Skip to main content
Cellular and Molecular Immunology logoLink to Cellular and Molecular Immunology
. 2012 Feb 13;9(3):225–231. doi: 10.1038/cmi.2011.64

Human allograft rejection in humanized mice: a historical perspective

Michael A Brehm 1, Leonard D Shultz 2
PMCID: PMC4012846  PMID: 22327213

Abstract

Basic research in transplantation immunology has relied primarily on rodent models. Experimentation with rodents has laid the foundation for our basic understanding of the biological events that precipitate rejection of non-self or allogeneic tissue transplants and supported the development of novel strategies to specifically suppress allogeneic immune responses. However, translation of these studies to the clinic has met with limited success, emphasizing the need for new models that focus on human immune responses to allogeneic tissues. Humanized mouse models are an exciting alternative that permits investigation of the rejection of human tissues mediated by human immune cells without putting patients at risk. However, the use of humanized mice is complicated by a diversity of protocols and approaches, including the large number of immunodeficient mouse strains available, the choice of tissue to transplant and the specific human immune cell populations that can be engrafted. Here, we present a historical perspective on the study of allograft rejection in humanized mice and discuss the use of these novel model systems in transplant biology.

Keywords: transplantation, immunodeficient mice, human

Introduction

Transplantation of allogeneic or ‘non-self' tissues stimulates a robust immune response leading to graft rejection.1, 2, 3 The survival of allogeneic organ transplants requires lifelong immune suppression,4, 5, 6 although recent work has suggested that this may not be necessary in select populations of patients.7 Excellent outcomes notwithstanding, contemporary immunosuppressive medications are toxic, are often not taken by patients, and pose long-term risks of infection and malignancy.8, 9, 10 The ultimate goal in transplantation research is to develop new treatments that will supplant the need for general immunosuppression. While novel protocols have been developed using murine transplantation systems, translation of these approaches to the clinic has been limited. This failure is related to the species-specific differences in the rodent and human immune systems, as well as the difficulty of studying the generation of alloreactive immune responses in patients.

Humanized mice offer a novel approach to study human biology by implanting functional human cells and tissues into immunodeficient mice. Humanized mouse models have been utilized for a number of years in an attempt to understand the basic immunological mechanisms underlying allogeneic transplant rejection. However, these early studies suffered from low levels of chimerism and limited functionality of the engrafted human immune cells. Advancements in generating humanized mice have greatly enhanced the functionality of the engrafted human immune systems and have facilitated the study of human immunobiology. Recent studies have demonstrated robust rejection of human allografts, including skin, islet and artery grafts in humanized models. Moreover, these new model systems are proving to be valuable tools for the preclinical evaluation of human-specific therapeutics to induce transplantation tolerance.

Strain development for humanized mice

The successful transplantation of xenogeneic tissues into mice requires elimination or severe suppression of the murine innate and adaptive immune systems.11 Initial efforts to engraft human immune systems into mice focused on eliminating the adaptive host murine immune response by using the CB17-scid mouse, which has impaired development of B and T cells.12 The scid mutation is within the catalytic subunit of DNA-dependent protein kinase, which is required for the repair of double-stranded DNA breaks and carrying out V(D)J recombination.13, 14 The CB17-scid mouse will engraft with human peripheral blood mononuclear cells (PBMC), 15 hematopoietic stem cells (HSC)16 and fetal tissues,17 but the overall levels of engraftment are extremely low and the engrafted cells have minimal functionality. An alternative to eliminate murine adaptive immunity is the use of mice deficient in the expression of either recombination activating gene (Rag1) (Rag1null) or Rag2 (Rag2null).18, 19 Rag1 and Rag2 function synergistically to create double-stranded DNA breaks and are essential for V(D)J recombination and the development of functional T cells and B cells.20, 21 While these early immunodeficient mouse strains allowed for low-level human immune cell engraftment, the usefulness of these models to study human immunobiology and allograft rejection was limited.

The adaptive immune system is effectively eliminated in scid, Rag1null and Rag2null mice, but the murine innate immune system remains intact and prevents high-level engraftment of human HSC and immune cells. In an attempt to diminish the murine innate immune system, new genetic stocks of scid, Rag1null or Rag2null mice were created that also harbored targeted mutations in the IL2 receptor common gamma chain (IL2rγ) gene.22, 23, 24, 25, 26, 27, 28 The IL2rγ chain is required for high-affinity ligand binding and signaling through multiple cytokine receptors, including IL2, IL4, IL7, IL9, IL15 and IL21.29 Importantly, the absence of IL15 prevents the development of murine natural killer (NK) cells, which are extremely efficient in the rejection of non-self hematopoietic cells in vivo. Immunodeficient mice bearing mutations within the IL2rγ gene support significantly higher levels of human hematolymphoid engraftment than all previous immunodeficient stocks and allow for the development of a functional human immune system comprised of multiple lymphoid and myeloid cell lineages.

An additional variable that will significantly influence the engraftment of human cells and tissues into immunodeficient mice is the specific strain background of the recipient mouse. For example, immunodeficiency mutations on the nonobese diabetic (NOD) mouse background support higher engraftment levels of human hematopoietic cells and immune cells as compared to other backgrounds, such as BALB/c, C3H and C57BL/6.30, 31, 32, 33, 34, 35 The NOD mouse background offers a number of genetic advantages that promote the engraftment of human immune systems.34 A direct comparison of immunodeficient IL2rγnull mice on either a NOD background (NOD-scid IL2rγnull (NSG) and NOD-Rag1null IL2rγnull (NRG)) or BALB/c (BALB-Rag2null IL2rγnull) background revealed that the NOD background supported significantly higher levels of human cell engraftment following injection of human HSC.30, 32 NOD mice have a number of defects in innate immune functionality that may facilitate the engraftment of human cells, including reduced NK cell numbers and function, defects in macrophage function, impaired dendritic cell maturation and a lack of hemolytic complement. In addition, the signal regulatory protein-alpha (SIRPα) polymorphism that is expressed by phagocytic cells on the NOD background allows for heightened self-recognition interactions between murine phagocytic cells and human CD47 expressing hematopoietic cells, minimizing the phagocytosis of human cells.36 In contrast, BALB/c mice express a SIRPα polymorphism that is not protective against phagocytosis. Two recent studies using distinct approaches have demonstrated that the SIRPα–CD47 interactions are critically important for the engraftment of human HSC and immune cell development in IL2rγnull mice.37, 38 It was first shown that transgenic expression of human SIRPα by mice on a mixed (129×BALB/c) strain background significantly improved the engraftment of human HSC.38 The second study showed that retrogenic expression of murine CD47 in human HSC prior to transplantation significantly improved human immune system development.37 Overall, these studies highlight the importance of strain selection in the efficient generation of humanized mice.

