Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2011 Apr 1.
Published in final edited form as: Curr Opin Endocrinol Diabetes Obes. 2010 Apr;17(2):120–125. doi: 10.1097/MED.0b013e328337282f

Humanized Mouse Models to Study Human Diseases

Michael A Brehm 1, Leonard D Shultz 2, Dale L Greiner 1
PMCID: PMC2892284  NIHMSID: NIHMS213355  PMID: 20150806

Abstract

Purpose of review

Update on humanized mouse models and their use in biomedical research.

Recent findings

The recent description of immunodeficient mice bearing a mutated IL-2 receptor gamma chain (IL2ry) facilitated greatly the engraftment and function of human hematolymphoid cells and other cells and tissues. These mice permit the development of human immune systems, including functional T and B cells, following engraftment of hematopoietic stem cells (HSC). The engrafted functional human immune systems are capable of T and B cell-dependent immune responses, antibody production, anti-viral responses, and allograft rejection. Immunodeficient IL2rynull mice also support heightened engraftment of primary human cancers and malignant progenitor cells, permitting in vivo investigation of pathogenesis and function. In addition, human-specific infectious agents for which animal models were previously unavailable can now be studied in vivo using these new generation humanized mice.

Summary

Immunodeficient mice bearing an IL2rynull mutated gene can be engrafted with functional human cells and tissues, including human immune systems, following engraftment with human hematolymphoid cells. These mice are now used as in vivo models to study human hematopoiesis, immunity, regeneration, stem cell function, cancer, and human-specific infectious agents without putting patients at risk.

Keywords: IL2ry, NOD-SCID, NSG, Humanized mice

Introduction

Studies of human cell and tissue function have traditionally been limited to ex vivo analyses, non-invasive procedures, or clinical trials that are costly and severely limited due to ethical constraints. Small animal models of human cell and tissue function would overcome these limitations. A major breakthrough in the generation of humanized mice was the development of immunodeficient mice bearing a targeted IL2ry mutation. These mice permit functional in vivo studies of human cells and tissues [13]. This report summarizes recent progress in the use of humanized mice for the study of human diseases.

History of Humanized Mice

Human cell and tissue functions in murine hosts have been investigated since the first description of athymic (nude) mice [4]. Humanization of immunodeficient mice advanced greatly with the discovery of the severe combined immune deficiency (scid) mutation [5] and the knockout of recombination activating genes 1 and 2 [6;7]. Additional manipulation of the mouse genome by knockout and transgenic technology led to increased engraftment and function of human cells and tissues [1]. The recent description of immunodeficient mice bearing a mutated IL-2 receptor gamma chain (IL2rynull) facilitated greatly the in vivo engraftment and function of human cells. The history of humanized mice in biomedical research and the tremendous advances in this field resulting from the generation of immunodeficient mice with a mutated IL2ry gene has been reviewed recently [13]. The present review briefly summarizes the different models of humanized mice available for experiments, highlights recent advances in the model systems, and summarizes new findings that have emerged over the last year.

Models of Humanized Mice

We use a simple definition of humanized mice as “mice engrafted with functional human cells or tissues or expressing human transgenes.” Depending on the experimental question, different models of immunodeficient humanized mice are utilized (Table 1). Hu-PBL-SCID mice are engrafted with peripheral blood mononuclear cells [PBMC, 8]. Hu-SRC-SCID mice are engrafted with human hematopoietic stem cells [HSC, 9]. SCID-hu mice are engrafted with human fetal liver and thymus [10]. These models represent humanized mice engrafted with functional human immune systems. Immunocompetent mice expressing human transgenes also provide insights into human biology [1113]. This review will focus on humanized mice engrafted with functional human cells and tissues.

Table 1.

Models of humanized mice

Model Approach Characteristics Use
Hu-PBL-SCID (Human Peripheral Blood Lymphocyte) Engraftment of mature PBMC obtained from blood, spleen or lymph nodes Predominately engrafts human CD3 T cells. Study of mature T cell function Study of mature immune cell function
Hu-SRC-SCID (Human Scid Repopulating Cell) Engraftment of hematopoietic stem cells Develops human hematopoietic and naïve immune systems Study of complete hematopoietic system and naïve immune system
SCID-Hu Engraftment of human fetal liver and thymus Develops human hematopoietic and immune systems Study of complete hematopoietic system and naïve immune system
Hu-Tg Mice Transgenic expression of human genes Expresses specific human genes in vivo In vivo study of human gene function

Continuing Improvements in Humanized Mouse Technology

Human cell and tissue function was enhanced greatly by the generation of immunodeficient IL2rynull mice [13], but there remain a number of limitations that are continuously being recognized and overcome. One of the first questions in the field arose from the bewildering number of available mouse strains and engraftment protocols. Recognizing this problem, standardized methodologies for establishment of humanized mice were recently published [14;15]. Optimal approaches for engraftment of newborn and adult NOD-scid IL2rynull mice with HSC (Hu-SRC-SCID) as well as engraftment of human PBMC (Hu-PBL-SCID) are detailed by Pearson et al [14]. Optimal approaches for engraftment of BALB/c-Rag2null IL2rynull mice with human HSC (Hu-SRC-SCID) are described by Legrand et al [15]. It is recommended that investigators working with humanized mouse models initially establish the models based on these standardized guidelines.

