxin directed against human B-cell lymphoma. Br. J. Haematol. 134, 157–170 (2006).

    Article  CAS  PubMed  Google Scholar 

  108. Trieu, Y. et al. Soluble interleukin-13Rα2 decoy receptor inhibits Hodgkin's lymphoma growth in vitro and in vivo. Cancer Res. 64, 3271–3275 (2004).

    Article  CAS  PubMed  Google Scholar 

  109. Dewan, M. Z. et al. Rapid tumor formation of human T-cell leukemia virus type 1-infected cell lines in novel NOD-SCID/γcnull mice: suppression by an inhibitor against NF-κB. J. Virol. 77, 5286–5294 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Watanabe, M. et al. A novel NF-κB inhibitor DHMEQ selectively targets constitutive NF-κB activity and induces apoptosis of multiple myeloma cells in vitro and in vivo. Int. J. Cancer 114, 32–38 (2005).

    Article  CAS  PubMed  Google Scholar 

  111. Chalandon, Y. et al. BCR–ABL-transduced human cord blood cells produce abnormal populations in immunodeficient mice. Leukemia 19, 442–448 (2005).

    Article  CAS  PubMed  Google Scholar 

  112. Serakinci, N. & Keith, W. N. Therapeutic potential of adult stem cells. Eur. J. Cancer 42, 1243–1246 (2006).

    Article  CAS  PubMed  Google Scholar 

  113. Solter, D. From teratocarcinomas to embryonic stem cells and beyond: a history of embryonic stem cell research. Nature Rev. Genet. 7, 319–327 (2006).

    Article  CAS  PubMed  Google Scholar 

  114. Kakinuma, S. et al. Human umbilical cord blood as a source of transplantable hepatic progenitor cells. Stem Cells 21, 217–227 (2003).

    Article  PubMed  Google Scholar 

  115. Tateno, C. et al. Near completely humanized liver in mice shows human-type metabolic responses to drugs. Am. J. Pathol. 165, 901–912 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Ishikawa, F. et al. Purified human hematopoietic stem cells contribute to the generation of cardiomyocytes through cell fusion. FASEB J. 20, 950–952 (2006).

    Article  CAS  PubMed  Google Scholar 

  117. Ma, N. et al. Umbilical cord blood cell transplantation for myocardial regeneration. Transplant Proc. 38, 771–773 (2006).

    Article  CAS  PubMed  Google Scholar 

  118. Torrente, Y. et al. Human circulating AC133+ stem cells restore dystrophin expression and ameliorate function in dystrophic skeletal muscle. J. Clin. Invest. 114, 182–195 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Ishikawa, F. et al. Human cord blood- and bone marrow-derived CD34+ cells regenerate gastrointestinal epithelial cells. FASEB J. 18, 1958–1960 (2004).

    Article  CAS  PubMed  Google Scholar 

  120. Droetto, S. et al. Vasculogenic potential of long term repopulating cord blood progenitors. FASEB J. 18, 1273–1275 (2004).

    Article  CAS  PubMed  Google Scholar 

  121. Taguchi, A. et al. Administration of CD34+ cells after stroke enhances neurogenesis via angiogenesis in a mouse model. J. Clin. Invest. 114, 330–338 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Pauwelyn, K. A. & Verfaillie, C. M. Transplantation of undifferentiated, bone marrow-derived stem cells. Curr. Top. Dev. Biol. 74, 201–251 (2006).

    Article  CAS  PubMed  Google Scholar 

  123. Hess, D. et al. Bone marrow-derived stem cells initiate pancreatic regeneration. Nature Biotechnol. 21, 763–770 (2003).

    Article  CAS  Google Scholar 

  124. Yoshida, S. et al. Human cord blood-derived cells generate insulin-producing cells in vivo. Stem Cells 23, 1409–1416 (2005).

    Article  PubMed  Google Scholar 

  125. Vassilopoulos, G., Wang, P. R. & Russell, D. W. Transplanted bone marrow regenerates liver by cell fusion. Nature 422, 901–904 (2003).

    Article  CAS  PubMed  Google Scholar 

  126. Zhang, S. et al. Both cell fusion and transdifferentiation account for the transformation of human peripheral blood CD34+ cells into cardiomyocytes in vivo. Circulation 110, 3803–3807 (2004).

    Article  PubMed  Google Scholar 

  127. Stamm, C., Liebold, A., Steinhoff, G. & Strunk, D. Stem cell therapy for ischemic heart disease: beginning or end of the road? Cell Transplant. 15 (Suppl. 1), 47–56 (2006).

    Article  Google Scholar 

  128. Brolen, G. K., Heins, N., Edsbagge, J. & Semb, H. Signals from the embryonic mouse pancreas induce differentiation of human embryonic stem cells into insulin-producing β-cell-like cells. Diabetes 54, 2867–2874 (2005).

    Article  CAS  PubMed  Google Scholar 

  129. Cooke, M. J., Stojkovic, M. & Przyborski, S. A. Growth of teratomas derived from human pluripotent stem cells is influenced by the graft site. Stem Cells Dev. 15, 254–259 (2006).

    Article  CAS  PubMed  Google Scholar 

  130. Przyborski, S. A. Differentiation of human embryonic stem cells after transplantation in immune-deficient mice. Stem Cells 23, 1242–1250 (2005).

    Article  PubMed  Google Scholar 

  131. Wang, L. et al. Generation of hematopoietic repopulating cells from human embryonic stem cells independent of ectopic HOXB4 expression. J. Exp. Med. 201, 1603–1614 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Tian, X., Woll, P. S., Morris, J. K., Linehan, J. L. & Kaufman, D. S. Hematopoietic engraftment of human embryonic stem cell-derived cells is regulated by recipient innate immunity. Stem Cells 24, 1370–1380 (2006).

