16
PERSPECTIVE dmm.biologists.org 318 Introduction A major complication and limitation to the broad application of hematopoietic stem cell transplantation (HSCT) is graft-versus- host disease (GvHD), which results from immune-mediated attack of recipient tissue by donor T cells contained in the transplant. GvHD accounts for 15-30% of deaths that occur following allogeneic HSCT that is carried out to treat malignant diseases, and is a major cause of morbidity in up to 50% of transplant recipients (Ferrara et al., 2009). This high incidence is despite the use of aggressive immunosuppressive therapies after transplantation, as well as allele-level human leukocyte antigen (HLA) matching between donor and recipient. Despite recent advances to reduce the incidence of GvHD through altering prophylactic regimens and reducing the intensity of conditioning prior to transplantation, effective treatments for GvHD are lacking. Thus, accurate and reproducible experimental models of GvHD are essential for advancing our fundamental understanding of this disorder and for developing new treatments. In this Perspective, we provide an overview of available mouse models of acute and chronic GvHD, discuss their uses and limitations, and provide guidance on selecting an appropriate model. We also briefly discuss the graft-versus-leukemia (GVL) effect, although readers are referred to other recent reviews for more detail on this topic (Bleakley and Riddell, 2004; Ringden et al., 2009). We begin by describing the clinical symptoms and immunopathology of GvHD. GvHD immunopathology in humans and mice In humans, GvHD manifests as acute GvHD (aGvHD), which occurs within 100 days of transplant, or as chronic GvHD (cGvHD), which typically develops 100 days after transplantation and can take 2-5 years to become clinically evident. This temporal distinction does not necessarily translate to mouse models of the disease, which can differ in time of onset and are mainly defined by their clinical phenotype: cGvHD develops within weeks after transplantation in most mouse models. In addition, in humans, aGvHD typically precedes cGvHD, although in some cases cGvHD can occur without the occurrence of clinically obvious aGvHD. One factor that confounds the translation of findings in mouse models to the human disease is that most patients are given immunosuppressive therapy to prevent aGvHD, and these medications might influence the development of cGvHD. Interestingly, interventions to prevent aGvHD in the clinic have, in general, not significantly decreased rates of cGvHD in humans (Lee, 2010). The sequence of events that lead to the development of GvHD has largely been defined using mouse models. Early work established that T-cell alloreactivity is the underlying cause of the disease (Korngold and Sprent, 1978; Sprent et al., 1986). The pathology of both acute and chronic mouse models of GvHD relies on T-cell alloreactivity, but each form has a different phenotype owing to differential involvement of cytotoxic (CD8 + ) or helper (CD4 + ) T-cell subsets. Donor CD8 + T cells are activated when their T-cell receptor (TCR) binds to recipient peptides presented in the context of recipient class I major histocompatibility complex (MHC) molecules (see Box 1 for a glossary of immunological terms). T-cell activation also requires the engagement of co- stimulatory receptors on activated donor T cells by their cognate ligands expressed on donor antigen-presenting cells (APCs). CD8 + T-cell cytotoxicity is mediated by perforin, granzymes and Fas ligand, and inflammatory cytokines augment this response. Donor CD4 + T cells are activated when their TCR binds to exogenous Disease Models & Mechanisms 4, 318-333 (2011) doi:10.1242/dmm.006668 Mouse models of graft-versus-host disease: advances and limitations Mark A. Schroeder 1 and John F. DiPersio 1, * 1 Division of Oncology, Siteman Cancer Center, Washington University School of Medicine, St Louis, MO 63110, USA *Author for correspondence ([email protected]) © 2011. Published by The Company of Biologists Ltd This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial Share Alike License (http://creativecommons.org/licenses/by- nc-sa/3.0), which permits unrestricted non-commercial use, distribution and reproduction in any medium provided that the original work is properly cited and all further distributions of the work or adaptation are subject to the same Creative Commons License terms. The limiting factor for successful hematopoietic stem cell transplantation (HSCT) is graft-versus-host disease (GvHD), a post-transplant disorder that results from immune-mediated attack of recipient tissue by donor T cells contained in the transplant. Mouse models of GvHD have provided important insights into the pathophysiology of this disease, which have helped to improve the success rate of HSCT in humans. The kinetics with which GvHD develops distinguishes acute from chronic GvHD, and it is clear from studies of mouse models of GvHD (and studies of human HSCT) that the pathophysiology of these two forms is also distinct. Mouse models also further the basic understanding of the immunological responses involved in GvHD pathology, such as antigen recognition and presentation, the involvement of the thymus and immune reconstitution after transplantation. In this Perspective, we provide an overview of currently available mouse models of acute and chronic GvHD, highlighting their benefits and limitations, and discuss research and clinical opportunities for the future. Disease Models & Mechanisms DMM

