Parameters for establishing humanized mouse models to study human immunity: Analysis of human hematopoietic stem cell engraftment in three immunodeficient strains of mice bearing the IL2rγnull mutation

https://doi.org/10.1016/j.clim.2009.12.008Get rights and content

Abstract

“Humanized” mouse models created by engraftment of immunodeficient mice with human hematolymphoid cells or tissues are an emerging technology with broad appeal across multiple biomedical disciplines. However, investigators wishing to utilize humanized mice with engrafted functional human immune systems are faced with a myriad of variables to consider. In this study, we analyze HSC engraftment methodologies using three immunodeficient mouse strains harboring the IL2rγnull mutation; NOD-scid IL2rγnull, NOD-Rag1null IL2rγnull, and BALB/c-Rag1null IL2rγnull mice. Strategies compared engraftment of human HSC derived from umbilical cord blood following intravenous injection into adult mice and intracardiac and intrahepatic injection into newborn mice. We observed that newborn recipients exhibited enhanced engraftment as compared to adult recipients. Irrespective of the protocol or age of recipient, both immunodeficient NOD strains support enhanced hematopoietic cell engraftment as compared to the BALB/c strain. Our data define key parameters for establishing humanized mouse models to study human immunity.

Introduction

The discovery of the severe combined immunodeficiency mutation (Prkdcscid, abbreviated as “scid”) in a stock of C.B-17 mice in 1983 [1] led to the assessment of their ability to support engraftment with human peripheral blood mononuclear cells (PBMC, termed Hu-PBL-SCID) [2] or fetal (termed SCID-Hu) [3] or adult (termed Hu-SRC-SCID) [4] hematopoietic stem cells (HSC). These human hematolymphoid engrafted mouse models have been used in multiple biomedical disciplines for the study of human immunobiology and hematopoiesis (reviewed in refs. [5–8]). However, the utility of the early strains of immunodeficient mice transplanted with human hematolymphoid cells was hindered by the inability to achieve multilineage hematopoietic engraftment leading to the generation of a functional human immune system [5], [6]. In the Hu-PBL-SCID model, although all lineages of peripheral blood mononuclear cells (PBMC) are injected, the majority of human cells engrafted are T cells, with few B cells, myeloid cells or natural killer (NK) cells surviving. In the Hu-SRC-SCID model, the major obstacle was the inability of HSC-engrafted mice to develop all components of a functional human immune system, particularly T cells [5].

A major technological breakthrough occurred when investigators created genetic stocks of scid, Rag1null or Rag2null mice that also harbor mutations in the IL2 receptor common gamma chain (IL2rγ) gene [9], [10], [11], [12]. IL2rγ is required for high-affinity signaling through multiple cytokine receptors, including IL2, IL4, IL7, IL9, IL15 and IL21 [13]. Immunodeficient mice bearing a mutated IL2rγ gene support much higher levels of human hematolymphoid engraftment than all previous immunodeficient stocks. Multiple immune lineages develop following engraftment of human HSC that can lead to the generation of a functional human immune system [5], [6], [7], [8].

Confounding widespread establishment of humanized mouse models in multiple laboratories is the diversity of approaches that have been reported for engrafting human HSC. For example, IL2rγnull mice are available on a number of strain backgrounds. The ability of immunodeficient mice to support engraftment of human hematolymphoid cells has been shown to be strongly affected by the genetic background of the host [5], [14], [15], [16], [17]. In addition to strain background, reports differ significantly with regard to engraftment methodologies, which include intravenous (IV) engraftment into adult mice [11], or intrahepatic (IH) [10], intraperitoneal (IP) [12], and IV [9], [18] injection into newborn mice.

In the present study, we compared a number of variables of human HSC engraftment, including strain background, age of recipient, and engraftment route. The three strains of immunodeficient mice tested were NOD-scid IL2rγnull, NOD-Rag1null IL2rγnull, and BALB/c-Rag1null IL2rγnull. To provide a consistent comparison, we used human HSC derived from umbilical cord blood (UCB) in all of our experiments. We matched UCB donors within an individual experiment, allowing direct comparison of the engraftment characteristics in each strain, independent of donor-to-donor variability. We observed that in the variables tested, newborn immunodeficient NOD strain mice provided the optimal hosts with respect to their ability to support human hematopoietic cell engraftment.