Humanized mouse models

There are a variety of humanized mouse models that can be used to study immune cell function (Table 1), including the Hu-PBL-SCID, the Hu-SRC (scid-repopulating cell)-SCID and the SCID-Hu or BLT (bone marrow, liver, thymus) models.39 In the Hu-PBL-SCID model, mice are injected with mature human PBMC, which results in robust engraftment of human T cells, and provides a useful tool to examine the functionality of human T cells in vivo.40, 41 This mouse model has been successfully used to examine both allo-immunity and viral immunity and to recapitulate HIV infection.40, 42, 43, 44, 45 One complicating factor with the Hu-PBL-SCID model is the development of a xenogeneic graft-versus-host disease (GVHD) that develops with successful engraftment as human T cells recognize murine major histocompatibility complex.27, 41, 46, 47, 48 This GVHD limits the time frame of experiments that can be done with the HU-PBL-SCID mice, but T cells expanded during the xenogeneic reaction still are able to mediate rejection of human skin allografts.49 In the Hu-SRC-SCID model, preconditioned mice are injected with human HSC derived from a variety of sources, including umbilical cord blood, bone marrow, fetal liver and peripheral blood of granulocyte colony-stimulating factor-treated individuals. Human HSC engraft at high levels in NSG and NRG stocks, and these mice develop functional innate and adaptive human immune systems.30, 50 The Hu-SRC-SCID model has been used to study many aspects of human immunobiology, including infectious disease, transplantation rejection and immune responses.39, 51 In the SCID-Hu or BLT model, mice are implanted under the renal capsule with human fetal thymus and fetal liver.17, 52 In some instances CD34+ cells derived from the autologous fetal liver tissue are also injected intravenously to provide a peripheral source of HSC. BLT mice develop a functional immune system and have primarily been used to study HIV infection, but have also been used to study other aspects of human infectious and immunological disease.17, 52, 53, 54 The advantage of the BLT model is robust engraftment and the presence of autologous human thymic epithelium for T-cell development. The Hu-PBL-SCID and Hu-SRC-SCID models have been predominantly used in studies of human allograft rejection and have proven to be promising tools to investigate mechanisms of rejection.

Table 1. Humanized mouse engraftment models used to study human allograft rejection.

Humanized model Advantages Disadvantages Allograft rejection models
Hu-PBL-SCID Ease of engraftment T cell-dominated engraftment Human islet
  Consistent Xenogeneic GVHD Human skin
  Robust T-cell engraftment   Human artery
Hu-SRC-SCID Functional naive T cells and B cells Lack of human thymic epithelium for T-cell education Human islet
  Innate immune cell development Immature status of B cells Human skin (required injection of autologous PBMC)
      Human artery
SCID-Hu or BLT Functional naive T cells and B cells Immature status of B cells Non-human xenogeneic tissue grafts
  Innate immune cell development Development of GVHD as mice age  
  Human thymic epithelium for T-cell development    

Abbreviations: BLT, bone marrow, liver, thymus; GVHD, graft-versus-host disease; PBMC, peripheral blood mononuclear cells.

HUMAN SKIN ALLOGRAFTS

Human skin allografts have been used extensively to study transplantation immunology in humanized mouse models. There are numerous advantages for the use of human skin in these transplantation experiments. Tissue specimens from healthy individuals are readily available. Preparation of tissue and the transplant protocol is straightforward. The healed-in tissues resemble healthy human skin complete with epidermal and dermal layers and vasculature. It is also possible to recover autologous PBMC from the skin donor. Finally, skin allografts are highly immunogenic and stimulate robust immune responses.55

Early attempts to study rejection of human skin allografts in humanized mice utilized the CB17-scid host.56, 57, 58 In 1992, Kawamura and colleagues evaluated rejection of human skin allografts on CB17-scid mice engrafted with either PBMC from a donor that had been previously sensitized to alloantigen or with PBMC from a donor that had not been previously exposed to alloantigens. PBMC from the non-sensitized donor were unable to reject skin allografts in this model. In contrast, 37% of mice injected with PBMC from the presensitized donor rejected skin allografts from an individual that shared at least one human histocompatibility leukocyte antigen (HLA) allele with the sensitizing donor. During the rejection process human T cells were detectable within the human skin by immunohistochemistry. These findings suggested that PBMC injected into CB17-scid mice do not maintain functionality unless pre-existing alloreactive memory cells were present.

Although human PBMC engraft in CB17-scid mice, the levels of detectable human cells are extremely low. The next effort by transplantation biologists focused on improving engraftment and maintaining functionality of human immune cells following injection into CB17-scid mice. One approach to improve the model included the injection of extremely high numbers (3×108) of human PBMC and the depletion of host murine NK cells by treatment with anti-asialo GM1 polyclonal antibody.58 Although complete rejection of human skin allografts was not demonstrated with this approach, perivascular infiltrates of human T cells were consistently observed and damage to the human microvessels was evident in >95% of engrafted mice. A second strategy to improve the model was to irradiate (2 cGy) the CB17-scid mice prior to injection of human splenocytes.56 The injection of human splenocytes into irradiated hosts resulted in significantly higher levels of human cell engraftment as compared to human PBMC. Consistent with earlier studies, injection of human splenocytes resulted in primarily T-cell engraftment that consisted of both CD4 and CD8 T cells with an activated phenotype: CD25+, HLA-DR+, CD45RA+. Within 3 weeks of human skin transplantation, 75% of splenocyte-engrafted CB17-scid mice had completely rejected the skin allografts, and depletion of T cells prevented rejection. Together, these initial studies suggested that under the appropriate conditions the CB17-scid mouse model could be used to study the rejection of human skin allografts. However, this model required the injection of very high numbers of human cells and preconditioning with either irradiation or depletion of NK cells, and the levels of engraftment were still low.

The enhancement of human cell engraftment by NK cell depletion suggested that the innate immune system of CB17-scid mice was impeding human cell survival. In an effort to improve human cell engraftment and function in immunodeficient mice, Pober and colleagues initiated a series of studies using CB17-scid mice co-expressing the beige (Lystbg) mutation (SCID/beige).59, 60, 61 The beige mutation disrupts a gene required for lysosomal trafficiking and results in impaired NK cell function.62, 63 The use of SCID/beige mice allowed engraftment of human PBMC without depletion of NK cells, but still required the injection of high numbers of cells (1×108–3×108 cells). Moreover, SCID/beige mice readily accepted human skin grafts, with minimal injury or infiltration by murine immune cells.64 Injection of human PBMC or T-cell lines into SCID/beige mice bearing human skin allografts produced allograft injury that was characterized by vascular damage, and this injury was mediated by human CD4 and CD8 T cells.59, 60, 61 The SCID/beige model has since proven useful in characterizing allograft injury mediated by human T cells. Inhibition of human T-cell function by treatment of skin allograft bearing SCID/beige mice with a combination of cyclosporine and rapamycin significantly reduced the level of human cell infiltrate and microvascular damage.59 The blockade of CD2–CD58 interactions, which are important for T-cell interactions with antigen-presenting cells, using an antibody to CD58 or CD58-Ig fusion protein blocked T-cell infiltration of human skin allografts and prevented allograft injury.61 Costimulation blockade by treatment with monoclonal antibodies specific for 4-1BBL, ICOSL or OX40L was not effective in preventing T-cell infiltration of human skin allografts after PBMC injection, but all three treatments were able to diminish allograft injury, including endothelial injury and thrombosis. Moreover, blockade of 4-1BBL, ICOSL or OX40L also reduced the expression of Fas ligand and perforin by the injected T cells.65

