Skip to main content
Wiley Open Access Collection logoLink to Wiley Open Access Collection
. 2025 Oct 13;5(10):e70224. doi: 10.1002/cpz1.70224

Humanized Mouse Models for Type 1 Diabetes

David V Serreze 1,, Marissa Tousey‐Pfarrer 1, Jeremy J Racine 1
PMCID: PMC12517312  PMID: 41081710

Abstract

T cell–mediated autoimmune type 1 diabetes (T1D) is under complex polygenic control in both humans and the NOD mouse model. However, in both species, particular major histocompatibility complex (MHC; designated HLA in humans) haplotypes provide the primary T1D risk factor. Both MHC/HLA class I and II variants interactively contribute to T1D by respectively driving autoreactive CD8 and CD4 T cell responses that cooperatively destroy insulin‐producing pancreatic β cells. While NOD mice have provided important insights to the pathogenic basis of T1D, the model has so far provided only a limited means to identify possible clinically translatable disease intervention approaches. This highlights a need to humanize NOD mice in ways that their pathogenic basis of T1D development becomes more similar to that characterizing the disease course in patients. In this review, we discuss the use of CRISPR/Cas9‐generated murine‐MHC‐deficient NOD mice as a platform for introduction of patient‐relevant HLA and T cell receptor molecules. These mice provide ever‐improving models for development of clinically applicable interventions for T1D and other autoimmune diseases. © 2025 The Author(s) Current Protocols published by Wiley Periodicals LLC.

Keywords: autoimmune T1D, humanized mice, MHC/HLA, TCRs

Introduction

Most cases of type 1 diabetes (T1D) in humans result from T cell–mediated autoimmune destruction of insulin‐producing pancreatic β cells. There are multiple models for studying T1D in mice (Leiter & Schile, 2013). One such model, the NOD mouse (Leiter, 2001), develops T cell–mediated T1D and has provided important insights into the pathogenic basis of this disease (Leiter, 2001; Serreze et al., 2024). However, NOD mice have provided only a limited means to identify possible clinically translatable T1D intervention approaches (Atkinson & Leiter, 1999). One way to better design mouse studies so that pathogenic mechanisms of disease development more closely resemble that in patients is through humanization.

In both humans and NOD mice, T1D results from complex interactions between multiple susceptibility genes (designated IDDM and Idd in humans and mice, respectively; Chen et al., 2018; Grant et al., 2020). While polygenic in nature, particular major histocompatibility (MHC) and human leukocyte antigen (HLA) haplotypes provide the strongest drivers of T1D susceptibility in both species. In the H2g7 MHC haplotype of NOD mice, homozygous expression of H2‐Ag7 and the absence of H2‐E class II gene products are key components of T1D development by driving pathogenic autoreactive CD4 T cell responses. Certain HLA class II molecules, including DR4 and DQ8, are also strong drivers of diabetogenic CD4 T cell responses in patients (Grant et al., 2020). In addition, it is now appreciated that, while they represent common variants also found in some non‐autoimmune‐prone strains, the Kd and Db MHC class I molecules encoded in the H2g7 haplotype additionally exert essential diabetogenic functions in NOD mice by driving pathogenic CD8 T cell responses (Serreze et al., 2024). It is also now known that some HLA class I molecules, including the relatively common A2 and the rarer B39 variants, drive diabetogenic CD8 T cell responses in humans (Howson et al., 2009). Thus, we reasoned that it could be of clinical translational value to generate NOD mouse stocks in which murine diabetogenic MHC class I and/or II molecules were replaced by T1D relevant human variants. The advent of targeted CRISPR/Cas9 protocols (Harms et al., 2014; Qin et al., 2015, 2016) made development of such NOD stocks possible and is the subject of this review. Other targeting protocols are also available (Gaj et al., 2013; Wefers et al., 2012).

NOD Mice Expressing T1D‐patient‐relevant HLA Class I Variants

The necessary role of MHC class I molecules for T1D development in NOD mice by driving pathogenic CD8 T cells was first demonstrated by showing that introduction of an inactivated variant of the β2m gene (β2mnull ) required for MHC class I expression resulted in complete disease resistance (Serreze et al., 1994; Wicker et al., 1994). Subsequent efforts were made to introduce human HLA class I alleles into NOD background mice. An early approach crossed a full‐length genomic construct encoding the HLA‐A*02:01 allele originally introduced into the C57BL/6 (B6) strain (Le et al., 1989) into NOD mice (Marron et al., 2002). There were three issues with this approach: (1) crossing transgenes from one strain to another also introduces linked genetic material from the donor strain; (2) the genomic construct may not be optimized for interaction between human and murine molecules; and (3) protocols for generation of random transgene insertion (Conner, 2004) can effectively knock out a recipient strain gene. This particular allele is now designated Mcph1Tg(HLA‐A2.1)1Enge (MGI:3056375) since it integrated in, and functionally knocked out, Mcph1.

A chimeric “HHD” transgene was developed that bypasses some of concern 2 above. This construct encodes a monochain fusing human β2m to the α1 and α2 antigen‐binding domains of the HLA‐A*02:01 allele, which in turn were fused to the α3, transmembrane, and cytoplasmic domains of the mouse H2‐Db variant (Pascolo et al., 1997). The original HLA‐A2.1 genomic construct was crossed into NOD mice still expressing murine MHC I molecules (Marron et al., 2002). The HHD transgene, however, was directly introduced into NOD zygotes and then paired with the β2mnull mutation (Takaki et al., 2006). These mice were important in identifying HLA‐A2‐restricted epitopes of insulin and islet‐specific glucose‐6‐phosphatase catalytic subunit‐related protein (originally designated IGRP, now G6pc2) targeted by CD8 T cells contributing to T1D pathogenesis (Jarchum et al., 2007; Jarchum & DiLorenzo, 2010; Takaki et al., 2006) as well as identifying means of potentially attenuating such responses (Niens et al., 2011). A modified HHD transgenic vector has now been used to introduce the HLA‐A*39:06 class I allele into NOD mice (Schloss et al., 2018).

Introduction of the β2mnull mutation unfortunately also eliminates expression of FcRn, a non‐classical MHC class I molecule critical for serum homeostasis of IgG and albumin (Roopenian & Akilesh, 2007). Hence, β2m‐deficient NOD mice cannot be used to evaluate possible antibody‐ or serum‐albumin‐based T1D interventions (Larsen et al., 2016). To overcome this limitation, we utilized the CRISPR/Cas9 workflow employed by the Jackson Laboratory Genetic Engineering Technologies group (Qin et al., 2016) to directly generate NOD stocks in which the classical H2‐Kd and H2‐Db class I genes were ablated individually or in tandem (NOD‐H2‐K–/–, NOD‐H2‐D–/–, and NOD‐cMHCI–/–, respectively; Racine et al., 2018). Here, the d and b designations refer to the specific alleles of the H2‐K1 and H2‐D1 genes found in NOD mice. The design of CRISPR/Cas9 guides targeting genes in the MHC region must take into account the high degree of allelic variation that occurs between different mouse strains at this locus. A listing of haplotypes for various mouse strains can be found in Appendix F of Flurkey et al. (2009).

