Elsevier

Human Immunology

Volume 70, Issue 10, October 2009, Pages 790-802
Human Immunology

Comparison of human fetal liver, umbilical cord blood, and adult blood hematopoietic stem cell engraftment in NOD-scid/γc−/−, Balb/c-Rag1−/−γc−/−, and C.B-17-scid/bg immunodeficient mice

https://doi.org/10.1016/j.humimm.2009.06.005Get rights and content

Abstract

Immunodeficient mice bearing components of a human immune system present a novel approach for studying human immune responses. We investigated the number, phenotype, developmental kinetics, and function of developing human immune cells following transfer of CD34+ hematopoietic stem cell (HSC) preparations originating from second trimester human fetal liver (HFL), umbilical cord blood (UCB), or granulocyte colony-stimulating factor-mobilized adult blood (G-CSF-AB) delivered via intrahepatic injection into sublethally irradiated neonatal NOD-scid/γc−/−, Balb/c-Rag1−/−γc−/−, and C.B-17-scid/bg mice. HFL and UCB HSC provided the greatest number and breadth of developing cells. NOD-scid/γc−/− and Balb/c-Rag1−/−γc−/− harbored human B and dendritic cells as well as human platelets in peripheral blood, whereas NOD-scid/γc−/− mice harbored higher levels of human T cells. NOD-scid/γc−/− mice engrafted with HFL CD34+ HSC demonstrated human immunological competence evidenced by white pulp expansion and increases in total human immunoglobulin following immunization with T-dependent antigens and delayed-type hypersensitivity-infiltrating leukocytes in response to antigenic challenge. In conclusion, we describe an encouraging base system for studying human hematopoietic lineage development and function utilizing human HFL or UCB HSC-engrafted NOD-scid/γc−/− mice that is well suited for future studies toward the development of a fully competent humanized mouse model.

Introduction

Immunodeficient mice harboring human cells or tissues, frequently referred to as “humanized mice,” are promising tools for studying complex processes in human biology. Mice bearing human immune systems, in particular, are being developed to investigate immune-mediated disease pathogenesis [1], [2] and allogeneic tissue rejection and tolerance in vivo[3], [4], [5]. Two decades ago, the original “SCID-hu” model was developed by McCune and co-workers using C.B-17-scid mice as recipients for human hematopoietic tissues including fetal liver, bone, and/or thymus originating from second-trimester human fetuses [6], [7], [8]. The engrafted human hematopoietic tissues gave rise to low levels of human T and B cells that were capable of producing a primary antibody response when autologous fetal skin, serving as an additional source of dendritic cells, was coengrafted along with thymus, bone marrow, and lymph node [9]. Adoptive transfer of peripheral blood mononuclear cells (PBMC) in this same mouse strain supported low levels of engraftment of T, B and dendritic cells [10]. Further compromising the innate immune system of C.B-17-scid animals by either introducing a beige mutation or crossing with mice of the NOD background allowed recipients to accept higher levels of mature human T and B cells [11], [12].

Two major technical advances enhance the applicability of humanized mice for studying the human immune system. First, isolated hematopoietic stem cells (HSC) engrafted in C.B-17-scid [[13], [14]] and NOD-scid mice [14]. Second, the development of more severely immunodeficient mouse lines lacking the common cytokine receptor gamma chain (γc) [15], [16] and thus more profoundly deficient in the innate immune system supported even higher levels of HSC engraftment and de novo differentiation of T, B, and dendritic cells [17]. Most recent work has centered on HSC engraftment in neonatal γc-deficient mice on either the NOD-scid[18] or the Balb/c-Rag1−/−[19] background. Although such animals demonstrate varying levels of splenic lymphoid development, human immune system function is incomplete. Nevertheless, these systems offer great promise for routine use as humanized mouse models, especially if HSC can be reproducibly generated from embryonic stem cells [20], [21] or inducible pluripotent stem cells [22]. In addition to the requirement of severe defects in both innate and adaptive immunity in the recipient mouse, engraftment efficiency also appears dependent on the genetic background and age of the recipient mouse, route of engraftment and conditioning regime, and type and individual aliquot of injected HSC (for review, see [2]). We were interested in comparing the NOD-scid/γc−/− and Balb/c-Rag1−/−γc−/− mouse strains for preparing humanized mice for our ongoing transplant immunology and disease pathogenesis studies. To this end, we performed a side-by-side comparison of the kinetics and breadth of the developing immune system following intrahepatic, neonatal transplantation using matched aliquots of HSC originating from human fetal liver (HFL), umbilical cord blood (UCB), and granulocyte colony-stimulating factor-mobilized adult blood (G-CSF-AB) in NOD-scid/γc−/− and Balb/c-Rag1−/−γc−/− mice and also considered engraftment of our currently utilized strain, C.B-17-scid/bg.