The SCID/beige model has also been used to define the role of human T-cell subsets in the rejection of human skin allografts. Preliminary experiments first tested the ability of CD45RA+ naive T cells or CD45RO+ effector T cells to mediate allograft injury.65 CD45RA+ and CD45RO+ T cells were purified by flow cytometry-based sorting and injected into SCID/beige mice. Although both T-cell subsets were able to engraft, only the CD45RO+ cells mediated vascular injury, suggesting that memory phenotype T cells were essential for the rejection of human skin. While CD45RA/RO status is not completely definitive to differentiate memory from naive T cells, greater than 90% of CD45RA+ CD4 T cells and between 50% and 60% of CD45RA+ CD8 T cells identified by this approach are functionally naive.66 More detailed analysis of T-cell subsets involved in skin allograft injury revealed that sort-purified effector memory CD4+ T cells (CD45RO+/CCR7/CD62L) mediated rejection of human skin allografts in SCID/beige mice, correlating with the ability of these cells to produce interferon (IFN)-γ in response to stimulation with human allogeneic endothelial cells.67 In contrast, central memory CD4 T cells (CD45RO+/CCR7+/CD62L+) were unable to engraft in SCID/beige mice after transfer, and therefore could not be evaluated for the ability to mediate allograft injury. Together, these data demonstrate that the SCID/beige mouse can be used to study mechanisms by which human T cells mediate injury to human skin allografts.

One limitation for the Hu-PBL-SCID model is that engraftment is dominated by T cells (Table 1), with minimal survival of innate immune cells and B cells.46 In an effort to develop a model that would allow the investigation of human macrophages in skin allograft rejection, adult SCID/beige mice were irradiated and injected with human CD34+ HSC derived from peripheral blood of granulocyte colony-stimulating factor-mobilized individuals to allow the development of a complete human immune system (Hu-SRC-SCID model).64 Unfortunately, SCID/beige mice engrafted only at very low levels with human immune cells in the blood, spleen and bone marrow, but human macrophages (CD68+ and CD14+) were detectable. Following transplantation of human skin allografts onto HSC-engrafted SCID/beige mice, human macrophages were detectable within the graft, but there was no evidence of graft injury. However, injection of autologous PBMC into the HSC-engrafted SCID/beige mice strongly activated the macrophages within the skin allograft and significantly enhanced allograft injury, as compared to mice engrafted with HSC or PBMC alone.64 Overall, these studies indicate that the SCID/beige mouse can be used to study the biology of human skin allograft rejection, but this model is limited by relatively low-level engraftment of human immune cells and the inability to develop a complete human immune system after HSC engraftment.

As described above the NOD background supports high-level engraftment with human cells and tissues. Initial studies on skin allograft rejection in immunodeficient mice on the NOD background were done in NOD-scid mice that were deficient in the expression of β2-microglobulin (Β2mnull).68 The β2-microglobulin deficiency impairs cell surface expression of major histocompatibility complex class I, depressing NK cell development and enhancing human immune cell engraftment.69, 70 For these experiments, NOD-scid B2mnull mice were first depleted of NK cells by pre-treatment with a monoclonal antibody specific for murine CD122. NK cell-depleted mice were then transplanted with human skin allografts and injected with human PBMC or splenocytes. All NOD-scid B2mnull mice injected with human cells were engrafted at high levels and demonstrated significant graft injury characterized by erythema, thrombosis and epithelial sloughing. Antibody-mediated in vivo depletion of human CD4 and CD8 T cells demonstrated that both T-cell subsets could mediate skin allograft rejection. This study demonstrated that T cell-mediated allograft rejection could be studied in immunodeficient mice on a NOD background.68

NSG mice engraft at very high levels with human PBMC in the absence of pretreatment with irradiation or NK cell-depleting antibodies,25, 27, 40 and appeared to be an excellent potential model to study skin allograft rejection mediated by human PBMC. However, human skin grafts on unmanipulated NSG mice showed extensive perivascular infiltration of murine immune cells that resulted in graft injury and precluded the study of rejection mediated by human immune cells.44 The depletion of Gr1+ murine innate immune cells from the recipient NSG mice allowed transplanted human skin to heal efficiently and significantly improved the overall graft morphology, as evident by the maintenance of epidermal and dermal structures. Moreover, human ‘passenger' leukocytes were readily detectable in human skin grafts on Gr1-depleted NSG, an observation that had not been reported previously. Injection of human PBMC into GR1-depleted NSG mice bearing human skin allografts resulted in severe graft injury within 21 days. This early graft injury was characterized by the remodeling of epidermal and dermal layers, extensive destruction of human endothelium, and a human CD45 infiltrate. By 31 days after PBMC injection, all engrafted mice showed extensive destruction of grafted tissues, with near complete loss of human vasculature and destruction of epidermal and dermal layers. Both human CD4 and CD8 T cells were able to mediate the rejection of human skin allografts.44 The high engraftment of human cells and consistent rejection of human skin allografts in NSG mice and the recent description of additional studies done in other IL2rγnull mouse models71, 72 demonstrate the power and utility of this model system to study transplantation rejection.

Human islet allografts

β-cell replacement therapies are the ultimate goal to cure diabetes.73, 74, 75 One example of a β-cell replacement therapy is human islet transplantation, which offers the advantage of a minimally invasive surgical procedure with limited complications.76, 77, 78 However, to date, the long-term success rate of islet transplantation has not reached the same level as that for pancreatic organ transplants for reasons that are not clear.79 Humanized mouse models have been used to study the rejection of allogeneic human islets and are a valuable resource as a preclinical model for investigating mechanisms of rejection and how to impede them.

As with the study of human skin allograft rejection, the initial experiments to study rejection of human islet transplants were done in CB17-scid mice.80, 81 In 1991, London and colleagues demonstrated that the transplantation of human islets to the renal subcapsular space restored normoglycemia in CB17-scid mice that had been rendered diabetic by the injection of streptozotocin (STZ).80 Injection of human splenocytes allogeneic to the implanted islets resulted in rejection within 7 days. The rejection was characterized by strong infiltration of human CD8 T cells at the graft site. In contrast, human islet grafts were not rejected following the injection of autologous splenocytes. Levels of human cell engraftment were not shown in this manuscript, but based on our current knowledge the levels would be predicted to be low. To improve engraftment of human immune cells in CB17-scid mice, a subsequent study showed that an initial injection of human PBMC followed 2 days later by an injection of autologous PBMC that had been stimulated in vitro with anti-CD3 antibody resulted in higher levels of peripheral engraftment as compared to a single injection.81 Human islet allografts transplanted into CB17-scid mice in this two-stage injection model were rejected within 21 days of implant as determined by a loss of detectable human C-peptide. In contrast, human C-peptide was detectable for over 60 days in unmanipulated CB17-scid transplanted with human islets. Moreover, human T cells recovered from the graft site of PBMC-injected mice showed cytotoxic activity against allogeneic targets cells that shared HLA with the implanted islets, confirming the activation of alloreactive T cells.81