The second question is the optimal recipient strain. HSC engraftment in different strains of immunodeficient mice following intrahepatic injection into newborns has been compared [16]. NOD-scid IL2rynull mice and BALB/c-Rag2null IL2rynull l mice are equivalent in their generation of human B cells, dendritic cells (DC), and platelets whereas NOD-scid IL2rynull mice are superior in supporting human T cell development. Fetal liver and umbilical cord blood (UCB) HSC supported higher percentages of human engraftment than G-CSF-mobilized peripheral blood HSCs. Bone marrow HSC was not tested in this report. We have confirmed that UCB HSC-engrafted newborn NOD-scid IL2rynull mice are superior to BALB/c-Rag1null IL2rynull mice in their ability to support human T cell development, and further found that intrahepatic and intracardiac (i.v.) injections are equivalent (Brehm et al, submitted). The NOD vs. BALB/c support of T cell engraftment is not based on the scid vs. Rag1/2null mutations as NOD-scid IL2rynull mice and NOD-Rag1null IL2rynull mice are equivalent in their support of human HSC engraftment [17]. These data suggest that investigators establishing humanized mice requiring a fully functional human immune system in the Hu-SRC-SCID model should consider basing their work on newborn engraftment of NOD-scid IL2rynull or NOD-Rag1null IL2rynull mice.

Another limitation being addressed is the species-specificity of a number of molecules. Examples include species-specific human cytokines. Transgenic expression of human IL15 enhanced human NK cell development and differentiation in HSC-engrafted BALB/c-Rag2null IL2rynull mice, and documented the critical role that IL15 trans-presentation has in regulating human NK cell homeostasis [18*]. A second cytokine, B Lymphocyte Stimulating factor (BLyS also termed BAFF) is important in B cell survival and differentiation [19;20]. Mouse BLyS cannot support human B cell survival, and administration of human recombinant BLyS to NOD-Rag1null Prf1null mice engrafted with human PBMC increased human B and, surprisingly, T cell engraftment [21*]. Generation of BLyS transgenic NOD-scid IL2rynull mice should enhance human B and T cell immune function in humanized mice and creation of these transgenic mice as well as other human-specific cytokine transgenic mice is underway [22].

Human Immune Function in Humanized Mice

Humanized mice have been used in the past year to investigate multiple types of human immune responses and to test potential therapeutics that modulate human immunity. The Hu-PBL-SCID model has an ~30 day window of analysis due to development of xenogeneic graft-versus-host disease [GVHD, 23]. Investigators have used this observation to establish an in vivo model of human immune-mediated GVHD. Using the Hu-PBL-SCID model based on NOD-scid mice, activation of human regulatory T cells by HIV-1 envelope protein gp120 delayed development of GVHD [24]. A similar delay of GVHD was observed in the Hu-PBL-SCID model based on NOD-scid IL2rynull mice following treatment with soluble Fas ligand [25]. King et al examined kinetics of engraftment and development of GVHD in the Hu-PBL-SCID model based on NOD-scid IL2rynull mice [26*]. They observed that most of the GVHD was directed against mouse MHC class I and II and mice deficient in MHC class I exhibited delayed GVHD. TNF inhibitors are used in the clinic to treat GVHD [27;28], and similarly, etanercept, a TNF inhibitor delayed the development of GVHD in this model system [26*].

Additional analyses of human T and B cells in Hu-SRC-SCID mice generated following engraftment of adult NOD-scid IL2rynull mice revealed that although human B cells develop, they are developmentally blocked [29], likely due to the inability of mouse BLyS to signal human B cells [21*]. The authors further suggested that human T cells selected in the thymus on mouse MHC class II is at least partially responsible for decreased human T cell immune responses.

One approach to enhance human T cell selection in the mouse thymus is to provide a human thymus autologous with the human HSC. This model, termed SCID-hu (Table 1), has been used extensively in the study of infectious agents (see below), and was recently used to establish a porcine islet xenograft rejection model [30] and an approach for induction of xenograft tolerance [31*]. Human HSC model systems supporting rejection of human allografts have to date not been reported. However, SCID-beige immunodeficient mice engrafted with human HSC generate multiple hematopoietic cells but not T cells [16;32]. Macrophages infiltrated human skin allografts in these mice, but produced little injury. However, when combined with adoptive transfer of autologous T cells to activate the infiltrating macrophages, the macrophages produced intimal expansion and calcification, reminiscent of atherogenesis or end-stage renal disease [33;34]. Regarding T cell-mediated allograft rejection, Racki et al demonstrated using the Hu-PBL-SCID model based on NOD-scid IL2rynull mice that either purified human CD4 or CD8 T cells can mediate human skin allograft rejection [35].

Additional human immune responses have been described in humanized mice. A model for asthma was used to identify a role for DC-derived CCL17 and CCL22 in attraction of Th2 cells and induction of airway inflammation [36]. In a humanized mouse model of sepsis, human lymphocyte apoptosis and cytokine production recapitulated the findings in patients with septic shock [37]. As new models are generated, investigation of multiple aspects of both immune and autoimmune responses of human immune systems will be possible.