Individual deletion of the H2‐Kd or H2‐Db genes significantly reduced, but did not completely eliminate, T1D development in NOD mice (Racine et al., 2018). Similar to the result obtained by introduction of the β2mnull mutation, NOD‐cMHCI–/– mice were completely T1D resistant (Racine et al., 2018). The availability of NOD‐cMHCI–/– mice gave us the ability to functionally test whether particular HLA class I variants could contribute to T1D development in patients, a possibility that was only supported epidemiologically at the time (Fennessy et al., 1994; Nejentsev et al., 2007; Noble et al., 2010). To do this, we again turned to HHD‐based transgene constructs (Pascolo et al., 1997) to induce expression of the proposed T1D‐patient‐relevant A2 or B39 HLA class I variants in NOD‐cMHCI–/– mice. We introduced the original HHD transgene driving expression of the HLA‐A2 variant or the modified version encoding the HLA‐B39 antigen presenting domains (Schloss et al., 2018) into NOD‐cMHCI–/– mice (Racine et al., 2018). Expression of either transgene‐driven HLA‐A2 or B39 HLA class I molecules restored aggressive T1D development to NOD‐cMHCI–/– mice, which as described above are normally completely disease resistant (Racine et al., 2018; Fig. 1). These findings provide functional evidence the HLA‐A2 or B39 class I variants can indeed contribute to T1D in patients. FcRn activity also remained intact in CRISPR‐generated NOD‐cMHCI–/– mice, as evidenced by their retention of transfused antibodies for at least 30 days (Racine et al., 2018). Thus, NOD mice with direct CRISPR‐mediated ablation of murine MHC molecules could be used to test possible antibody‐mediated T1D interventions (Racine et al., 2018). Additionally, unlike β2mnull HLA‐A humanized mice, NOD‐cMHCI–/– versions also retained expression of non‐classical MHC I molecules including Cd1d and Qa‐2 (Racine et al., 2018). A comprehensive listing of potential non‐classical MHC class I molecules that could be impacted by using humanized mice with a β2mnull mutation can be found in Forman & Lindahl (2002).

Figure 1.

Figure 1

Introduction of HHD expression vector–driven human HLA‐A2 or B39 class I variants restores T1D susceptibility to NOD mice made deficient in classical murine MHC molecules using a CRISPR approach.

NOD Mice with CRISPR Ablation of Both Classical Murine MHC Class I and II Molecules

While it is now appreciated that MHC/HLA class I–restricted CD8 T cells contribute to T1D in both NOD mice and humans, particular class II variants including DR4 were originally identified as strong disease‐driving components in patients (Grant et al., 2020). This led to an effort to determine if B6 mice transgenically expressing DR4 in the absence of endogenous murine class II molecules (B6‐DR4) could provide a clinically relevant T1D model (Verhagen et al., 2018). Despite also expressing the T cell co‐stimulatory B7 molecule on pancreatic β cells, such B6‐DR4 mice remained resistant to spontaneous T1D. However, priming these B6‐DR4 mice with insulin peptides did induce T1D. NOD background mice transgenically expressing DR4 were also found to be resistant to spontaneous T1D (Pow Sang et al., 2015).

These findings illustrate a need to test another approach that might generate NOD mice that develop T1D through autoreactive T cell responses at least partly mediated by HLA class II variants. As noted earlier, the H2g7 MHC haplotype of NOD encodes a single class II molecule designated H2‐Ag7. Thus, we utilized CRISPR to ablate the Abg7 subunit gene in NOD‐cMHCI–/– mice (Racine et al., 2018). This resulted in NOD mice lacking expression of both classical murine MHC class I and II molecules (designated NOD‐cMHCI/II–/– mice). These mice now provide a platform for transgenic introduction of any desired combination of HLA class I and/or II variants.

Functional T1D‐patient‐derived Insulin/Proinsulin TCR Molecules in NOD‐cMHCI/II–/–.DQ8 Mice

As described earlier, HLA‐DQ8 (encoded by the HLA‐DQA*0301 and DQB*0302 alleles) is a class II variant strongly associated with disease susceptibility in T1D patients. Thus, we produced NOD‐cMHCI/II–/– mice transgenically expressing HLA‐DQ8 (designated NOD‐cMHCI/II–/–.DQ8; Racine et al., 2023), and subsequently introduced into these mice transgenes derived from T1D patients encoding rearranged T cell receptor (TCR) α and β chains from DQ8‐restricted insulin/proinsulin‐autoreactive CD4 T cell clones (Racine et al., 2023). The transgenic 20D11 and 6H9 TCRs both recognize the insulin B‐chain 9‐23 peptide (Michels et al., 2017), whereas that from the A1.9 clone is specific for human C‐peptide 42‐50 (Pathiraja et al., 2015). TCRα sequences were packaged into pCD2 (Zhumabekov et al., 1995) and TCRβ sequences were incorporated into p428 (Sawada et al., 1994). These vectors drive TCR chain expression by CD2 and CD4, respectively. An additional genomic transgene designated (TgINS*)172 and encoding a human proinsulin allele with the class I (26‐63) ACAGGGGTGTGGGG variable number of tandem repeats (VNTR) sequence associated with T1D susceptibility (Rotwein et al., 1986) was also introduced into the NOD‐cMHCI/II–/–.DQ8 strain additionally carrying the A1.9 TCR.

None of the human‐TCR‐expressing NOD‐cMHCI/II–/–.DQ8 strains developed overt T1D. However, they all developed significant levels of pancreatic islet leukocytic infiltration (termed insulitis), which can be quantitatively scored as described in Leiter (2001) and Ratiu et al. (2017). As assessed by histological examination, individual islets were scored as follows: 0, no lesions; 1, peri‐insular aggregates; 2, <25% islet destruction; 3, >25% islet destruction; and 4, >75% islet destruction (for examples, see Fig. 2). A mean insulitis score (MIS) for each mouse was determined by dividing the summed score for the pancreas by the total number of islets examined. The development of significant levels of insulitis indicated the human insulin/proinsulin‐autoreactive TCRs introduced into NOD‐cMHCI/II–/–.DQ8 mice were functionally active. These T cell responses in NOD‐cMHCI/II–/–.DQ8 TCR transgenic stocks recapitulated those in DQ8‐expressing T1D patients and thus allow analyses for how such effectors undergo selection. The ability to quantitate insulitis levels in these stocks could also enable their use for evaluating the efficacy of potential clinically applicable T1D intervention approaches.