    Article  CAS  PubMed  Google Scholar 

  133. Galic, Z. et al. T-lineage differentiation from human embryonic stem cells. Proc. Natl Acad. Sci. USA 103, 11742–11747 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Robert, J. S. The science and ethics of making part-human animals in stem cell biology. FASEB J. 20, 838–845 (2006).

    Article  CAS  PubMed  Google Scholar 

  135. Sotiropoulou, P. A., Perez, S. A., Gritzapis, A. D., Baxevanis, C. N. & Papamichail, M. Interactions between human mesenchymal stem cells and natural killer cells. Stem Cells 24, 74–85 (2006).

    Article  PubMed  Google Scholar 

  136. Flanagan, S. P. 'Nude', a new hairless gene with pleiotropic effects in the mouse. Genet. Res. 8, 295–309 (1966).

    Article  CAS  PubMed  Google Scholar 

  137. Mosier, D. E., Stell, K. L., Gulizia, R. J., Torbett, B. E. & Gilmore, G. L. Homozygous scid/scid;beige/beige mice have low levels of spontaneous or neonatal T cell-induced B cell generation. J. Exp. Med. 177, 191–194 (1993).

    Article  CAS  PubMed  Google Scholar 

  138. Lapidot, T. Mechanism of human stem cell migration and repopulation of NOD/SCID and B2mnull NOD/SCID mice. The role of SDF-1/CXCR4 interactions. Ann. NY Acad. Sci. 938, 83–95 (2001).

    Article  CAS  PubMed  Google Scholar 

  139. Nicolini, F. E., Cashman, J. D., Hogge, D. E., Humphries, R. K. & Eaves, C. J. NOD/SCID mice engineered to express human IL-3, GM-CSF and Steel factor constitutively mobilize engrafted human progenitors and compromise human stem cell regeneration. Leukemia 18, 341–347 (2004).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank E. Leiter, D. Serreze, M. Berry and A. Rossini for valuable discussions. We are supported by the β-Cell Biology Consortium and Autoimmunity Prevention Centers of the National Institutes of Health (NIH; USA), the Juvenile Diabetes Research Foundation (International), the American Diabetes Association, and the Diabetes Endocrinology Research Center, the National Cancer Institute Center, the National Institute of Allergy and Infectious Diseases and the National Heart, Lung and Blood Institute of the NIH. The contents of this publication are solely the responsibility of the authors and do not necessarily represent the official views of the NIH.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Leonard D. Shultz.

Ethics declarations

Competing interests

Leonard D. Shultz and Dale L. Greiner are consultants for and receive financial support from Viacell, Inc. and Pfizer, Inc.

Related links

Related links

FURTHER INFORMATION

The Jackson Laboratory

Leonard Shultz's homepage

Glossary

Severe combined immunodeficiency

(scid). Mice homozygous for the scid mutation at the protein kinase, DNA activated, catalytic polypeptide (Prkdcscid) locus have a complete absence of mature T and B cells.

Leakiness

The spontaneous rearrangement of T- and B-cell receptors in scid mice, leading to the generation of mature T and B cells.

Recombination-activating gene 1 (Rag1) and Rag2

Two linked genes in which targeted mutations result in the complete inability to generate T and B cells expressing antigen-specific receptors.

Nude mice

Mice homozygous for a mutation in the forkhead box N1 (Foxn1) gene, which causes both hairlessness and impaired development of the thymus, resulting in an inability to generate mature T cells.

SCID-hu mice

Severe combined immunodeficiency (scid) mice engrafted with human fetal liver and thymus tissue under the renal capsule.

Hu-SRC-SCID mice

Severe combined immunodeficiency (scid) mice that have been sublethally irradiated and injected with human haematopoietic stem cells (HSCs). The HSCs are obtained from three main sources: bone marrow, umbilical cord blood or granulocyte colony-stimulating factor (G-CSF)-mobilized peripheral blood, which contains large numbers of human HSCs.

Xenoreactivity

An immune reactivity of cells or antibody from one animal directed against cells or tissues of a different species.

NOD/SCID-hu BLT mice

(BLT mice). Non-obese diabetic (NOD)-severe combined immunodeficiency (scid) mice engrafted with human fetal liver (L) and thymus (T) under the renal capsule. Three weeks later, mice are irradiated and then injected with a suspension of CD34+ cells from the same human fetal liver sample. The injected fetal liver cells seed to the mouse bone marrow (B).

Graves' disease

A type of autoimmune disease in which autoantibodies produced by the immune system overstimulate the thyroid gland, causing hyperthyroidism.

Retrogenic

A term used for T-cell receptor (TCR)-transgenic mice generated by retrovirus-mediated transduction of haematopoietic stem cells (HSCs) with a vector carrying a TCR transgene. These transduced HSCs are then injected into conditioned mice to reconstitute the mice with T cells expressing the TCR transgene.

Transdifferentiation

Refers to the ability of a non-stem cell to transform into a different type of cell lineage, or when an already partly differentiated stem cell creates cells of different lineages or cell types.

Teratoma

A tumour that derives from pluripotent germ cells, comprising disorganized tissues derived from all three embryonic germ layers (ectoderm, mesoderm and endoderm). It can arise spontaneously in the human gonads.

Myocardial infarction

An episode of acute cardiac ischaemia that leads to death of heart-muscle cells. It is usually caused by a thrombotic atherosclerotic plaque.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Shultz, L., Ishikawa, F. & Greiner, D. Humanized mice in translational biomedical research. Nat Rev Immunol 7, 118–130 (2007). https://doi.org/10.1038/nri2017

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nri2017

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