Human-Specific Infectious Disease in Humanized Mice

One of the most prevalent uses of humanized mice is the study of human-specific infectious agents, particularly HIV [38;39]. Using the SCID-hu system, robust virus-specific immune responses following HIV infection were observed [40*]. Despite these robust responses, HIV viral load remained high and correlated with increased PD1 expression on human T cells, an observation also made in humans [4143]. In a model based on NOD-scid Jak3null mice, a nucleoside reverse transcriptase inhibitor blocked HIV infection in Hu-PBL-SCID mice [44]. Using the Hu-SRC-SCID model based on BALB/c-Rag1null IL2rynull mice, in vivo RNAi gene therapy against HIV-1 was investigated [45]. Human HSC were transduced with a lentiviral vector expressing a shRNA against HIV-1 nef gene and engrafted into newborn BALB/c-Rag1null IL2rynull mice. Evidence was obtained that the mature human CD4 T cells recovered from the HSC-engrafted mice exhibited an inhibition of virus replication, confirming efficacy of the shRNA therapy.

In a key series of experiments using both the Hu-PBL-SCID and Hu-SRC-SCID models based on NOD-scid IL2rynull mice, Kumar et al validated a novel new drug for the prevention and treatment of HIV infection [46**]. They used a modified single chain antibody (scFv) to the human T cell marker CD7 to deliver siRNA in vivo against CCR5 and viral Vif and Tat genes to human CD4 T cells in humanized mice. They documented that HIV infection could be controlled in a prophylactic setting in both model systems when viral challenge was performed after initiation of siRNA treatment, as well as in a post-infection setting, where mice were engrafted with PBMC from an HIV-infected subject.

A number of reports have described Dengue virus humanized mouse models for which no animal model system previously existed. Using newborn HSC-engrafted NOD-scid IL2rynull mice infected with eight different viral strains representing the four genotypes of Dengue viruses, viremia, a thrombocytopenia, increase in body temperature and erythema were observed corresponding to clinical characteristics in Dengue-infected humans [47;48].

Another approach in addition to the SCID-hu model to enhance human T cell selection during development in the thymus is to transgenically express human HLA in the mouse recipient. In a report using Hu-SRC-SCID mice based on NOD-scid IL2rynull HLA-A2 transgenic mice engrafted with HLA-A2 HSC, Jaiswal et al documented the development of virus-specific HLA-A2-restricted human T cell responses to Dengue virus infection [49*]. This is one of the first two reports using HLA-transgenic mice to document a human T cell HLA-restricted immune response. The other report used Epstein Barr Virus (EBV) infection in a model also based on NOD-scid IL2rynull HLA-A2 transgenic mice engrafted with HLA-A2 HSC [50**]. HLA-A2-restricted cytotoxic and IFNy-producing human T cells against multiple EBV HLA-A2 epitopes were observed exhibiting similar patterns of reactivity to that detected in human EBV carriers. These two reports document that HLA expressed transgenically in mouse thymus can positively select developing T cells and lead to HLA-restricted immune responses in mice engrafted with human HSC. Development of additional HLA-transgenic immunodeficient mice is currently underway [2;39].

NOD-scid IL2rynull mice engrafted as newborns with HSC were used to document a novel approach for enhancing immune responses following immunization. Targeting EBV antigen to human DC in vivo stimulated human T cell responses to EBV and induced anti-EBV antibody responses [51]. Similarly, NOD-scid β2mnull mice engrafted with human HSC and autologous mature T cells and then infected with live attenuated trivalent influenza virus generated human T cell responses to influenza [52]. The authors proposed this as a model for investigating antigen-presenting cells in immune responses as the response was completely dependent on reconstitution of the human myeloid compartment.

Finally, the first humanized mouse model for the study of Plasmodium falciparum was recently reported [53*]. NOD-scid IL2rynull mice were injected repeatedly with human red blood cells, which could then support productive infection with P. falciparum. Therapeutic efficacy of a number of anti-malarial agents was tested, permitting determination of the protective ED90 of the drugs against infection.

Overall, infectious disease studies in humanized mice are providing important pre-clinical model systems for the investigation of the pathogenesis of human-specific infectious agents, evaluation of therapeutics, and platforms to understand mechanisms of human immune responses following vaccination.

Humanized Mice and Cancer

Immunodeficient IL2rynull mice permit engraftment with a number of primary human tumors [1]. Comparing growth of human melanoma lung metastases in NOD-scid, NOD-scid β2mnull, and NOD-scid IL2rynull mice [54] the absolute NK deficiency in NOD-scid IL2rynull mice appeared to be a major factor in its enhanced support of primary tumor growth [54]. The enhanced engraftment of NOD-scid IL2rynull mice was confirmed in a report showing that human acute leukemia cells generate a faster and more efficient disease as compared to that observed in NOD-scid and NOD-scid β2mnull mice [55]. Human immune responses via antibody-dependent cellular cytotoxicity against primary adult T cell leukemia/lymphomas, Hodgkin lymphoma, and cutaneous T cell lymphoma in NOD-scid IL2rynull mice could be potentiated by defucosylated anti-CCR4 antibody [56;57*], suggesting a novel approach to enhance anti-tumor immunity. Confirming the utility of humanized mice for studies of cancer, it was shown that primary lung tumors transplanted into NOD-scid IL2rynull mice recapitulated in vivo tumor characteristics, including maintenance of stroma and passenger leukocytes that could, when activated with IL12, become tumor effector CTLs [58].