Figure 2.

Figure 2

Examples of NOD islets with insulitis scores of 0 to 4. Reproduced from Ratiu et al. (2017).

The above models used traditional transgene methodology, including allowing for random insertion. Briefly, TCR chain transgenes were removed from their respective plasmid backbones by restriction digestion (NotI/SalI and NotI, respectively, for pCD2 and p428), gel‐purified, and mixed together for microinjection into embryos (Racine et al., 2023). This process can be fraught with obstacles and multiple founders must be tested. In the course of our studies, we found several lines that either did not develop any insulitis (indicating potential issues with protein expression that we could not resolve through flow cytometry) or breeding issues (likely due to insertion into genes affecting viability). For our A1.9 model, our initial four founder lines were reduced to a single viable model due to three separate issues: lack of germline transmission, integration into the Y chromosome, and breeding issues.

One approach to overcome these issues is to couple CRISPR/Cas9 methodology (Harms et al., 2014; Qin et al., 2015, 2016) with targeting of a safe harbor locus such as Rosa26 for insertion (Chu et al., 2016). An alternative to dedicated human TCR transgenic mouse strains is the use of TCR retrogenic methodologies (Kong et al., 2019). In this application, transformation takes place in bone marrow cells that are then transplanted into recipient mice. While this requires that experimental mice be repeatedly developed on an as‐needed basis, it provides flexibility by eliminating the need for multiple mouse colonies each expressing a different human TCR.

Retrogenic technology has introduced a glutamic acid decarboxylase (GAD)–reactive TCR into NOD.HLA‐DR4 Tg.H2Ab1−/−.Rag1−/− mice (Jing et al., 2022). While NOD.HLA‐DR4 Tg.H2Ab1−/− mice develop insulitis, NOD.HLA‐DR4 Tg.H2Ab1−/−.Rag1−/− mice carrying retrogenically introduced GAD65115–127 reactive TCR required peptide priming to induce insulitis. We previously hypothesized that the discrepancy between our above DQ8 model and the DR4 model mentioned here may be due to differing abilities of murine CD4 to interact with HLA‐DQ8 versus HLA‐DR4 (Racine et al., 2023), but it could also be due to the intracellular signaling domains. The HHD vector for MHC I transgenes was designed to allow for (1) mouse CD8 to interact with a murine rather than human α3 domain (Pascolo et al., 1997) and (2) the transmembrane and intracellular domains to also be murine‐derived. Despite HLA‐DQ8 being structurally equivalent to H2‐Ag7, and the capacity of the H2‐Ag7‐restricted BDC2.5 clone to be selected by HLA‐DQ8 (Wen et al., 2002), and the ability of conserved epitopes of insulin, GAD, and HSP 60 to bind both H2‐Ag7 and HLA‐DQ8 (Yu et al., 2000), polyclonal T cells selected by H2‐Ag7 and HLA‐DQ8 seem to have different fates than those selected by H2‐Ag7 and HLA‐DR4. Moreover, pairing of a human CD4 transgene did not recover insulitis or T1D in another HLA‐DQ8 model co‐expressing human CD4 (Liu et al., 1999). It is therefore possible that future iterations of HLA‐humanized NOD mice will require the creation of chimeric constructs that allow optimal murine CD4/HLA class II interactions.

Additional Autoimmunity in NOD‐cMHCI/II–/–.DQ8 Mice

In addition to T1D, NOD mice can also develop other autoimmune pathologies (Leiter, 1998). It has been reported that NOD mice transgenically expressing the HLA‐DQ8 variant in conjunction with murine MHC class I molecules could develop lethal autoimmune myocarditis (Elliott et al., 2003; Taneja et al., 2007; Taylor et al., 2004). We have more recently found that subclinical levels of myocarditis develop in ∼90% of NOD‐cMHCI/II–/–.DQ8 mice (Racine et al., 2023, 2024). We also observed myocarditis development in NOD‐cMHCI/II–/–.DQ8‐A2 mice and in cMHCI/II–/–.DQ8‐6H9 mice (which lacked allelic exclusion), but not in cMHCI/II–/–.DQ8‐20D11 or cMHCI/II–/–.DQ8‐A1.9 mice (Racine et al., 2023). However, treatment of NOD‐cMHCI/II–/–.DQ8 mice with the anti‐PD‐1 immune checkpoint inhibitor (ICI) elicited virtually universal lethal myocarditis within a two‐week period (Racine et al., 2024). ICI‐treated NOD‐cMHCI/II–/–.DQ8 mice also subsequently developed severe myositis that was highly pronounced in the diaphragm and soleus. TCR sequencing indicated that, while limited, there was some overlap in the clonotypes infiltrating the heart and skeletal muscle of ICI‐treated NOD‐cMHCI/II–/–.DQ8 mice. Hence, there appears to be at least some extent of shared antigen recognition at both sites. We have also recently found that NOD‐cMHCI/II–/–.DQ8, but not NOD‐cMHCI/II–/–.DQ8‐20D11 mice, have increased immune cell infiltration into the sciatic nerve, including T cells (Fig. 3). Resistance in the latter strain indicates that neuritis does not develop if the TCR repertoire is skewed towards insulin. Thus, NOD‐cMHCI/II–/–.DQ8 mice may be of use to study autoimmune neuritis, another autoimmune disease that can occasionally develop in the NOD background (Leiter, 1998). Whether this phenotype is exacerbated by ICI therapy, or whether these mice also develop the adverse event optic neuritis (Francis et al., 2020; Kartal & Atas, 2018; Mori et al., 2018; Sengul Samanci et al., 2020) is currently not known.

Figure 3.

Figure 3

Neuritis develops in NOD‐cMHCI/II–/–.DQ8 mice but not NOD‐cMHCI/II–/–.DQ8‐20D11 mice.

Collectively, these studies indicate that DQ8 expression is sufficient to initiate myocarditis development in NOD background mice, but spontaneous progression of disease to a highly penetrant lethal state likely requires T cell responses mediated by additional MHC molecules. Interestingly, in a previous DQ8 model retaining murine MHC I molecules, CD8 T cells were required to initiate the disease (Hayward et al., 2006). Therefore, it is likely the addition of MHC class I mediated CD8 T cell responses is required for myocarditis to spontaneously progress to a high rate lethal state in NOD background mice. This conclusion is further supported by our finding that lethal myocarditis can develop with higher penetrance in the absence of ICI treatment in NOD‐cMHCI/II–/–.DQ8 mice co‐expressing the HLA‐A2 class I variant (Racine et al., 2024). The fact that any myocarditis develops in NOD‐cMHCI/II–/–.DQ8 mice (Racine et al., 2024) but not in HLA‐DQ8 NOD carrying the β2mnull mutation (Hayward et al., 2006) indicates that non‐classical MHC I–selected T cells may play a role in this pathology.