Mouse models of graft-versus-host disease: advances and … › content › dmm › 4 › 3 › 318.full.pdf · PERSPECTIVE 318 dmm.biologists.org Introduction A major complication

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Mouse models of graft-versus-host disease: advances and … › content › dmm › 4 › 3 › 318.full.pdf · PERSPECTIVE 318 dmm.biologists.org Introduction A major complication

PERSPECTIVE

dmm.biologists.org318

IntroductionA major complication and limitation to the broad application ofhematopoietic stem cell transplantation (HSCT) is graft-versus-host disease (GvHD), which results from immune-mediated attackof recipient tissue by donor T cells contained in the transplant.GvHD accounts for 15-30% of deaths that occur followingallogeneic HSCT that is carried out to treat malignant diseases,and is a major cause of morbidity in up to 50% of transplantrecipients (Ferrara et al., 2009). This high incidence is despitethe  use of aggressive immunosuppressive therapies aftertransplantation, as well as allele-level human leukocyte antigen(HLA) matching between donor and recipient. Despite recentadvances to reduce the incidence of GvHD through alteringprophylactic regimens and reducing the intensity of conditioningprior to transplantation, effective treatments for GvHD are lacking.Thus, accurate and reproducible experimental models of GvHDare essential for advancing our fundamental understanding of thisdisorder and for developing new treatments. In this Perspective,we provide an overview of available mouse models of acute andchronic GvHD, discuss their uses and limitations, and provideguidance on selecting an appropriate model. We also briefly discussthe graft-versus-leukemia (GVL) effect, although readers arereferred to other recent reviews for more detail on this topic(Bleakley and Riddell, 2004; Ringden et al., 2009). We begin bydescribing the clinical symptoms and immunopathology of GvHD.

GvHD immunopathology in humans and miceIn humans, GvHD manifests as acute GvHD (aGvHD), whichoccurs within 100 days of transplant, or as chronic GvHD (cGvHD),which typically develops 100 days after transplantation and can take2-5 years to become clinically evident. This temporal distinctiondoes not necessarily translate to mouse models of the disease, whichcan differ in time of onset and are mainly defined by their clinicalphenotype: cGvHD develops within weeks after transplantation inmost mouse models. In addition, in humans, aGvHD typicallyprecedes cGvHD, although in some cases cGvHD can occurwithout the occurrence of clinically obvious aGvHD. One factorthat confounds the translation of findings in mouse models tothe  human disease is that most patients are givenimmunosuppressive therapy to prevent aGvHD, and thesemedications might influence the development of cGvHD.Interestingly, interventions to prevent aGvHD in the clinic have,in general, not significantly decreased rates of cGvHD in humans(Lee, 2010).