Section snippets

Mice

NOD.Cg-Prkdcscid Il2rgtm1Wjl/Sz (NOD-scid IL2rγnull) and NOD.Cg-Rag1tm1Mom IL2rgtm1Wjl (NOD-Rag1null IL2rγnull) mice have been described previously [11], [19]. C.Cg-Rag1tm1Mom Il2rgtm1Wjl (BALB/c-Rag1null IL2rγnull) mice were generated by LDS by crossing C.129S7(B6)-Rag1tm1Mom mice (BALB/c-Rag1null) with C.129S4-Il2rg tm1Wjl(BALB/c-IL2rγnull) mice and then intercrossing the F1 progeny to fix the Rag1null and the IL2rγnull alleles to homozygosity. Mice were housed in a specific pathogen free

Development of BALB/c-Rag1null IL2rγnull Mice

The optimal mouse strains (described in Table 1) that support engraftment of human HSC harbor mutations in the IL2 receptor common gamma chain (IL2rγnull) and are available on a number of genetic backgrounds, including NOD-scid, NOD-Rag1null and BALB/c-Rag2null [10], [11], [19]. As the BALB/c-Rag2null IL2rγnull stocks are not commercially available, we generated a BALB/c-Rag1null IL2rγnull stock by crossing the BALB/c-Rag1null strain with the BALB/c-IL2rγnull strain. These parental stocks were

Discussion

In this study we have compared NOD-scid IL2rγnull, NOD-Rag1null IL2rγnull, and BALB/c-Rag1null IL2rγnull strains of mice for their ability to support engraftment of human HSC from the same cord blood donor within individual experiments. We observed that newborn mice engrafted at higher levels than adult mice and that in all parameters tested, NOD strain mice provided the optimal environment for engraftment of human hematopoietic cells. Although all strains of newborn mice engrafted with human

Acknowledgments

We thank Jean Leaf, Linda Paquin, and Allison Ingalls for technical assistance. This work was supported by National Institutes of Health research grants AI46629, DK53006, HL077642, CA34196, an institutional Diabetes Endocrinology Research Center (DERC) grant DK32520 and grants from the Beta Cell Biology Consortium of NIDDK, grants from the Juvenile Diabetes Foundation, International and a grant from the University of Massachusetts Center for AIDS Research, P30 AI042845. The contents of this

References (41)

  • R.S. van Rijn et al.

    A new xenograft model for graft-versus-host disease by intravenous transfer of human peripheral blood mononuclear cells in RAG2−/− double-mutant mice

    Blood

    (2003)
  • T. Yahata et al.

    Competitive repopulation assay of two gene-marked cord blood units in NOD/SCID/gammac(null) mice

    Mol. Ther.

    (2004)
  • E. Gluckman et al.

    Donor selection for unrelated cord blood transplants

    Curr. Opin. Immunol.

    (2006)
  • F. Ishikawa et al.

    An assay for long-term engrafting human hematopoietic cells based on newborn NOD/SCID/beta2-microglobulin(null) mice

    Exp. Hematol.

    (2002)
  • K. Ohbo et al.

    Modulation of hematopoiesis in mice with a truncated mutant of the interleukin-2 receptor gamma chain

    Blood

    (1996)
  • G.C. Bosma et al.

    A severe combined immunodeficiency mutation in the mouse

    Nature

    (1983)
  • D.E. Mosier et al.

    Transfer of a functional human immune system to mice with severe combined immunodeficiency

    Nature

    (1988)
  • J.M. McCune et al.

    The SCID-hu mouse: murine model for the analysis of human hematolymphoid differentiation and function

    Science

    (1988)
  • T. Lapidot et al.

    Cytokine stimulation of multilineage hematopoiesis from immature human cells engrafted in SCID mice

    Science

    (1992)
  • L.D. Shultz et al.

    Humanized mice in translational biomedical research

    Nat. Rev. Immunol.

    (2007)
  • Cited by (211)

    View all citing articles on Scopus
    View full text