Immunodeficient NOD‐related Strains Expressing Various HLA Molecules Can Be Engrafted with Human Cells

It should be noted that the above models, although HLA and TCR humanized, still rely on components of the murine immune system. Use of immunodeficient mouse strains allows for the humanization of multiple immunological components through a number of different protocols (Chuprin et al., 2023; Fu & Kim, 2020; Pearson et al., 2008; Phelps et al., 2023; Verma & Wesa, 2020). There is a growing array of immunodeficient mouse strains that can be used as recipients for human cells and tissues (Chuprin et al., 2023). However, NOD background stocks made deficient in murine lymphocytes by introduction of the Prkdcscid or Rag1null mutations are most efficiently engrafted by human cells or tissues. A further modification that has been made to these stocks is introduction of an inactivated Il2rg (IL‐2 common‐γ chain receptor) gene. The inactivated Il2rg gene eliminates signaling through the IL‐2, IL‐4, IL‐7, IL‐9, IL‐15, and IL‐21 receptors, with the resultant strains carrying this mutation designated NSG and NRG (Shultz et al., 2012). The introduced mutations combined with other NOD strain characteristics, such as the lack of hemolytic complement activity and diminished myeloid cell phagocytic activity, eliminates NK cells in addition to lymphocytes, enabling NSG or NRG mice to be efficiently engrafted with human cells.

NSG and NRG mice have now been further modified by introducing transgenes encoding the HLA class I or II variants listed in Table 1. Additional HLA variant alleles existing within the NOD‐background could also be introduced into NSG and NRG mice (Table 2). This allows NSG and NRG stocks to be used for functional testing of engrafted T cells from human donors sharing the indicated HLA variant. One example is a human CD8 T cell line recognizing the β cell autoantigen IGRP (now designated G6pc2) in an HLA‐A2‐restricted fashion, which was able to transfer insulitis to NSG recipients expressing this human class I variant (Unger et al., 2012). This indicated that human diabetogenic T cells can be functionally activated in NSG recipients expressing appropriate HLA restriction elements. Efforts are also underway to express an array of HLA variants in a CRISPR/Cas9‐generated NSG‐cMHCI/II–/– stock, the first of which was HLA‐DQ8 (Racine et al., 2023). We have used these mice as recipients of murine heart and skeletal muscle infiltrating T cells to show cross reactivity of autoreactive T cells targeting these tissues after anti‐PD‐1 administration (Racine et al., 2024).

Table 1.

HLA Class I and II–Expressing NSG/NRG Stocks at The Jackson Laboratory

Strain number HLA alleles Formal strain Other humanized genes
009617 HLA‐A2.1 NOD.Cg‐Mcph1Tg(HLA‐A2.1)1Enge Prkdcscid Il2rgtm1Wjl/Sz
012479 HLA‐DR1 NOD.Cg‐Tg(HLA‐DRA*0101, HLA‐DRB1*0101)1Dmz Prkdcscid Il2rgtm1Wjl/GckRolyJ
014570 HLA‐A2.1 NOD.Cg‐Prkdcscid Il2rgtm1Wjl Tg(HLA‐A/H2‐D/B2M)1Dvs/SzJ
017637 HLA‐DR4 NOD.Cg‐Prkdcscid Il2rgtm1Wjl H2‐Ab1b‐tm1Doi Tg(HLA‐DRB1)31Dmz/SzJ
017914 HLA‐DR4 NOD.Cg‐Rag1tm1Mom Il2rgtm1Wjl Tg(HLA‐DRA, HLA‐DRB1*0401)39‐2Kito/ScasJ
026561 HLA‐DQ8 NOD.Cg‐Prkdcscid H2‐Ab1b‐tm1Doi Il2rgtm1Wjl Tg(HLA‐DQA1, HLA‐DQB1)1Dv/SzJ
026936 HLA‐DQ8 NOD.Cg‐Prkdcscid H2‐Ab1b‐tm1Doi Il2rgtm1Wjl Tg(HLA‐DQA1, HLA‐DQB1)1Dv Tg(INS*)172Dvs/DvsJ INS
029295 HLA‐DR4 NOD.Cg‐Prkdcscid Il2rgtm1Wjl Tg(H2‐Ea‐HLA‐DRB1*0401*)1Dv/SzJ
030331 HLA‐DR1 NOD.Cg‐Tg(HLA‐DRA*0101, HLA‐DRB1*0101)1Dmz Prkdcscid H2‐Ab1b‐tm1Doi Il2rgtm1Wjl/SzJ
031566 HLA‐DR4 NOD.Cg‐Prkdcscid H2‐Ab1b‐tm1Doi Il2rgtm1Wjl Tg(H2‐Ea‐HLA‐DRB1*0401*)1Dv/SzJ
033127 HLA‐A2.1 NOD.Cg‐Rag1tm1Mom Flt3tm1Irl Mcph1Tg(HLA‐A2.1)1Enge Il2rgtm1Wjl/J
035844 HLA‐A2.1 NOD.Cg‐Flt3em1Akp Prkdcscid Il2rgtm1Wjl Tg(CMV‐IL3, CSF2, KITLG)1Eav Tg(HLA‐A/H2‐D/B2M)1Dvs/J IL3, CSF2, KITLG
035855 HLA‐A2.1, HLA‐DR4 NOD.Cg‐Rag1tm1Mom Il2rgtm1Wjl Tg(HLA‐A/H2‐D/B2M)Dvs Tg(HLA‐DRA, HLA‐DRB1*0401)39‐2Kito/J
039055 HLA‐DQ8 NOD.Cg‐Prkdcscid H2‐K1d‐em1Dvs H2‐Ab1g7‐em1Dvs H2‐D1b‐em5Dvs Il2rgtm1Wjl Tg(HLA‐DQA1, HLA‐DQB1)1Dv/DvsJ

Table 2.