Humanized Mouse Models for Human Cell Therapy

Cellular therapy for treatment of multiple human diseases, particularly diabetes, is a promising approach. Successful human islet transplantation [59], has expanded into a number of clinical trials (www.clinicaltrials.gov). Because sufficient islets are not available for the tremendous need, focus has turned to development of beta cells from stem cells (http://www.betacell.org/). However, animal models for testing the safety and efficacy of this and other forms of human cellular therapy, such as regulatory T cells [60] or embryonic stem cells [61], are needed and humanized mice are being developed to address these critical needs. For example, immunodeficient NOD-Rag1null Prf1null Ins2Akita mice that spontaneously develop a non-autoimmune hyperglycemia [6265] can be engrafted with human islets [66]. This model can be used to test the in vivo function of human beta stem cells. Backcrossing of the Ins2Akita mutation to the NOD-Rag1null IL2rγnull strain will permit human beta stem/progenitor cells to be transplanted into mice bearing a functional human immune system [17;67], similar to the situation that occurs during transplantation in the clinic.

Conclusion

Humanized mice as pre-clinical models for the in vivo study of human cells and tissues have been under development for over 30 years. With the recent generation of immunodeficient IL2rγnull mice, the ability of humanized mice to serve as preclinical models is becoming a reality, but not yet ideal. Additional modifications of the model systems and genetic manipulation of the host continue to improve the ability of humanized mice to more accurately recapitulate the in vivo function of human cells and tissues. Novel insights into human disease are now possible due to the availability of humanized mice wherein human cells and tissues can be studied in vivo without putting patients at risk.

Acknowledgements

This work was supported by National Institutes of Health (NIH) Grants AI46629, DK53006, HL077642, an institutional Diabetes Endocrinology Research Center (DERC) grant DK32520, a Cancer Center Core grant CA34196, the Beta Cell Biology Consortium, grants from the Juvenile Diabetes Foundation, International, the Brehm Foundation, and the Helmsley Foundation. The contents of this publication are solely the responsibility of the authors and do not necessarily represent the official views of the NIH.

Funding National Institutes of Health, the Juvenile Diabetes Foundation, International, The Brehm Foundation, and the Helmsley Foundation.

Footnotes

The authors have no conflict of interest to report.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