Section snippets

Mice

Mice were housed in approved facilities at the Yale Animal Resources Center and handled according to Guide for the Care and Use of Laboratory Animals. All experimentation was approved by the Yale Institutional Animal Care and Use Committee. The original breeding pairs of NOD-scid/γc−/− and Balb/c-Rag1−/−γc−/− mice were generous donations from Dr. Leonard Shultz at The Jackson Laboratories (Bar Harbor, ME) and Dr. Drew Pardoll, Johns Hopkins University, respectively. C.B-17-scid/bg breeders were

Characterization of HSC preparations

To assess the potential origin of human leukocytes detected in HSC-reconstituted mice, each inoculum was analyzed for lineage marker expression (Table 1). Eighty-four percent of G-CSF-AB-column cells expressed human CD34, and 9.3 and 6.3% of cells in the preparation were CD3+ and CD11c+, reflecting contaminating T cells and DCs, respectively. Fewer numbers of cells in the preparation (<2%) were either CD19+ B cells or CD56+ natural killer (NK) cells (Table 1). In contrast, 92% of

Discussion

In this study, we compared the engraftment of human CD34+ HSC in NOD-scid/γc−/−, Balb/c-Rag1−/−γc−/−, and C.B-17-scid/bg mice. To allow direct comparisons in these three mouse strains, a number of critical parameters were held constant throughout the study, including an established conditioning regime for each mouse strain, an intrahepatic injection site, age at the time of engraftment, and an identical number of input cells from each HSC source in a given experimental cohort of mice.

Acknowledgments

We thank Dr. Leonard Shultz, Jackson Laboratories, and Dr. Drew Pardoll, Johns Hopkins University for the donations of breeding pairs of NOD-scid−/− and Balb/c-Rag1γ−/− mice, respectively; Drs. Li Wen and Sara Rockwell for the donations of control NOD and Balb/c mice, respectively; Dr. Bradford Poulos of the Human Fetal Tissue Repository, Albert Einstein College of Medicine, for human fetal liver tissue; Dr. Diane Krause and Wendy Haskell at the Yale Center of Excellence in Molecular

References (44)

  • W. Li et al.

    The SLAM-associated protein signaling pathway is required for development of CD4+ T cells selected by homotypic thymocyte interaction

    Immunity

    (2007)
  • M.W. Melkus et al.

    Humanized mice mount specific adaptive and innate immune responses to EBV and TSST-1

    Nat Med

    (2006)
  • L.D. Shultz et al.

    Humanized mice in translational biomedical research

    Nat Rev Immunol

    (2007)
  • T.M. Zollner et al.

    Proteasome inhibition reduces superantigen-mediated T cell activation and the severity of psoriasis in a SCID-hu model

    J Clin Invest

    (2002)
  • J.S. Pober et al.

    Immunopathology of human T cell responses to skin, artery and endothelial cell grafts in the human peripheral blood lymphocyte/severe combined immunodeficient mouse

    Springer Semin Immunopathol

    (2003)
  • J.M. McCune et al.

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

    Science

    (1988)
  • R. Namikawa et al.

    Long-term human hematopoiesis in the SCID-hu mouse

    J Exp Med

    (1990)
  • J.M. Carballido et al.

    Generation of primary antigen-specific human T- and B-cell responses in immunocompetent SCID-hu mice

    Nat Med

    (2000)
  • D.E. Mosier et al.

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

    Nature

    (1988)
  • R.M. Hesselton et al.

    High levels of human peripheral blood mononuclear cell engraftment and enhanced susceptibility to human immunodeficiency virus type 1 infection in NOD/LtSz-scid/scid mice

    J Infect Dis

    (1995)
  • D.A. Tereb et al.

    Human T cells infiltrate and injure pig coronary artery grafts with activated but not quiescent endothelium in immunodeficient mouse hosts

    Transplantation

    (2001)
  • T. Lapidot et al.

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

    Science

    (1992)
  • Cited by (0)

    View full text