Additional HLA Class I and II Alleles Available in the NOD Background at The Jackson Laboratory

Strain number HLA alleles Formal strain Other humanized genes
003354 HLA‐B27 NOD.B6‐Tg(HLA‐B27)30‐4Trg/DvsJ
004342 HLA‐B27 NOD.Cg‐Tg(B2M, HLA‐B*27:05)56‐3Trg/DvsJ B2M
006023 HLA‐DQ6 NOD.Cg‐H2‐Ab1b‐tm1Gru Tg(CD4, HLA‐DQA1, HLA‐DQB1)N8Ell/EllJ CD4
006024 HLA‐DQ6 NOD.Cg‐Rag1tm1Mom H2‐Ab1b‐tm1Gru Tg(CD4, HLA‐DQA1, HLA‐DQB1)N8Ell/EllJ CD4
010565 HLA‐A24 NOD.129P2(B6)‐B2mtm1Unc Tg(HLA‐A24/H2‐D/B2M)3Dvs/J
018907 HLA‐B7 NOD.Cg‐B2mtm1Unc Tg(B2M)55Hpl Tg(HLA‐B*0702)#Chmb/TdilJ B2M
027849 HLA‐A11 NOD.Cg‐B2mtm1Unc Tg(B2M)55Hpl Tg(HLA‐A*1101/H2‐Kb)#Sette/TdilJ
030434 HLA‐DR3 NOD.Cg‐H2‐Ab1b‐tm1Doi Tg(HLA‐DRA, HLA‐DRB1*0301)#Gjh/DvYtomJ
031857 HLA‐B39 NOD/ShiLtDvs‐H2‐K1d‐em1Dvs H2‐D1b‐em5Dvs Tg(HLA‐B39/H2‐D/B2M)2Dvs/DvsJ

Concluding Remarks

NOD stocks in which CRISPR/Cas9 technologies have been used to ablate various combinations of endogenous H2g7 MHC genes provide a means to test the potential of their human counterparts to contribute to autoimmune T1D susceptibility using a transgenic replacement approach. Such NOD‐HLA stocks also provide a means to test how T cells expressing autoreactive TCR molecules contributing to T1D in humans may undergo selection or potentially be functionally attenuated. Murine‐MHC‐deficient NOD stocks that instead express various human counterparts also allow the study of other clinically relevant autoimmune pathologies, including those that can arise as an undesirable side effect of ICI therapy. In these ways, the use of CRISPR/Cas9‐generated murine‐MHC‐deficient NOD mice as a platform for introduction of patient‐relevant HLA and TCR molecules provides ever‐improving models for development of clinically applicable interventions for T1D and other autoimmune diseases.

Author Contributions

David Serreze: Conceptualization; supervision; writing—original draft; writing—review and editing. Marissa Tousey‐Pfarrer: Investigation; writing—review and editing. Jeremy Racine: Conceptualization; formal analysis; investigation; writing—original draft; writing—review and editing.

Conflict of Interest

J.J.R. and D.V.S. hold a patent related to this work (Murine‐MHC‐deficient HLA‐transgenic NOD‐mouse Models for T1D Therapy Development; US‐11712026‐B2).

Acknowledgments

J.J.R. was supported by NIH grant R01DK136472 and D.V.S. was supported by NIH grant R01DK095735.

Serreze, D. V. , Tousey‐Pfarrer, M. , & Racine, J. J. (2025). Humanized mouse models for type 1 diabetes. Current Protocols, 5, e70224. doi: 10.1002/cpz1.70224

Published in the Immunology section

Data Availability Statement

Data presented in the figures are available upon request.