REFERENCES

  • 1.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]
  • 2.Manz MG, Di Santo JP. Renaissance for mouse models of human hematopoiesis and immunobiology. Nat.Immunol. 2009;10:1039–1042. doi: 10.1038/ni1009-1039. [DOI] [PubMed] [Google Scholar]
  • 3.Pearson T, Greiner DL, Shultz LD. Humanized SCID mouse models for biomedical research. Curr.Top.Microbiol.Immunol. 2008;324:25–51. doi: 10.1007/978-3-540-75647-7_2. [DOI] [PubMed] [Google Scholar]
  • 4.Flanagan SP. `Nude', a new hairless gene with pleiotropic effects in the mouse. Genet.Res. 1966;8:295–309. doi: 10.1017/s0016672300010168. [DOI] [PubMed] [Google Scholar]
  • 5.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]
  • 6.Mombaerts P, Iacomini J, Johnson RS, Herrup K, Tonegawa S. 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]
  • 7.Shinkai Y, Rathbun G, Lam KP, Oltz EM, Stewart V, Mendelsohn M, Datta M, Young F, Stall AM, 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]
  • 8.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]
  • 9.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]
  • 10.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]
  • 11.Kong YC, Flynn JC, Banga JP, David CS. Application of HLA class II transgenic mice to study autoimmune regulation. Thyroid. 2007;17:995–1003. doi: 10.1089/thy.2007.0196. [DOI] [PubMed] [Google Scholar]
  • 12.Pascolo S. HLA class I transgenic mice: development, utilisation and improvement. Expert.Opin.Biol.Ther. 2005;5:919–938. doi: 10.1517/14712598.5.7.919. [DOI] [PubMed] [Google Scholar]
  • 13.Friese MA, Jensen LT, Willcox N, Fugger L. Humanized mouse models for organ-specific autoimmune diseases. Curr.Opin.Immunol. 2006;18:704–709. doi: 10.1016/j.coi.2006.09.003. [DOI] [PubMed] [Google Scholar]
  • 14.Pearson T, Greiner DL, Shultz LD. Creation of “humanized” mice to study human immunity. Curr.Protoc.Immunol. 2008 doi: 10.1002/0471142735.im1521s81. Chapter 15:Unit. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Legrand N, Weijer K, Spits H. Experimental model for the study of the human immune system: production and monitoring of “human immune system” Rag2−/− gamma c−/− mice. Methods Mol.Biol. 2008;415:65–82. doi: 10.1007/978-1-59745-570-1_4. [DOI] [PubMed] [Google Scholar]
  • 16.Lepus CM, Gibson TF, Gerber SA, Kawikova I, Szczepanik M, Hossain J, Ablamunits V, Kirkiles-Smith N, Herold KC, Donis RO, Bothwell AL, Pober JS, Harding MJ. Comparison of human fetal liver, umbilical cord blood, and adult blood hematopoietic stem cell engraftment in NOD-scid/gammac−/−, Balb/c-Rag1−/− gammac−/−, 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]
  • 17.Pearson T, Shultz LD, Miller D, King M, Laning J, Fodor W, Cuthbert A, Burzenski L, Gott B, Lyons B, Foreman O, Rossini AA, Greiner DL. Non-obese diabetic-recombination activating gene-1 (NOD-Rag1 null) interleukin (IL)-2 receptor common gamma chain (IL2r gamma null) 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]
  • *18.Huntington ND, Legrand N, Alves NL, Jaron B, Weijer K, Plet A, Corcuff E, Mortier E, Jacques Y, Spits H, Di Santo JP. IL-15 trans-presentation promotes human NK cell development and differentiation in vivo. J.Exp.Med. 2008 doi: 10.1084/jem.20082013. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study demonstrates that human NK cell development and function in Hu-SRC-SCID mice are augmented by treatment with human IL-15 and that this effect requires trans-presentation of IL-15.
  • 19.Woodland RT, Schmidt MR, Thompson CB. BLyS and B cell homeostasis. Semin.Immunol. 2006;18:318–326. doi: 10.1016/j.smim.2006.06.001. [DOI] [PubMed] [Google Scholar]
  • 20.Woodland RT, Schmidt MR. Homeostatic proliferation of B cells. Semin.Immunol. 2005;17:209–217. doi: 10.1016/j.smim.2005.02.006. [DOI] [PubMed] [Google Scholar]
  • *21.Schmidt MR, Appel MC, Giassi LJ, Greiner DL, Shultz LD, Woodland RT. Human BLyS facilitates engraftment of human PBL derived B cells in immunodeficient mice. PLoS.ONE. 2008;3:e3192. doi: 10.1371/journal.pone.0003192. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study demonstrates that treatment with recombinant human BLyS/BAFF enhances survival and function of human B cells in the Hu-PBL-model.
  • 22.Manz MG. Human-hemato-lymphoid-system mice: opportunities and challenges. Immunity. 2007;26:537–541. doi: 10.1016/j.immuni.2007.05.001. [DOI] [PubMed] [Google Scholar]
  • 23.King M, Pearson T, Shultz LD, Leif J, Bottino R, Trucco M, Atkinson MA, Wasserfall C, Herold KC, Woodland RT, Schmidt MR, Woda BA, thompson mj, Rossini AA, Greiner DL. 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]
  • 24.Becker C, Taube C, Bopp T, Becker C, Michel K, Kubach J, Reuter S, Dehzad N, Neurath MF, Reifenberg K, Schneider FJ, Schmitt E, Jonuleit H. Protection from graft-versus-host disease by HIV-1 envelope protein gp120-mediated activation of human CD4+CD25+ regulatory T cells. Blood. 2009;114:1263–1269. doi: 10.1182/blood-2009-02-206730. [DOI] [PubMed] [Google Scholar]