A similar two-stage injection protocol of human PBMC was used in NOD-scid mice to demonstrate human islet allograft rejection.82 In these experiments, NOD-scid mice were first transplanted with human islet allografts and then 3 days later injected with PBMC that were activated by in vitro stimulation with anti-CD3 antibody. Between 2 and 4 days later, these mice received a second injection with PBMC that had been stimulated in vitro with irradiated allogeneic splenocytes from the islet donor. Detection of human C-peptide was significantly reduced in the PBMC-injected mice by day 28 and was undetectable by day 42, indicating that the injected cells were able to reject the graft. Rejection of human islet allografts was prevented by the co-injection of T cell-depleted bone marrow derived from the islet donor, suggesting that the establishment of microchimerism was sufficient to induce tolerance in this humanized model.82 The NOD-scid mouse has also been used to study the rejection of human islet allografts after depletion of murine NK cells with a monoclonal antibody specific for CD122.42 In this study, transplantation of human islet allografts into STZ-treated diabetic NOD-scid mice efficiently restored normoglycemia. Injection of human PBMC that were allogeneic to the graft into the islet-implanted mice resulted in a reversion to hyperglycemia, indicating that the allografts were rejected. Histological evaluation of the graft site revealed a loss of insulin-positive cells and an extensive human T-cell infiltrate, confirming rejection. As an alternative to NOD-scid mice to study allograft rejection, perforin-deficient NOD-Rag1null Prf1null mice were shown to support human PBMC engraftment in the absence of NK cell-depletion and to allow the rejection of HLA-A2 expressing murine islets, isolated from NOD-scid HLA-A2 transgenic mice, by A2-negative human PBMC, but not by A2 positive PBMC.42

As described above, NSG mice support high-level engraftment of human PBMC and allow the study of human T-cell function, following injection of small numbers of human PBMC. Transplantation of human islet allografts into STZ-treated diabetic NSG mice was effective in regulating blood glucose levels but co-injection of human PBMC that were allogeneic to the islets resulted in a rapid reversion to hyperglycemia and a loss of detectable human C-peptide.40 The rejection was confirmed histologically by the absence of insulin-positive cells at the graft site and the presence of a marked cellular infiltrate. This study suggests that immunodeficient mice bearing mutations within the IL2rγ chain are the ideal hosts to study islet allograft rejection in the Hu-PBL-SCID model.

As described above, a limitation of the Hu-PBL-SCID model is the engraftment of predominately T cells. Two recent studies have evaluated the rejection of human islet allografts in the Hu-SRC-SCID model with mixed results.83, 84 Balb/c-Rag2null IL2rγnull mice engrafted with human fetal liver-derived HSC failed to reject human islet allografts.83 Human C-peptide levels were not reduced in HSC-engrafted mice by 35 days postislet transplant, insulin-positive cells were detectable at the graft site and there was minimal human cell infiltration. In contrast, recently published data from our laboratory demonstrated human islet allograft rejection in an NRG Ins2Akita mouse strain that spontaneously develops hyperglycemia.84 NRG mice heterozygous for the Ins2Akita mutation develop a spontaneous hyperglycemia due to misfolding of the insulin-2 protein, leading to endoplasmic reticulum stress and β-cell apoptosis.85, 86, 87 Importantly, the β-cell death occurs in the absence of an immune response and does not require the administration of β-cell toxins such as STZ, which have detrimental effects on many other tissues.88 Euglycemia can be restored following subrenal capsule transplantation with mouse or human islets.84 NRG Ins2Akita mice were injected with human HSC as newborns, and mice that engrafted with human immune systems were transplanted with allogeneic human islets. By 30 days post-transplant, 60% of islet transplanted HSC-engrafted NRG-Akita mice that had initially become normoglycemic reverted back to a hyperglycemic state, and histological evaluation of the grafts confirmed a marked human cell infiltration and loss of most insulin-positive cells. These results are consistent with an immune-mediated rejection of the allogeneic human islets. Interestingly in HSC-engrafted NRG-Akita mice that did not revert back to hyperglycemia, histological examination revealed a human mononuclear cell infiltration at the graft site, but these mice still had abundant insulin-positive cells. Thus, the Hu-SRC-SCID mouse model can be used to study human islet rejection, but this system still requires optimization.

Human artery allografts

A critical target during acute and chronic rejection of allogeneic tissues is the allograft blood vessels, specifically the endothelial cell lining.89 Endothelial cells strongly activate human T cells and are susceptible to direct injury mediated by effector T cells. SCID/beige mice have been used extensively in the Hu-PBL-SCID model to study the process of human vascular rejection.90

In 1999, Pober and colleagues demonstrated that human arterial grafts could be interposed into the murine infrarenal artery of SCID/beige mice and the tissues would remain viable.91 By 28 days after injection of human PBMC allogeneic to the arterial graft, the majority of mice showed severe human cell infiltration of the allograft and histological changes consistent with vascular rejection. IFN-γ production was shown to be critical during the rejection of human artery allografts, mediating vascular dysfunction.92 Transforming growth factor-β produced by arterial allografts diminished the ability of human T cells to produce IFN-γ, and blockade of transforming growth factor-β enhanced vascular rejection.93 In contrast, blockade of IL-1 with a human-specific IL-1R antagonist, diminished the infiltration of human immune cells and IL17 production and minimized arterial graft injury, suggesting the IL1α produced by the human arterial allograft promotes vascular rejection.94 The use of arterial grafts with pre-existing modest atherosclerosis did not accelerate T cell-mediated vascular rejection, suggesting that atherosclerotic plaques do not stimulate more robust immune responses.95 Finally, the Hu-SRC-SCID model has been used to study artery graft rejection in the SCID/beige model.64 As described above, injection of human HSC into SCID/beige mice results in low overall engraftment characterized by the lack of T cells or B cells, but human macrophage are detectable. Human artery allografts were rapidly infiltrated with human macrophages following transplant on HSC-engrafted SCID/beige mice and were extensively calcified. Together, these studies demonstrated that humanized models can be used to study the mechanisms of artery graft rejection.

Conclusions

Significant progress has been made in the study of human allograft rejection in humanized mouse models. New mouse strains, such as those bearing mutations in the IL2rγ chain, allow for optimal engraftment of functional human immune systems and the ability to consistently observe allograft rejection. These advanced model systems are currently being used to test novel immune therapies to suppress rejections. For example, four recent studies have shown that in vitro expanding human T regulatory cells will prevent rejection of human skin and artery grafts in the Hu-PBL-SCID model.71, 72, 96, 97 Efforts are continuing to improve humanized mouse models by creating new immunodeficient strains that express HLA and human cytokines and growth factor to enhance human immune system development.11, 98

Acknowledgments

This work was supported by National Institutes of Health research grants AI46629, HL077642, CA34196, DK089572, an institutional Diabetes Endocrinology Research Center (DERC) grant DK32520, a grant from the University of Massachusetts Center for AIDS Research, P30 AI042845 and grants from the JDRF, the Helmsley Foundation and USAMRIID. The contents of this publication are solely the responsibility of the authors and do not necessarily represent the official views of the National Institutes of Health.