Literature Cited

  1. Atkinson, M. A. , & Leiter, E. H. (1999). The NOD mouse model of type 1 diabetes: As good as it gets? Nature Medicine, 5(6), 601–604. 10.1038/9442 [DOI] [PubMed] [Google Scholar]
  2. Chen, Y. G. , Mathews, C. E. , & Driver, J. P. (2018). The role of NOD mice in type 1 diabetes research: Lessons from the past and recommendations for the future. Frontiers in Endocrinology, 9, 51. 10.3389/fendo.2018.00051 [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Chu, V. T. , Weber, T. , Graf, R. , Sommermann, T. , Petsch, K. , Sack, U. , Volchkov, P. , Rajewsky, K. , & Kuhn, R. (2016). Efficient generation of Rosa26 knock‐in mice using CRISPR/Cas9 in C57BL/6 zygotes. BMC Biotechnology, 16, 4. 10.1186/s12896-016-0234-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Chuprin, J. , Buettner, H. , Seedhom, M. O. , Greiner, D. L. , Keck, J. G. , Ishikawa, F. , Shultz, L. D. , & Brehm, M. A. (2023). Humanized mouse models for immuno‐oncology research. Nature Reviews Clinical Oncology, 20(3), 192–206. 10.1038/s41571-022-00721-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Conner, D. A. (2004). Transgenic mouse production by zygote injection. Current Protocols in Molecular Biology, 68, 23.9.1–23.9.29. 10.1002/0471142727.mb2309s68 [DOI] [PubMed] [Google Scholar]
  6. Elliott, J. F. , Liu, J. , Yuan, Z.‐N. , Bautista‐Lopez, N. , Wallbank, S. L. , Suzuki, K. , Rayner, D. , Nation, P. , Robertson, M. A. , Liu, G. , & Kavanagh, K. M. (2003). Autoimmune cardiomyopathy and heart block development develop spontaneously in HLA‐DQ8 transgenic IAβ knockout NOD mice. Proceedings of the National Academy of Sciences of the United States of America, 100, 13447–13452. 10.1073/pnas.223555210 [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Fennessy, M. , Metcalfe, K. , Hitman, G. A. , Niven, M. , Biro, P. A. , Tuomilehto, J. , & Tuomilehto‐Wolf, E. (1994). A gene in the HLA class I region contributes to susceptibility to IDDM in the Finnish population. Diabetologia, 37, 937–944. 10.1007/BF00400951 [DOI] [PubMed] [Google Scholar]
  8. Flurkey, K. , Currer, J. M. , Leiter, E. H. , Witham, B. , & Jackson, L. (2009). The Jackson Laboratory Handbook on Genetically Standardized Mice (6th ed.). Bar Harbor, ME: Jackson Laboratory. [Google Scholar]
  9. Forman, J. , & Lindahl, K. F. (2002). Listing, location, binding motifs, and expression of nonclassical class I and related genes and molecules. Current Protocols in Immunology, 49, A.1M.1–A.1M.13. 10.1002/0471142735.ima01ms49 [DOI] [PubMed] [Google Scholar]
  10. Francis, J. H. , Jaben, K. , Santomasso, B. D. , Canestraro, J. , Abramson, D. H. , Chapman, P. B. , Berkenstock, M. , & Aronow, M. E. (2020). Immune checkpoint inhibitor‐associated optic neuritis. Ophthalmology, 127(11), 1585–1589. 10.1016/j.ophtha.2020.05.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Fu, J. , & Kim, Y. J. (2020). Autologously humanized mice for immune‐oncologic studies. Current Protocols in Pharmacology, 89(1), e76. 10.1002/cpph.76 [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Gaj, T. , Gersbach, C. A. , & Barbas, C. F. 3rd. (2013). ZFN, TALEN, and CRISPR/Cas‐based methods for genome engineering. Trends Biotechnology, 31(7), 397–405. 10.1016/j.tibtech.2013.04.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Grant, S. F. A. , Wells, A. D. , & Rich, S. S. (2020). Next steps in the identification of gene targets for type 1 diabetes. Diabetologia, 63(11), 2260–2269. 10.1007/s00125-020-05248-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Harms, D. W. , Quadros, R. M. , Seruggia, D. , Ohtsuka, M. , Takahashi, G. , Montoliu, L. , & Gurumurthy, C. B. (2014). Mouse genome editing using the CRISPR/Cas system. Current Protocols in Human Genetics, 83, 15.7.1–15.7.27. 10.1002/0471142905.hg1507s83 [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Hayward, S. L. , Bautista‐Lopez, N. , Suzuki, K. , Atrazhev, A. , Dickie, P. , & Elliott, J. F. (2006). CD4 T cells play major effector role and CD8 T cells initiating role in spontaneous autoimmune myocarditis of HLA‐DQ8 transgenic IAb knockout nonobese diabetic mice. Journal of Immunology, 176(12), 7715–7725. 10.4049/jimmunol.176.12.7715 [DOI] [PubMed] [Google Scholar]
  16. Howson, J. M. , Walker, N. M. , Clayton, D. , Todd, J. A. , & Diabetes Genetics Consortium . (2009). Confirmation of HLA class II independent type 1 diabetes associations in the major histocompatibility complex including HLA‐B and HLA‐A. Diabetes, Obesity and Metabolism, 11(1), 31–45. 10.1111/j.1463-1326.2008.01001.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Jarchum, I. , Baker, J. C. , Yamada, T. , Takaki, T. , Marron, M. P. , Serreze, D. V. , & DiLorenzo, T. P. (2007). In vivo cytotoxicity of insulin‐specific CD8+ T‐cells in HLA‐A*0201 transgenic NOD mice. Diabetes, 56(10), 2551–2560. 10.2337/db07-0332 [DOI] [PubMed] [Google Scholar]
  18. Jarchum, I. , & DiLorenzo, T. P. (2010). Ins2 deficiency augments spontaneous HLA‐A*0201‐restricted T cell responses to insulin. Journal of Immunology, 184(2), 658–665. 10.4049/jimmunol.0903414 [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Jing, Y. , Kong, Y. , McGinty, J. , Blahnik‐Fagan, G. , Lee, T. , Orozco‐Figueroa, S. , Bettini, M. L. , James, E. A. , & Bettini, M. (2022). T‐cell receptor/HLA humanized mice reveal reduced tolerance and increased immunogenicity of posttranslationally modified GAD65 epitope. Diabetes, 71(5), 1012–1022. 10.2337/db21-0993 [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Kartal, O. , & Atas, E. (2018). Bilateral optic neuritis secondary to nivolumab therapy: A case report. Medicina, 54(5), 82. 10.3390/medicina54050082 [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Kong, Y. , Jing, Y. , & Bettini, M. (2019). Generation of T cell receptor retrogenic mice. Current Protocols in Immunology, 125(1), e76. 10.1002/cpim.76 [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Larsen, M. T. , Kuhlmann, M. , Hvam, M. L. , & Howard, K. A. (2016). Albumin‐based drug delivery: Harnessing nature to cure disease. Molecular Cell Therapy, 4, 3. 10.1186/s40591-016-0048-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Le, A. X. , Bernhard, E. J. , Holterman, M. J. , Strub, S. , Parham, P. , Lacy, E. , & Engelhard, V. H. (1989). Cytotoxic T cell responses in HLA‐A2.1 transgenic mice. Recognition of HLA alloantigens and utilization of HLA‐A2.1 as a restriction element. Journal of Immunology, 142(4), 1366–1371. 10.4049/jimmunol.142.4.1366 [DOI] [PubMed] [Google Scholar]
  24. Leiter, E. (1998). NOD mice and related strains: Origins, husbandry, and biological introduction. In Leitner E. & Atkinson M. (Eds.), NOD mice and related strains: Research applications in diabetes, AIDA, cancer and other diseases (pp. 1–35). Austin: R. G. Landes. [Google Scholar]