  • 25.Bohana-Kashtan O, Morisot S, Hildreth R, Brayton C, Levitsky HI, Civin CI. Selective reduction of graft-versus-host disease-mediating human T cells by ex vivo treatment with soluble Fas ligand. J.Immunol. 2009;183:696–705. doi: 10.4049/jimmunol.0800561. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • *26.King MA, Covassin L, Brehm MA, Racki W, Pearson T, Leif J, Laning J, Fodor W, Foreman O, Burzenski L, Chase T, Gott B, Rossini AA, Bortell R, Shultz LD, Greiner DL. Hu-PBL-NOD-scid IL2rgnull mouse model of xenogeneic graft-versus-host-like disease and the role of host MHC. Clinical & Experimental Immunology. 2009;157:104–118. doi: 10.1111/j.1365-2249.2009.03933.x. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study demonstrates that xenogeneic-GVHD in the Hu-PBL-SCID model is directed against murine MHC.
  • 27.Uberti JP, Ayash L, Ratanatharathorn V, Silver S, Reynolds C, Becker M, Reddy P, Cooke KR, Yanik G, Whitfield J, Jones D, Hutchinson R, Braun T, Ferrara JL, Levine JE. Pilot trial on the use of etanercept and methylprednisolone as primary treatment for acute graft-versus-host disease. Biol Blood Marrow Transplant. 2005;11:680–687. doi: 10.1016/j.bbmt.2005.05.009. [DOI] [PubMed] [Google Scholar]
  • 28.Hattori K, Hirano T, Miyajima H, Yamakawa N, Tateno M, Oshimi K, Kayagaki N, Yagita H, Okumura K. Differential effects of anti-Fas ligand and anti-tumor necrosis factor alpha antibodies on acute graft-versus-host disease pathologies. Blood. 1998;91:4051–4055. [PubMed] [Google Scholar]
  • 29.Watanabe Y, Takahashi T, Okajima A, Shiokawa M, Ishii N, Katano I, Ito R, Ito M, Minegishi M, Minegishi N, Tsuchiya S, Sugamura K. The analysis of the functions of human B and T cells in humanized NOD/shi-scid/gammac(null) (NOG) mice (hu-HSC NOG mice) Int.Immunol. 2009;21:843–858. doi: 10.1093/intimm/dxp050. [DOI] [PubMed] [Google Scholar]
  • 30.Tonomura N, Shimizu A, Wang S, Yamada K, Tchipashvili V, Weir GC, Yang YG. Pig islet xenograft rejection in a mouse model with an established human immune system. Xenotransplantation. 2008;15:129–135. doi: 10.1111/j.1399-3089.2008.00450.x. [DOI] [PubMed] [Google Scholar]
  • *31.Habiro K, Sykes M, Yang YG. Induction of human T-cell tolerance to pig Xenoantigens via thymus transplantation in mice with an established human immune system. Am.J.Transplant. 2009;9:1324–1329. doi: 10.1111/j.1600-6143.2009.02646.x. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study demonstrates that porcine thymic grafts tolerize developing human T cells to porcine antigens in the SCID-Hu model.
  • 32.Kirkiles-Smith NC, Harding MJ, Shepherd BR, Fader SA, Yi T, Wang Y, McNiff JM, Snyder EL, Lorber MI, Tellides G, Pober JS. 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]
  • 33.Goodman WG, London G, Amann K, Block GA, Giachelli C, Hruska KA, Ketteler M, Levin A, Massy Z, McCarron DA, Raggi P, Shanahan CM, Yorioka N. Vascular calcification in chronic kidney disease. Am.J.Kidney Dis. 2004;43:572–579. doi: 10.1053/j.ajkd.2003.12.005. [DOI] [PubMed] [Google Scholar]
  • 34.Doherty TM, Asotra K, Fitzpatrick LA, Qiao JH, Wilkin DJ, Detrano RC, Dunstan CR, Shah PK, Rajavashisth TB. Calcification in atherosclerosis: bone biology and chronic inflammation at the arterial crossroads. Proc.Natl.Acad.Sci.U.S.A. 2003;100:11201–11206. doi: 10.1073/pnas.1932554100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Racki WJ, Covassin L, Brehm M, Pino S, Ignotz R, Dunn R, Laning J, Rossini AA, Shultz LD, Greiner DL. NOD-scid IL2rgnull (NSG) mouse model of human skin transplantation and allograft rejection. Transplantation. 2009 doi: 10.1097/TP.0b013e3181c90242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Perros F, Hoogsteden HC, Coyle AJ, Lambrecht BN, Hammad H. Blockade of CCR4 in a humanized model of asthma reveals a critical role for DC-derived CCL17 and CCL22 in attracting Th2 cells and inducing airway inflammation. Allergy. 2009;64:995–1002. doi: 10.1111/j.1398-9995.2009.02095.x. [DOI] [PubMed] [Google Scholar]
  • 37.Unsinger J, McDonough JS, Shultz LD, Ferguson TA, Hotchkiss RS. Sepsis-induced human lymphocyte apoptosis and cytokine production in “humanized” mice. J.Leukoc.Biol. 2009;86:219–227. doi: 10.1189/jlb.1008615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Denton PW, Garcia JV. Novel humanized murine models for HIV research. Curr.HIV./AIDS Rep. 2009;6:13–19. doi: 10.1007/s11904-009-0003-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Legrand N, Ploss A, Balling R, Becker PD, Borsotti C, Brezillon N, Debarry J, de JY, Deng H, Di Santo JP, Eisenbarth S, Eynon E, Flavell RA, Guzman CA, Huntington ND, Kremsdorf D, Manns MP, Manz MG, Mention JJ, Ott M, Rathinam C, Rice CM, Rongvaux A, Stevens S, Spits H, Strick-Marchand H, Takizawa H, van Lent AU, Wang C, Weijer K, Willinger T, Ziegler P. 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]