References

  1. Lechler RI, Sykes M, Thomson AW, Turka LA. Organ transplantation—how much of the promise has been realized. Nat Med. 2005;11:605–613. doi: 10.1038/nm1251. [DOI] [PubMed] [Google Scholar]
  2. Suchin EJ, Langmuir PB, Palmer E, Sayegh MH, Wells AD, Turka LA. Quantifying the frequency of alloreactive T cells in vivo: new answers to an old question. J Immunol. 2001;166:973–981. doi: 10.4049/jimmunol.166.2.973. [DOI] [PubMed] [Google Scholar]
  3. Heeger PS. T-cell allorecognition and transplant rejection: a summary and update. Am J Transplant. 2003;3:525–533. doi: 10.1034/j.1600-6143.2003.00123.x. [DOI] [PubMed] [Google Scholar]
  4. Chen G, Dong JH. Individualized immunosuppression: new strategies from pharmacokinetics, pharmacodynamics and pharmacogenomics. Hepatobiliary Pancreat Dis Int. 2005;4:332–338. [PubMed] [Google Scholar]
  5. Odorico JS, Sollinger HW. Technical and immunosuppressive advances in transplantation for insulin-dependent diabetes mellitus. World J Surg. 2002;26:194–211. doi: 10.1007/s00268-001-0207-0. [DOI] [PubMed] [Google Scholar]
  6. Shapiro R, Young JB, Milford EL, Trotter JF, Bustami RT, Leichtman AB. Immunosuppression: evolution in practice and trends, 1993–2003. Am J Transplant. 2005;5:874–886. doi: 10.1111/j.1600-6135.2005.00833.x. [DOI] [PubMed] [Google Scholar]
  7. Newell KA, Asare A, Kirk AD, Gisler TD, Bourcier K, Suthanthiran M, et al. Identification of a B cell signature associated with renal transplant tolerance in humans. J Clin Invest. 2010;120:1836–1847. doi: 10.1172/JCI39933. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Laederach-Hofmann K, Bunzel B. Noncompliance in organ transplant recipients: a literature review. Gen Hosp Psychiatry. 2000;22:412–424. doi: 10.1016/s0163-8343(00)00098-0. [DOI] [PubMed] [Google Scholar]
  9. Soulillou JP, Giral M. Controlling the incidence of infection and malignancy by modifying immunosuppression. Transplantation. 2001;72:S89–S93. [PubMed] [Google Scholar]
  10. St Clair EW, Turka LA, Saxon A, Matthews JB, Sayegh MH, Eisenbarth GS, et al. New reagents on the horizon for immune tolerance. Annu Rev Med. 2007;58:329–346. doi: 10.1146/annurev.med.58.061705.145449. [DOI] [PubMed] [Google Scholar]
  11. Shultz LD, Ishikawa F, Greiner DL. Humanized mice in translational biomedical research. Nat Rev Immunol. 2007;7:118–130. doi: 10.1038/nri2017. [DOI] [PubMed] [Google Scholar]
  12. Bosma GC, Custer RP, Bosma MJ. A severe combined immunodeficiency mutation in the mouse. Nature. 1983;301:527–530. doi: 10.1038/301527a0. [DOI] [PubMed] [Google Scholar]
  13. Blunt T, Gell D, Fox M, Taccioli GE, Lehmann AR, Jackson SP, et al. Identification of a nonsense mutation in the carboxyl-terminal region of DNA-dependent protein kinase catalytic subunit in the scid mouse. Proc Natl Acad Sci USA. 1996;93:10285–10290. doi: 10.1073/pnas.93.19.10285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Fulop GM, Phillips RA. The scid mutation in mice causes a general defect in DNA repair. Nature. 1990;347:479–482. doi: 10.1038/347479a0. [DOI] [PubMed] [Google Scholar]
  15. Mosier DE, Gulizia RJ, Baird SM, Wilson DB. Transfer of a functional human immune system to mice with severe combined immunodeficiency. Nature. 1988;335:256–259. doi: 10.1038/335256a0. [DOI] [PubMed] [Google Scholar]
  16. Lapidot T, Pflumio F, Doedens M, Murdoch B, Williams DE, Dick JE. Cytokine stimulation of multilineage hematopoiesis from immature human cells engrafted in SCID mice. Science. 1992;255:1137–1141. doi: 10.1126/science.1372131. [DOI] [PubMed] [Google Scholar]
  17. McCune JM, Namikawa R, Kaneshima H, Shultz LD, Lieberman M, Weissman IL. The SCID-hu mouse: murine model for the analysis of human hematolymphoid differentiation and function. Science. 1988;241:1632–1639. doi: 10.1126/science.241.4873.1632. [DOI] [PubMed] [Google Scholar]
  18. Shinkai Y, Rathbun G, Lam KP, Oltz EM, Stewart V, Mendelsohn M, et al. RAG-2-deficient mice lack mature lymphocytes owing to inability to initiate V(D)J rearrangement. Cell. 1992;68:855–867. doi: 10.1016/0092-8674(92)90029-c. [DOI] [PubMed] [Google Scholar]
  19. Mombaerts P, Iacomini J, Johnson RS, Herrup K, Tonegawa S, Papaioannou VE. RAG-1-deficient mice have no mature B and T lymphocytes. Cell. 1992;68:869–877. doi: 10.1016/0092-8674(92)90030-g. [DOI] [PubMed] [Google Scholar]
  20. Oettinger MA, Schatz DG, Gorka C, Baltimore D. RAG-1 and RAG-2, adjacent genes that synergistically activate V(D)J recombination. Science. 1990;248:1517–1523. doi: 10.1126/science.2360047. [DOI] [PubMed] [Google Scholar]
  21. Gellert M. V(D)J recombination: RAG proteins, repair factors, and regulation. Annu Rev Biochem. 2002;71:101–132. doi: 10.1146/annurev.biochem.71.090501.150203. [DOI] [PubMed] [Google Scholar]
  22. Ishikawa F, Yasukawa M, Lyons B, Yoshida S, Miyamoto T, Yoshimoto G, et al. Development of functional human blood and immune systems in NOD/SCID/IL2 receptor γ chainnull mice. Blood. 2005;106:1565–1573. doi: 10.1182/blood-2005-02-0516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Ito M, Hiramatsu H, Kobayashi K, Suzue K, Kawahata M, Hioki K, et al. NOD/SCID/γcnull mouse: an excellent recipient mouse model for engraftment of human cells. Blood. 2002;100:3175–3182. doi: 10.1182/blood-2001-12-0207. [DOI] [PubMed] [Google Scholar]
  24. Legrand N, Weijer K, Spits H. Experimental models to study development and function of the human immune system in vivo. . J Immunol. 2006;176:2053–2058. doi: 10.4049/jimmunol.176.4.2053. [DOI] [PubMed] [Google Scholar]
  25. Shultz LD, Lyons BL, Burzenski LM, Gott B, Chen X, Chaleff S, et al. Human lymphoid and myeloid cell development in NOD/LtSz-scid IL2Rγnull mice engrafted with mobilized human hemopoietic stem cells. J Immunol. 2005;174:6477–6489. doi: 10.4049/jimmunol.174.10.6477. [DOI] [PubMed] [Google Scholar]
  26. Traggiai E, Chicha L, Mazzucchelli L, Bronz L, Piffaretti JC, Lanzavecchia A, et al. Development of a human adaptive immune system in cord blood cell-transplanted mice. Science. 2004;304:104–107. doi: 10.1126/science.1093933. [DOI] [PubMed] [Google Scholar]
  27. van Rijn RS, Simonetti ER, Hagenbeek A, Hogenes MCH, de Weger RA, Canninga-van Dijk MR, et al. A new xenograft model for graft-versus-host disease by intravenous transfer of human peripheral blood mononuclear cells in RAG2−/− gammac−/− double-mutant mice. Blood. 2003;102:2522–2531. doi: 10.1182/blood-2002-10-3241. [DOI] [PubMed] [Google Scholar]