  25. Leiter, E. H. (2001). The NOD mouse: A model for insulin‐dependent diabetes mellitus. Current Protocols in Immunology, 24, 15.9.1–15.9.23. 10.1002/0471142735.im1509s24 [DOI] [PubMed] [Google Scholar]
  26. Leiter, E. H. , & Schile, A. (2013). Genetic and pharmacologic models for type 1 diabetes. Current Protocols in Mouse Biology, 3(1), 9–19. 10.1002/9780470942390.mo120154 [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Liu, J. , Purdy, L. E. , Rabinovitch, S. , Jevnikar, A. M. , & Elliott, J. F. (1999). Major DQ8‐restricted T‐cell epitopes for human GAD65 mapped using human CD4, DQA1*0301, DQB1*0302 transgenic IA(null) NOD mice. Diabetes, 48(3), 469–477. 10.2337/diabetes.48.3.469 [DOI] [PubMed] [Google Scholar]
  28. Marron, M. P. , Graser, R. T. , Chapman, H. D. , & Serreze, D. V. (2002). Functional evidence for the mediation of diabetogenic T cell responses by HLA‐A2.1 MHC class I molecules through transgenic expression in NOD mice. Proceedings of the National Academy of Sciences of the United States of America, 99(21), 13753–13758. 10.1073/pnas.212221199 [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Michels, A. W. , Landry, L. G. , McDaniel, K. A. , Yu, L. , Campbell‐Thompson, M. , Kwok, W. W. , Jones, K. L. , Gottlieb, P. A. , Kappler, J. W. , Tang, Q. , Roep, B. O. , Atkinson, M. A. , Mathews, C. E. , & Nakayama, M. (2017). Islet‐derived CD4 T cells targeting proinsulin in human autoimmune diabetes. Diabetes, 66(3), 722–734. 10.2337/db16-1025 [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Mori, S. , Kurimoto, T. , Ueda, K. , Enomoto, H. , Sakamoto, M. , Keshi, Y. , Yamada, Y. , & Nakamura, M. (2018). Optic neuritis possibly induced by anti‐PD‐L1 antibody treatment in a patient with non‐small cell lung carcinoma. Case Reports in Ophthalmology, 9(2), 348–356. 10.1159/000491075 [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Nejentsev, S. , Howson, J. M. M. , Walker, N. M. , Szeszko, J. , Field, S. F. , Stevens, H. E. , Reynolds, P. , Hardy, M. , King, E. , Masters, J. , Hulme, J. , Maier, L. M. , Smyth, D. , Bailey, R. , Cooper, J. D. , Ribas, G. , Campbell, R. D. , Clayton, D. G. , Todd, J. A. , & The Wellcome Trust Case Control Consortium . (2007). Localization of type 1 diabetes susceptibility to the MHC class I genes HLA‐B and HLA‐A . Nature, 450, 887–892. 10.1038/nature06406 [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Niens, M. , Grier, A. E. , Marron, M. , Kay, T. W. , Greiner, D. L. , & Serreze, D. V. (2011). Prevention of "humanized" diabetogenic CD8 T‐cell responses in HLA‐transgenic NOD mice by a multipeptide coupled‐cell approach. Diabetes, 60(4), 1229–1236. 10.2337/db10-1523 [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Noble, J. A. , Valdes, A. M. , Varney, M. D. , Carlson, J. A. , Moonsamy, P. , Fear, A. L. , Lane, J. A. , Lavant, E. , Rappner, R. , Louey, A. , Concannon, P. , Mychaleckyj, J. C. , Erlich, H. A. , & Erlich, H. A. (2010). HLA class I and genetic susceptibility to type 1 diabetes: Results from the Type 1 Diabetes Genetics Consortium. Diabetes, 59(11), 2972–2979. 10.2337/db10-0699 [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Pascolo, S. , Bervas, N. , Ure, J. M. , Smith, A. G. , Lemonnier, F. A. , & Perarnau, B. (1997). HLA‐A2.1‐restricted education and cytolytic activity of CD8+ T lymphocytes from β2 microglobulin (β2m) HLA‐A2.1 monochain transgenic H‐2Db β2m double knockout mice. Journal of Experimental Medicine, 185(12), 2043–2051. 10.1084/jem.185.12.2043 [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Pathiraja, V. , Kuehlich, J. P. , Campbell, P. D. , Krishnamurthy, B. , Loudovaris, T. , Coates, P. T. , Brodnicki, T. C. , O'Connell, P. J. , Kedzierska, K. , Rodda, C. , Bergman, P. , Hill, E. , Purcell, A. W. , Dudek, N. L. , Thomas, H. E. , Kay, T. W. , & Mannering, S. I. (2015). Proinsulin‐specific, HLA‐DQ8, and HLA‐DQ8‐transdimer‐restricted CD4+ T cells infiltrate islets in type 1 diabetes. Diabetes, 64(1), 172–182. 10.2337/db14-0858 [DOI] [PubMed] [Google Scholar]
  36. Pearson, T. , Greiner, D. L. , & Shultz, L. D. (2008). Creation of "humanized" mice to study human immunity. Current Protocols in Immunology, 81, 15.21.1–15.21.21. 10.1002/0471142735.im1521s81 [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Phelps, C. , Huey, D. D. , & Niewiesk, S. (2023). Production of humanized mice through stem cell transfer. Current Protocols, 3(6), e800. 10.1002/cpz1.800 [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Pow Sang, L. , Surls, J. , Mendoza, M. , Casares, S. , & Brumeanu, T. (2015). HLA‐DR*0401 expression in the NOD mice prevents the development of autoimmune diabetes by multiple alterations in the T‐cell compartment. Cellular Immunology, 298(1–2), 54–65. 10.1016/j.cellimm.2015.09.003 [DOI] [PubMed] [Google Scholar]
  39. Qin, W. , Dion, S. L. , Kutny, P. M. , Zhang, Y. , Cheng, A. W. , Jillette, N. L. , Malhotra, A. , Geurts, A. M. , Chen, Y. G. , & Wang, H. (2015). Efficient CRISPR/Cas9‐mediated genome editing in mice by zygote electroporation of nuclease. Genetics, 200(2), 423–430. 10.1534/genetics.115.176594 [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Qin, W. , Kutny, P. M. , Maser, R. S. , Dion, S. L. , Lamont, J. D. , Zhang, Y. , Perry, G. A. , & Wang, H. (2016). Generating mouse models using CRISPR‐Cas9‐mediated genome editing. Current Protocols in Mouse Biology, 6(1), 39–66. 10.1002/9780470942390.mo150178 [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Racine, J. J. , Bachman, J. F. , Zhang, J. G. , Misherghi, A. , Khadour, R. , Kaisar, S. , Bedard, O. , Jenkins, C. , Abbott, A. , Forte, E. , Rainer, P. , Rosenthal, N. , Sattler, S. , & Serreze, D. V. (2024). Murine MHC‐deficient nonobese diabetic mice carrying human HLA‐DQ8 develop severe myocarditis and myositis in response to anti‐PD‐1 immune checkpoint inhibitor cancer therapy. Journal of Immunology, 212(8), 1287–1306. 10.4049/jimmunol.2300841 [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Racine, J. J. , Misherghi, A. , Dwyer, J. R. , Maser, R. , Forte, E. , Bedard, O. , Sattler, S. , Pugliese, A. , Landry, L. , Elso, C. , Nakayama, M. , Mannering, S. , Rosenthal, N. , & Serreze, D. V. (2023). HLA‐DQ8 supports development of insulitis mediated by insulin‐reactive human TCR‐transgenic T cells in nonobese diabetic mice. Journal of Immunology, 211(12), 1792–1805. 