  • *40.Brainard DM, Seung E, Frahm N, Cariappa A, Bailey CC, Hart WK, Shin HS, Brooks SF, Knight HL, Eichbaum Q, Yang YG, Sykes M, Walker BD, Freeman GJ, Pillai S, Westmoreland SV, Brander C, Luster AD, Tager AM. Induction of robust cellular and humoral virus-specific adaptive immune responses in human immunodeficiency virus-infected humanized BLT mice. J.Virol. 2009;83:7305–7321. doi: 10.1128/JVI.02207-08. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study shows that infection of SCID-Hu mice with HIV generates both a humoral and cellular anti-viral responses, including an epitope-specific CD8 T cell response.
  • 41.Trautmann L, Janbazian L, Chomont N, Said EA, Gimmig S, Bessette B, Boulassel MR, Delwart E, Sepulveda H, Balderas RS, Routy JP, Haddad EK, Sekaly RP. Upregulation of PD-1 expression on HIV-specific CD8+ T cells leads to reversible immune dysfunction. Nat.Med. 2006;12:1198–1202. doi: 10.1038/nm1482. [DOI] [PubMed] [Google Scholar]
  • 42.Petrovas C, Casazza JP, Brenchley JM, Price DA, Gostick E, Adams WC, Precopio ML, Schacker T, Roederer M, Douek DC, Koup RA. PD-1 is a regulator of virus-specific CD8+ T cell survival in HIV infection. J.Exp.Med. 2006;203:2281–2292. doi: 10.1084/jem.20061496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Day CL, Kaufmann DE, Kiepiela P, Brown JA, Moodley ES, Reddy S, Mackey EW, Miller JD, Leslie AJ, DePierres C, Mncube Z, Duraiswamy J, Zhu B, Eichbaum Q, Altfeld M, Wherry EJ, Coovadia HM, Goulder PJ, Klenerman P, Ahmed R, Freeman GJ, Walker BD. PD-1 expression on HIV-specific T cells is associated with T-cell exhaustion and disease progression. Nature. 2006;443:350–354. doi: 10.1038/nature05115. [DOI] [PubMed] [Google Scholar]
  • 44.Hattori S, Ide K, Nakata H, Harada H, Suzu S, Ashida N, Kohgo S, Hayakawa H, Mitsuya H, Okada S. Potent activity of a nucleoside reverse transcriptase inhibitor, 4'-ethynyl-2-fluoro-2'-deoxyadenosine, against human immunodeficiency virus type 1 infection in a model using human peripheral blood mononuclear cell-transplanted NOD/SCID Janus kinase 3 knockout mice. Antimicrob.Agents Chemother. 2009;53:3887–3893. doi: 10.1128/AAC.00270-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.ter BO, Legrand N, von Eije KJ, Centlivre M, Spits H, Weijer K, Blom B, Berkhout B. Evaluation of safety and efficacy of RNAi against HIV-1 in the human immune system (Rag-2(−/−)gammac(−/−)) mouse model. Gene Ther. 2009;16:148–153. doi: 10.1038/gt.2008.124. [DOI] [PubMed] [Google Scholar]
  • **46.Kumar P, Ban HS, Kim SS, Wu H, Pearson T, Greiner DL, Laouar A, Yao J, Haridas V, Habiro K, Yang YG, Jeong JH, Lee KY, Kim YH, Kim SW, Peipp M, Fey GH, Manjunath N, Shultz LD, Lee SK, Shankar P. 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]; This is the first demonstration that RNAi therapy can be used successfully in both the Hu-PBL-SCID and the Hu-SRC-SCID models. A CD7-specific single chain antibody was used to deliver siRNA targeting CCR5 and viral proteins to T cells and this blocked HIV infection in vivo.
  • 47.Gubler DJ. Dengue and dengue hemorrhagic fever. Clin.Microbiol.Rev. 1998;11:480–496. doi: 10.1128/cmr.11.3.480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Gubler DJ, Clark GG. Dengue/dengue hemorrhagic fever: the emergence of a global health problem. Emerg.Infect.Dis. 1995;1:55–57. doi: 10.3201/eid0102.952004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • *49.Jaiswal S, Pearson T, Friberg H, Shultz LD, Greiner DL, Rothman AL, Mathew A. Dengue virus infection and virus-specific HLA-A2 restricted immune responses in humanized NOD-scid IL2rgammanull mice. PLoS.ONE. 2009;4:e7251. doi: 10.1371/journal.pone.0007251. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study showing HLA-A2-restricted, epitope-specific CD8 T cell responses after Dengue virus infection in Hu-SRC-SCID mice.
  • **50.Strowig T, Gurer C, Ploss A, Liu YF, Arrey F, Sashihara J, Koo G, Rice CM, Young JW, Chadburn A, Cohen JI, Munz C. Priming of protective T cell responses against virus-induced tumors in mice with human immune system components. J.Exp.Med. 2009;206:1423–1434. doi: 10.1084/jem.20081720. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study demonstrates that T cells developing in Hu-SRC-SCID mice control the in vivo growth of EBV-associated tumors after infection with EBV. It also shows the generation of HLA-A2-restricted, EBV-epitope-specific CD8 T cell responses after infection.
  • 51.Gurer C, Strowig T, Brilot F, Pack M, Trumpfheller C, Arrey F, Park CG, Steinman RM, Munz C. Targeting the nuclear antigen 1 of Epstein-Barr virus to the human endocytic receptor DEC-205 stimulates protective T-cell responses. Blood. 2008;112:1231–1239. doi: 10.1182/blood-2008-03-148072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Yu CI, Gallegos M, Marches F, Zurawski G, Ramilo O, Garcia-Sastre A, Banchereau J, Palucka AK. 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]