  28. Brehm MA, Shultz LD, Greiner DL. Humanized mouse models to study human diseases. Curr Opin Endocrinol Diabetes Obes. 2010;17:120–125. doi: 10.1097/MED.0b013e328337282f. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Rochman Y, Spolski R, Leonard WJ. New insights into the regulation of T cells by γc family cytokines. Nat Rev Immunol. 2009;9:480–490. doi: 10.1038/nri2580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Brehm MA, Cuthbert A, Yang C, Miller DM, Diiorio P, Laning J, et al. 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. Clin Immunol. 2010;135:84–98. doi: 10.1016/j.clim.2009.12.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Hesselton RM, Greiner DL, Mordes JP, Rajan TV, Sullivan JL, Shultz LD. 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;172:974–982. doi: 10.1093/infdis/172.4.974. [DOI] [PubMed] [Google Scholar]
  32. Lepus CM, Gibson TF, Gerber SA, Kawikova I, Szczepanik M, Ablamunits V, et al. Comparison of Human Fetal Liver, Umbilical Cord Blood, and Adult Blood Hematopoietic Stem Cell Engraftment in NOD-scid/γc−/−, Balb/c-Rag2−/−γc−/−, and C.B-17-scid/bg immunodeficient mice. Hum Immunol. 2009;70:790–802. doi: 10.1016/j.humimm.2009.06.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Shultz LD, Lang PA, Christianson SW, Gott B, Lyons B, Umeda S, 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. 2000;164:2496–2507. doi: 10.4049/jimmunol.164.5.2496. [DOI] [PubMed] [Google Scholar]
  34. Shultz LD, Schweitzer PA, Christianson SW, Gott B, Schweitzer IB, Tennent B, et al. Multiple defects in innate and adaptive immunologic function in NOD/LtSz-scid mice. J Immunol. 1995;154:180–191. [PubMed] [Google Scholar]
  35. Tournoy KG, Depraetere S, Pauwels RA, Leroux-Roels GG. Mouse strain and conditioning regimen determine survival and function of human leucocytes in immunodeficient mice. Clin Exp Immunol. 2000;119:231–239. doi: 10.1046/j.1365-2249.2000.01099.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Takenaka K, Prasolava TK, Wang JCY, Mortin-Toth SM, Khalouei S, Gan OI, et al. Polymorphism in Sirpa modulates engraftment of human hematopoietic stem cells. Nat Immunol. 2007;8:1313–1323. doi: 10.1038/ni1527. [DOI] [PubMed] [Google Scholar]
  37. Legrand N, Huntington ND, Nagasawa M, Bakker AQ, Schotte R, Strick-Marchand H, et al. Functional CD47/signal regulatory protein alpha (SIRPα) interaction is required for optimal human T- and natural killer- (NK) cell homeostasis in vivo. . Proc Natl Acad Sci USA. 2011;108:13224–13229. doi: 10.1073/pnas.1101398108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Strowig T, Rongvaux A, Rathinam C, Takizawa H, Borsotti C, Philbrick W, et al. Transgenic expression of human signal regulatory protein alpha in Rag2−/−γc−/− mice improves engraftment of human hematopoietic cells in humanized mice. Proc Natl Acad Sci USA. 2011;108:13218–13223. doi: 10.1073/pnas.1109769108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Shultz LD, Pearson T, King M, Giassi L, Carney L, Gott B, et al. Humanized NOD/LtSz-scid IL2 receptor common gamma chain knockout mice in diabetes research. Ann NY Acad Sci. 2007;1103:77–89. doi: 10.1196/annals.1394.002. [DOI] [PubMed] [Google Scholar]
  40. King M, Pearson T, Shultz LD, Leif J, Bottino R, Trucco M, et al. A new Hu-PBL model for the study of human islet alloreactivity based on NOD-scid mice bearing a targeted mutation in the IL-2 receptor gamma chain gene. Clin Immunol. 2008;126:303–314. doi: 10.1016/j.clim.2007.11.001. [DOI] [PubMed] [Google Scholar]
  41. King MA, Covassin L, Brehm MA, Racki W, Pearson T, Leif J, et al. Human peripheral blood leucocyte non-obese diabetic-severe combined immunodeficiency interleukin-2 receptor gamma chain gene mouse model of xenogeneic graft-versus-host-like disease and the role of host major histocompatibility complex. Clin Exp Immunol. 2009;157:104–118. doi: 10.1111/j.1365-2249.2009.03933.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Banuelos SJ, Shultz LD, Greiner DL, Burzenski LM, Gott B, Lyons BL, 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. 2004;112:273–283. doi: 10.1016/j.clim.2004.04.006. [DOI] [PubMed] [Google Scholar]
  43. Yu CI, Gallegos M, Marches F, Zurawski G, Ramilo O, García-Sastre A, et al. Broad influenza-specific CD8+ T-cell responses in humanized mice vaccinated with influenza virus vaccines. Blood. 2008;112:3671–3678. doi: 10.1182/blood-2008-05-157016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Racki WJ, Covassin L, Brehm M, Pino S, Ignotz R, Dunn R, et al. NOD-scid IL2rγnull mouse model of human skin transplantation and allograft rejection. Transplantation. 2010;89:527–536. doi: 10.1097/TP.0b013e3181c90242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Kumar P, Ban HS, Kim SS, Wu H, Pearson T, Greiner DL, et al. T cell-specific siRNA delivery suppresses HIV-1 infection in humanized mice. Cell. 2008;134:577–586. doi: 10.1016/j.cell.2008.06.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Covassin L, Laning J, Abdi R, Langevin DL, Phillips NE, Shultz LD, et al. Human peripheral blood CD4 T cell-engrafted non-obese diabetic-scid IL2rγnull H2-Ab1tm1Gru Tg (human leucocyte antigen D-related 4) mice: a mouse model of human allogeneic graft-versus-host disease. Clin Exp Immunol. 2011;166:269–280. doi: 10.1111/j.1365-2249.2011.04462.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Hoffmann-Fezer G, Gall C, Zengerle U, Kranz B, Thierfelder S. Immunohistology and immunocytology of human T-cell chimerism and graft-versus-host disease in SCID mice. Blood. 1993;81:3440–3448. [PubMed] [Google Scholar]
  48. Sandhu JS, Gorczynski R, Shpitz B, Gallinger S, Nguyen HP, Hozumi N. A human model of xenogeneic graft-versus-host disease in SCID mice engrafted with human peripheral blood lymphocytes. Transplantation. 1995;60:179–184. [PubMed] [Google Scholar]
  49. Huppes W, Hoffmann-Fezer G. Peripheral blood leukocyte grafts that induce human to mouse graft-vs.-host disease reject allogeneic human skin grafts. Am J Pathol. 1995;147:1708–1714. [PMC free article] [PubMed] [Google Scholar]