10.4049/jimmunol.2300303 [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Racine, J. J. , Stewart, I. , Ratiu, J. , Christianson, G. , Lowell, E. , Helm, K. , Allocco, J. , Maser, R. S. , Chen, Y. G. , Lutz, C. M. , Roopenian, D. , Schloss, J. , DiLorenzo, T. P. , & Serreze, D. V. (2018). Improved murine MHC‐deficient HLA transgenic NOD mouse models for type 1 diabetes therapy development. Diabetes, 67(5), 923–935. 10.2337/db17-1467 [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Ratiu, J. J. , Racine, J. J. , Hasham, M. G. , Wang, Q. , Branca, J. A. , Chapman, H. D. , Zhu, J. , Donghia, N. , Philip, V. , Schott, W. H. , Wasserfall, C. , Atkinson, M. A. , Mills, K. D. , Leeth, C. M. , & Serreze, D. V. (2017). Genetic and small molecule disruption of the AID/RAD51 axis similarly protects nonobese diabetic mice from type 1 diabetes through expansion of regulatory B lymphocytes. Journal of Immunology, 198(11), 4255–4267. 10.4049/jimmunol.1700024 [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Roopenian, D. C. , & Akilesh, S. (2007). FcRn: The neonatal Fc receptor comes of age. Nature Reviews Immunology, 7(9), 715–725. 10.1038/nri2155 [DOI] [PubMed] [Google Scholar]
  46. Rotwein, P. , Yokoyama, S. , Didier, D. K. , & Chirgwin, J. M. (1986). Genetic analysis of the hypervariable region flanking the human insulin gene. American Journal of Human Genetics, 39(3), 291–299. [PMC free article] [PubMed] [Google Scholar]
  47. Sawada, S. , Scarborough, J. D. , Killeen, N. , & Littman, D. R. (1994). A lineage‐specific transcriptional silencer regulates CD4 gene expression during T lymphocyte development. Cell, 77(6), 917–929. 10.1016/0092-8674(94)90140-6 [DOI] [PubMed] [Google Scholar]
  48. Schloss, J. , Ali, R. , Racine, J. J. , Chapman, H. D. , Serreze, D. V. , & DiLorenzo, T. P. (2018). HLA‐B*39:06 efficiently mediates type 1 diabetes in a mouse model incorporating reduced thymic insulin expression. Journal of Immunology, 200(10), 3353–3363. 10.4049/jimmunol.1701652 [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Sengul Samanci, N. , Ozan, T. , Celik, E. , & Demirelli, F. H. (2020). Optic neuritis related to atezolizumab treatment in a patient with metastatic non‐small‐cell lung cancer. JCO Oncology Practice, 16(2), 96–98. 10.1200/JOP.19.00438 [DOI] [PubMed] [Google Scholar]
  50. Serreze, D. V. , Dwyer, J. R. , & Racine, J. J. (2024). Advancing animal models of human type 1 diabetes. Cold Spring Harbor Perspectives in Medicine, 14(10), a041587. 10.1101/cshperspect.a041587 [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Serreze, D. V. , Leiter, E. H. , Christianson, G. J. , Greiner, D. , & Roopenian, D. C. (1994). MHC class I deficient NOD‐B2mnull mice are diabetes and insulitis resistant. Diabetes, 43, 505–509. 10.2337/diab.43.3.505 [DOI] [PubMed] [Google Scholar]
  52. Shultz, L. D. , Brehm, M. A. , Garcia‐Martinez, J. V. , & Greiner, D. L. (2012). Humanized mice for immune system investigation: Progress, promise and challenges. Nature Reviews Immunology, 12(11), 786–798. 10.1038/nri3311 [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Takaki, T. , Marron, M. P. , Mathews, C. E. , Guttmann, S. T. , Bottino, R. , Trucco, M. , DiLorenzo, T. P. , & Serreze, D. V. (2006). HLA‐A*0201‐restricted T cells from humanized NOD mice recognize autoantigens of potential clinical relevance to type 1 diabetes. Journal of Immunology, 176(5), 3257–3265. 10.4049/jimmunol.176.5.3257 [DOI] [PubMed] [Google Scholar]
  54. Taneja, V. , Behrens, M. , Cooper, L. T. , Yamada, S. , Kita, H. , Redfield, M. M. , Terzic, A. , & David, C. (2007). Spontaneous myocarditis mimicking human disease occurs in the presence of an appropriate MHC and non‐MHC background in transgenic mice. Journal of Molecular and Cellular Cardiology, 42(6), 1054–1064. 10.1016/j.yjmcc.2007.03.898 [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Taylor, J. A. , Havari, E. , McInerney, M. F. , Bronson, R. , Wucherpfennig, K. W. , & Lipes, M. A. (2004). Spontaneous model for autoimmune myocarditis using the human MHC molecule HLa‐DQ8. Journal of Immunology, 172, 2651–2658. 10.4049/jimmunol.172.4.2651 [DOI] [PubMed] [Google Scholar]
  56. Unger, W. W. , Pearson, T. , Abreu, J. R. , Laban, S. , van der Slik, A. R. , der Kracht, S. M. , Kester, M. G. , Serreze, D. V. , Shultz, L. D. , Griffioen, M. , Drijfhout, J. W. , Greiner, D. L. , & Roep, B. O. (2012). Islet‐specific CTL cloned from a type 1 diabetes patient cause beta‐cell destruction after engraftment into HLA‐A2 transgenic NOD/scid/IL2RG null mice. PLoS One, 7(11), e49213. 10.1371/journal.pone.0049213 [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Verhagen, J. , Smith, E. L. , Whettlock, E. M. , Macintyre, B. , & Peakman, M. (2018). Proinsulin‐mediated induction of type 1 diabetes in HLA‐DR4‐transgenic mice. Scientific Reports, 8(1), 14106. 10.1038/s41598-018-32546-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Verma, B. , & Wesa, A. (2020). Establishment of humanized mice from peripheral blood mononuclear cells or cord blood CD34+ hematopoietic stem cells for immune‐oncology studies evaluating new therapeutic agents. Current Protocols in Pharmacology, 89(1), e77. 10.1002/cpph.77 [DOI] [PubMed] [Google Scholar]
  59. Wefers, B. , Meyer, M. , Hensler, S. , Panda, S. , Ortiz, O. , Wurst, W. , & Kuhn, R. (2012). Gene editing in one‐cell embryos by zinc‐finger and TAL nucleases. Current Protocols in Mouse Biology, 2(4), 347–364. 10.1002/9780470942390.mo120177 [DOI] [PubMed] [Google Scholar]
  60. Wen, L. , Wong, F. S. , Sherwin, R. , & Mora, C. (2002). Human DQ8 can substitute for murine I‐Ag7 in the selection of diabetogenic T cells restricted to I‐Ag7. Journal of Immunology, 168(7), 3635–3640. 10.4049/jimmunol.168.7.3635 [DOI] [PubMed] [Google Scholar]
  61. Wicker, L. S. , Leiter, E. H. , Todd, J. A. , Renjilian, R. J. , Peterson, E. , Fischer, P. A. , Podolin, P. L. , Zijlstra, M. , Jaenisch, R. , & Peterson, L. B. (1994). β2‐Microglobulin‐deficient NOD mice do not develop insulitis or diabetes. Diabetes, 43, 500–504. 10.2337/diab.43.3.500 [DOI] [PubMed] [Google Scholar]
  62. Yu, B. , Gauthier, L. , Hausmann, D. H. , & Wucherpfennig, K. W. (2000). Binding of conserved islet peptides by human and murine MHC class II molecules associated with susceptibility to type I diabetes. European Journal of Immunology, 30(9), 2497–2506. [DOI] [PubMed] [Google Scholar]
  63. Zhumabekov, T. , Corbella, P. , Tolaini, M. , & Kioussis, D. (1995). Improved version of a human CD2 minigene based vector for T cell‐specific expression in transgenic mice. Journal of Immunological Methods, 185(1), 133–140. 10.1016/0022-1759(95)00124-S [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Data presented in the figures are available upon request.


Articles from Current Protocols are provided here courtesy of Wiley

RESOURCES