  • *53.Jimenez-Diaz MB, Mulet T, Viera S, Gomez V, Garuti H, Ibanez J, varez-Doval A, Shultz LD, Martinez A, Gargallo-Viola D, Angulo-Barturen I. Improved murine model of malaria using Plasmodium falciparum competent strains and non-myelodepleted NOD-scid IL2Rgammanull mice engrafted with human erythrocytes. Antimicrob.Agents Chemother. 2009;53:4533–4536. doi: 10.1128/AAC.00519-09. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study shows that NOD-scid IL2rynull mice are the optimal mouse model to study infection with Plasmodium falciparum.
  • 54.Carreno BM, Garbow JR, Kolar GR, Jackson EN, Engelbach JA, Becker-Hapak M, Carayannopoulos LN, Piwnica-Worms D, Linette GP. Immunodeficient mouse strains display marked variability in growth of human melanoma lung metastases. Clin.Cancer Res. 2009;15:3277–3286. doi: 10.1158/1078-0432.CCR-08-2502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Agliano A, Martin-Padura I, Mancuso P, Marighetti P, Rabascio C, Pruneri G, Shultz LD, Bertolini F. Human acute leukemia cells injected in NOD/LtSz-scid/IL-2Rgamma null mice generate a faster and more efficient disease compared to other NOD/scid-related strains. Int.J.Cancer. 2008;123:2222–2227. doi: 10.1002/ijc.23772. [DOI] [PubMed] [Google Scholar]
  • 56.Ito A, Ishida T, Yano H, Inagaki A, Suzuki S, Sato F, Takino H, Mori F, Ri M, Kusumoto S, Komatsu H, Iida S, Inagaki H, Ueda R. Defucosylated anti-CCR4 monoclonal antibody exercises potent ADCC-mediated antitumor effect in the novel tumor-bearing humanized NOD/Shi-scid, IL-2Rgamma(null) mouse model. Cancer Immunol.Immunother. 2009;58:1195–1206. doi: 10.1007/s00262-008-0632-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • *57.Ito A, Ishida T, Utsunomiya A, Sato F, Mori F, Yano H, Inagaki A, Suzuki S, Takino H, Ri M, Kusumoto S, Komatsu H, Iida S, Inagaki H, Ueda R. Defucosylated anti-CCR4 monoclonal antibody exerts potent ADCC against primary ATLL Cells mediated by autologous human immune cells in NOD/Shi-scid, IL-2Rgamma(null) mice in vivo. J.Immunol. 2009;183:4782–4791. doi: 10.4049/jimmunol.0900699. [DOI] [PubMed] [Google Scholar]; This study demonstrates that antibody-based immunotherapies can be tested in humanized mice.
  • 58.Simpson-Abelson MR, Sonnenberg GF, Takita H, Yokota SJ, Conway TF, Jr., Kelleher RJ, Jr., Shultz LD, Barcos M, Bankert RB. Long-term engraftment and expansion of tumor-derived memory T cells following the implantation of non-disrupted pieces of human lung tumor into NOD-scid IL2Rgamma(null) mice. J.Immunol. 2008;180:7009–7018. doi: 10.4049/jimmunol.180.10.7009. [DOI] [PubMed] [Google Scholar]
  • 59.Shapiro AM, Ricordi C, Hering BJ, Auchincloss H, Lindblad R, Robertson RP, Secchi A, Brendel MD, Berney T, Brennan DC, Cagliero E, Alejandro R, Ryan EA, DiMercurio B, Morel P, Polonsky KS, Reems JA, Bretzel RG, Bertuzzi F, Froud T, Kandaswamy R, Sutherland DE, Eisenbarth G, Segal M, Preiksaitis J, Korbutt GS, Barton FB, Viviano L, Seyfert-Margolis V, Bluestone J, Lakey JR. International trial of the Edmonton protocol for islet transplantation. N.Engl.J.Med. 2006;355:1318–1330. doi: 10.1056/NEJMoa061267. [DOI] [PubMed] [Google Scholar]
  • 60.Golovina TN, Mikheeva T, Suhoski MM, Aqui NA, Tai VC, Shan X, Liu R, Balcarcel RR, Fisher N, Levine BL, Carroll RG, Warner N, Blazar BR, June CH, Riley JL. CD28 costimulation is essential for human T regulatory expansion and function. J.Immunol. 2008;181:2855–2868. doi: 10.4049/jimmunol.181.4.2855. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Tian X, Hexum MK, Penchev VR, Taylor RJ, Shultz LD, Kaufman DS. Bioluminescent Imaging Demonstrates Transplanted Human Embryonic Stem Cell-derived Cd34(+) Cells Preferentially Develop into Endothelial Cells. Stem Cells. 2009 doi: 10.1002/stem.204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.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]
  • 63.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]
  • 64.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]
  • 65.Izumi T, Yokota-Hashimoto H, Zhao S, Wang J, Halban PA, Takeuchi T. Dominant negative pathogenesis by mutant proinsulin in the Akita diabetic mouse. Diabetes. 2003;52:409–416. doi: 10.2337/diabetes.52.2.409. [DOI] [PubMed] [Google Scholar]
  • 66.Pearson T, Shultz LD, Lief J, Burzenski L, Gott B, Chase T, Foreman O, Rossini AA, Bottino R, Trucco M, Greiner DL. A new immunodeficient hyperglycaemic mouse model based on the Ins2Akita mutation for analyses of human islet and beta stem and progenitor cell function. Diabetologia. 2008;51:1449–1456. doi: 10.1007/s00125-008-1057-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Shultz LD, Lyons BL, Burzenski LM, Gott B, Chen X, Chaleff S, Gillies SD, King M, Mangada J, Greiner DL, Handgretinger R. Human lymphoid and myeloid cell development in NOD/LtSz-scid IL2rγnull mice engrafted with mobilized human hematopoietic stem cell. J.Immunol. 2005;174:6477–6489. doi: 10.4049/jimmunol.174.10.6477. [DOI] [PubMed] [Google Scholar]

RESOURCES