  50. Pearson T, Shultz LD, Miller D, King M, Laning J, Fodor W, et al. Non-obese diabetic-recombination activating gene-1 (NOD-Rag 1null) interleukin (IL)-2 receptor common gamma chain (IL 2 rγnull) null mice: a radioresistant model for human lymphohaematopoietic engraftment. Clin Exp Immunol. 2008;154:270–284. doi: 10.1111/j.1365-2249.2008.03753.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Legrand N, Ploss A, Balling R, Becker PD, Borsotti C, Brezillon N, et al. Humanized mice for modeling human infectious disease: challenges, progress, and outlook. Cell Host Microbe. 2009;6:5–9. doi: 10.1016/j.chom.2009.06.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Melkus MW, Estes JD, Padgett-Thomas A, Gatlin J, Denton PW, Othieno FA, et al. Humanized mice mount specific adaptive and innate immune responses to EBV and TSST-1. Nat Med. 2006;12:1316–1322. doi: 10.1038/nm1431. [DOI] [PubMed] [Google Scholar]
  53. Brainard D, Seung E, Frahm N, Cariappa A, Bailey C, Hart W, et al. Induction of robust cellular and humoral virus-specific adaptive immune responses in HIV-infected humanized BLT mice. J Virol. 2009;83:7305–7321. doi: 10.1128/JVI.02207-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Denton PW, Estes JD, Sun Z, Othieno FA, Wei BL, Wege AK, et al. Antiretroviral pre-exposure prophylaxis prevents vaginal transmission of HIV-1 in humanized BLT mice. PLoS Med. 2008;5:e16. doi: 10.1371/journal.pmed.0050016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Lee WP, Yaremchuk MJ, Pan YC, Randolph MA, Tan CM, Weiland AJ. Relative antigenicity of components of a vascularized limb allograft. Plast Reconstr Surg. 1991;87:401–411. doi: 10.1097/00006534-199103000-00001. [DOI] [PubMed] [Google Scholar]
  56. Alegre ML, Peterson LJ, Jeyarajah DR, Weiser M, Bluestone JA, Thistlethwaite JR. Severe combined immunodeficient mice engrafted with human splenocytes have functional human T cells and reject human allografts. J Immunol. 1994;153:2738–2749. [PubMed] [Google Scholar]
  57. Kawamura T, Niguma T, Fechner JH, Jr, Wolber R, Beeskau MA, Hullett DA, et al. Chronic human skin graft rejection in severe combined immunodeficient mice engrafted with human PBL from an HLA-presensitized donor. Transplantation. 1992;53:659–665. doi: 10.1097/00007890-199203000-00032. [DOI] [PubMed] [Google Scholar]
  58. Murray AG, Petzelbauer P, Hughes CC, Costa J, Askenase P, Pober JS. Human T-cell-mediated destruction of allogeneic dermal microvessels in a severe combined immunodeficient mouse. Proc Natl Acad Sci USA. 1994;91:9146–9150. doi: 10.1073/pnas.91.19.9146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Murray AG, Schechner JS, Epperson DE, Sultan P, McNiff JM, Hughes CC, et al. Dermal microvascular injury in the human peripheral blood lymphocyte reconstituted-severe combined immunodeficient (HuPBL-SCID) mouse/skin allograft model is T cell mediated and inhibited by a combination of cyclosporine and rapamycin. Am J Pathol. 1998;153:627–638. doi: 10.1016/S0002-9440(10)65604-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Rayner D, Nelson R, Murray AG. Noncytolytic human lymphocytes injure dermal microvessels in the huPBL-SCID skin graft model. Hum Immunol. 2001;62:598–606. doi: 10.1016/s0198-8859(01)00252-x. [DOI] [PubMed] [Google Scholar]
  61. Sultan P, Schechner JS, McNiff JM, Hochman PS, Hughes CC, Lorber MI, et al. Blockade of CD2-LFA-3 interactions protects human skin allografts in immunodeficient mouse/human chimeras. Nat Biotechnol. 1997;15:759–762. doi: 10.1038/nbt0897-759. [DOI] [PubMed] [Google Scholar]
  62. Mosier DE, Stell KL, Gulizia RJ, Torbett BE, Gilmore GL. Homozygous scid/scid;beige/beige mice have low levels of spontaneous or neonatal T cell-induced B cell generation. J Exp Med. 1993;177:191–194. doi: 10.1084/jem.177.1.191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Roder JC, Helfand SL, Werkmeister J, McGarry R, Beaumont TJ, Duwe A. Oxygen intermediates are triggered early in the cytolytic pathway of human NK cells. Nature. 1982;298:569–572. doi: 10.1038/298569a0. [DOI] [PubMed] [Google Scholar]
  64. Kirkiles-Smith NC, Harding MJ, Shepherd BR, Fader SA, Yi T, Wang Y, et al. Development of a humanized mouse model to study the role of macrophages in allograft injury. Transplantation. 2009;87:189–197. doi: 10.1097/TP.0b013e318192e05d. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Shiao SL, McNiff JM, Pober JS. Memory T cells and their costimulators in human allograft injury. J Immunol. 2005;175:4886–4896. doi: 10.4049/jimmunol.175.8.4886. [DOI] [PubMed] [Google Scholar]
  66. De Rosa SC, Herzenberg LA, Herzenberg LA, Roederer M. 11-color, 13-parameter flow cytometry: identification of human naive T cells by phenotype, function, and T-cell receptor diversity. Nat Med. 2001;7:245–248. doi: 10.1038/84701. [DOI] [PubMed] [Google Scholar]
  67. Shiao S, Kirkiles-Smith N, Shepherd B, Mcniff J, Carr E, Pober J. Human effector memory CD4+ T cells directly recognize allogeneic endothelial cells in vitro and in vivo. J Immunol. 2007;179:4397. doi: 10.4049/jimmunol.179.7.4397. [DOI] [PubMed] [Google Scholar]
  68. Turgeon NA, Banuelos SJ, Shultz LD, Lyons BL, Iwakoshi N, Greiner DL, et al. Alloimmune injury and rejection of human skin grafts on human peripheral blood lymphocyte-reconstituted non-obese diabetic severe combined immunodeficient beta2-microglobulin-null mice. Exp Biol Med (Maywood) 2003;228:1096–1104. doi: 10.1177/153537020322800918. [DOI] [PubMed] [Google Scholar]
  69. Christianson SW, Greiner DL, Hesselton RA, Leif JH, Wagar EJ, Schweitzer IB, et al. Enhanced human CD4+ T cell engraftment in beta2-microglobulin-deficient NOD-scid mice. J Immunol. 1997;158:3578–3586. [PubMed] [Google Scholar]
  70. Wagar EJ, Cromwell MA, Shultz LD, Woda BA, Sullivan JL, Hesselton RM, et al. Regulation of human cell engraftment and development of EBV-related lymphoproliferative disorders in Hu-PBL-scid mice. J Immunol. 2000;165:518–527. doi: 10.4049/jimmunol.165.1.518. [DOI] [PubMed] [Google Scholar]
  71. Issa F, Hester J, Goto R, Nadig SN, Goodacre TE, Wood K. Ex vivo-expanded human regulatory T cells prevent the rejection of skin allografts in a humanized mouse model. Transplantation. 2010;90:1321–1327. doi: 10.1097/TP.0b013e3181ff8772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Sagoo P, Ali N, Garg G, Nestle FO, Lechler RI, Lombardi G. Human regulatory T cells with alloantigen specificity are more potent inhibitors of alloimmune skin graft damage than polyclonal regulatory T cells. Sci Transl Med. 2011;3:83ra42. doi: 10.1126/scitranslmed.3002076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Halban PA, German MS, Kahn SE, Weir GC. Current status of islet cell replacement and regeneration therapy. J Clin Endocrinol Metab. 2010;95:1034–1043. doi: 10.1210/jc.2009-1819. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Naftanel MA, Harlan DM. Pancreatic islet transplantation. PLoS Med. 2004;1:e58; quiz e75. doi: 10.1371/journal.pmed.0010058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Harlan DM, Rother KI. Islet transplantation as a treatment for diabetes. N Engl J Med. 2004;350 doi: 10.1056/NEJM200405133502022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Harlan DM, Kenyon NS, Korsgren O, Roep BO. Current advances and travails in islet transplantation. Diabetes. 2009;58:2175–2184. doi: 10.2337/db09-0476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Rother KI, Harlan DM. Challenges facing islet transplantation for the treatment of type 1 diabetes mellitus. J Clin Invest. 2004;114:877–883. doi: 10.1172/JCI23235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Ricordi C, Strom TB. Clinical islet transplantation: advances and immunological challenges. Nat Rev Immunol. 2004;4:259–268. doi: 10.1038/nri1332. [DOI] [PubMed] [Google Scholar]
  79. Ryan EA, Paty BW, Senior PA, Bigam D, Alfadhli E, Kneteman NM, et al. Five-year follow-up after clinical islet transplantation. Diabetes. 2005;54:2060–2069. doi: 10.2337/diabetes.54.7.2060. [DOI] [PubMed] [Google Scholar]
  80. London NJ, Thirdborough SM, Swift SM, Bell PR, James RF. The diabetic ‘human reconstituted' severe combined immunodeficient (SCID-hu) mouse: a model for isogeneic, allogeneic, and xenogeneic human islet transplantation. Transplant Proc. 1991;23:749. [PubMed] [Google Scholar]
  81. Shiroki R, Poindexter NJ, Woodle ES, Hussain MS, Mohanakumar T, Scharp DW. Human peripheral blood lymphocyte reconstituted severe combined immunodeficient (hu-PBL-SCID) mice. A model for human islet allograft rejection. Transplantation. 1994;57:1555–1562. [PubMed] [Google Scholar]
  82. Mathew JM, Blomberg B, Ricordi C, Esquenazi V, Miller J. Evaluation of the tolerogenic effects of donor bone marrow cells using a severe combined immunodeficient mouse–human islet transplant model. Hum Immunol. 2008;69:605–613. doi: 10.1016/j.humimm.2008.07.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Jacobson S, Heuts F, Juarez J, Hultcrantz M, Korsgren O, Svensson M, et al. Alloreactivity but failure to reject human islet transplants by humanized Balb/c/Rag2−/−gc−/− mice. Scand J Immunol. 2010;71:83–90. doi: 10.1111/j.1365-3083.2009.02356.x. [DOI] [PubMed] [Google Scholar]
  84. Brehm MA, Bortell R, Diiorio P, Leif J, Laning J, Cuthbert A, et al. Human immune system development and rejection of human islet allografts in spontaneously diabetic NOD-Rag1null IL2rγnullIns2Akita mice. Diabetes. 2010;59:2265–2270. doi: 10.2337/db10-0323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Ron D. Proteotoxicity in the endoplasmic reticulum: lessons from the Akita diabetic mouse. J Clin Invest. 2002;109:443–445. doi: 10.1172/JCI15020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Mathews CE, Langley SH, Leiter EH. New mouse model to study islet transplantation in insulin-dependent diabetes mellitus. Transplantation. 2002;73:1333–1336. doi: 10.1097/00007890-200204270-00024. [DOI] [PubMed] [Google Scholar]
  87. Yoshioka M, Kayo T, Ikeda T, Koizumi A. A novel locus, Mody4, distal to D7Mit189 on chromosome 7 determines early-onset NIDDM in nonobese C57BL/6 (Akita) mutant mice. Diabetes. 1997;46:887–894. doi: 10.2337/diab.46.5.887. [DOI] [PubMed] [Google Scholar]
  88. Bolzan AD, Bianchi MS. Genotoxicity of streptozotocin. Mutat Res. 2002;512:121–134. doi: 10.1016/s1383-5742(02)00044-3. [DOI] [PubMed] [Google Scholar]
  89. Libby P, Pober JS. Chronic rejection. Immunity. 2001;14:387–397. doi: 10.1016/s1074-7613(01)00119-4. [DOI] [PubMed] [Google Scholar]
  90. Thomsen M, Galvani S, Canivet C, Kamar N, Bohler T. Reconstitution of immunodeficient SCID/beige mice with human cells: applications in preclinical studies. Toxicology. 2008;246:18–23. doi: 10.1016/j.tox.2007.10.017. [DOI] [PubMed] [Google Scholar]
  91. Lorber MI, Wilson JH, Robert ME, Schechner JS, Kirkiles N, Qian HY, et al. Human allogeneic vascular rejection after arterial transplantation and peripheral lymphoid reconstitution in severe combined immunodeficient mice. Transplantation. 1999;67:897–903. doi: 10.1097/00007890-199903270-00018. [DOI] [PubMed] [Google Scholar]
  92. Koh KP, Wang Y, Yi T, Shiao SL, Lorber MI, Sessa WC, et al. T cell-mediated vascular dysfunction of human allografts results from IFN-γdysregulation of NO synthase. J Clin Invest. 2004;114:846–856. doi: 10.1172/JCI21767. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Lebastchi AH, Khan SF, Qin L, Li W, Zhou J, Hibino N, et al. Transforming growth factor beta expression by human vascular cells inhibits interferon gamma production and arterial media injury by alloreactive memory T cells. Am J Transplant. 2011;11:2332–2341. doi: 10.1111/j.1600-6143.2011.03676.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Rao DA, Eid RE, Qin L, Yi T, Kirkiles-Smith NC, Tellides G, et al. Interleukin (IL)-1 promotes allogeneic T cell intimal infiltration and IL-17 production in a model of human artery rejection. J Exp Med. 2008;205:3145–3158. doi: 10.1084/jem.20081661. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Wang Y, Ahmad U, Yi T, Zhao L, Lorber MI, Pober JS, et al. Alloimmune-mediated vascular remodeling of human coronary artery grafts in immunodeficient mouse recipients is independent of preexisting atherosclerosis. Transplantation. 2007;83:1501–1505. doi: 10.1097/01.tp.0000264560.51845.67. [DOI] [PubMed] [Google Scholar]
  96. Feng G, Nadig SN, Backdahl L, Beck S, Francis RS, Schiopu A, et al. Functional regulatory T cells produced by inhibiting cyclic nucleotide phosphodiesterase type 3 prevent allograft rejection. Sci Transl Med. 2011;3:83ra40. doi: 10.1126/scitranslmed.3002099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Nadig SN, Wieckiewicz J, Wu DC, Warnecke G, Zhang W, Luo S, et al. In vivo prevention of transplant arteriosclerosis by ex vivo-expanded human regulatory T cells. Nat Med. 2010;16:809–813. doi: 10.1038/nm.2154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Willinger T, Rongvaux A, Strowig T, Manz MG, Flavell RA. Improving human hemato-lymphoid-system mice by cytokine knock-in gene replacement. Trends Immunol. 2011;32:321–327. doi: 10.1016/j.it.2011.04.005. [DOI] [PubMed] [Google Scholar]

Articles from Cellular and Molecular Immunology are provided here courtesy of Nature Publishing Group

RESOURCES