The sequence of events that lead to the development of GvHDhas largely been defined using mouse models. Early workestablished that T-cell alloreactivity is the underlying cause of thedisease (Korngold and Sprent, 1978; Sprent et al., 1986). Thepathology of both acute and chronic mouse models of GvHD relieson T-cell alloreactivity, but each form has a different phenotypeowing to differential involvement of cytotoxic (CD8+) or helper(CD4+) T-cell subsets. Donor CD8+ T cells are activated when theirT-cell receptor (TCR) binds to recipient peptides presented in thecontext of recipient class I major histocompatibility complex(MHC) molecules (see Box 1 for a glossary of immunologicalterms). T-cell activation also requires the engagement of co-stimulatory receptors on activated donor T cells by their cognateligands expressed on donor antigen-presenting cells (APCs). CD8+

T-cell cytotoxicity is mediated by perforin, granzymes and Fasligand, and inflammatory cytokines augment this response. DonorCD4+ T cells are activated when their TCR binds to exogenous

Disease Models & Mechanisms 4, 318-333 (2011) doi:10.1242/dmm.006668

Mouse models of graft-versus-host disease:advances and limitationsMark A. Schroeder1 and John F. DiPersio1,*

1Division of Oncology, Siteman Cancer Center, Washington University School ofMedicine, St Louis, MO 63110, USA*Author for correspondence ([email protected])© 2011. Published by The Company of Biologists LtdThis is an Open Access article distributed under the terms of the Creative CommonsAttribution Non-Commercial Share Alike License (http://creativecommons.org/licenses/by-nc-sa/3.0), which permits unrestricted non-commercial use, distribution and reproductionin any medium provided that the original work is properly cited and all furtherdistributions of the work or adaptation are subject to the same Creative Commons Licenseterms.

The limiting factor for successful hematopoietic stem cell transplantation (HSCT) is graft-versus-host disease (GvHD), apost-transplant disorder that results from immune-mediated attack of recipient tissue by donor T cells contained in thetransplant. Mouse models of GvHD have provided important insights into the pathophysiology of this disease, whichhave helped to improve the success rate of HSCT in humans. The kinetics with which GvHD develops distinguishesacute from chronic GvHD, and it is clear from studies of mouse models of GvHD (and studies of human HSCT) that thepathophysiology of these two forms is also distinct. Mouse models also further the basic understanding of theimmunological responses involved in GvHD pathology, such as antigen recognition and presentation, the involvementof the thymus and immune reconstitution after transplantation. In this Perspective, we provide an overview of currentlyavailable mouse models of acute and chronic GvHD, highlighting their benefits and limitations, and discuss researchand clinical opportunities for the future.

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Page 2: Mouse models of graft-versus-host disease: advances and … › content › dmm › 4 › 3 › 318.full.pdf · PERSPECTIVE 318 dmm.biologists.org Introduction A major complication

Disease Models & Mechanisms 319

Mouse models of GvHD PERSPECTIVE

peptides presented in the context of class II MHC molecules onrecipient APCs. CD4+ T-cell activation results in the developmentof a T-helper (Th)1 inflammatory response [marked by theproduction of interferon- (IFN), interleukin-12 (IL-12) and IL-2] or a Th2 response (marked by the production of IL-4, IL-5, IL-6 and IL-10). In mice, cGvHD mainly involves a Th2 response,whereas aGvHD is mainly Th1 biased.

Cellular isolates used for HSCT in mice contain, among othercells, stem cells and T cells. Within 24 hours after transplantation,T cells traffic to secondary lymphoid organs (Panoskaltsis-Mortariet al., 2004; Beilhack et al., 2005). This initial migration of donorT cells and their interaction with recipient APCs is believed to playa crucial role in alloantigen priming and donor T-cell activationprior to egress to primary target organs (Kim et al., 2003; Beilhacket al., 2005). Activated donor T cells traffic and cause cytotoxicityin the gut, skin, liver, lung, thymus and lymph nodes. This uniquetissue restriction is observed both in mouse models of GvHD andin human patients suffering from GvHD. Chemokines and adhesionmolecules also play a crucial role in T-cell trafficking (Wysocki etal., 2005).

aGvHD immunopathologyaGvHD involves the direct cytotoxic effects of donor T cells onrecipient tissues, the activation of APCs and an inflammatorycascade known as a ‘cytokine storm’, which involves the productionof cytokines, including IL-1, IL-6, IL-12, IFN and tumor necrosisfactor- (TNF). IFN has diverse roles, regulating immunesuppression as well as cytotoxicity (for a review, see Lu and Waller,2009). The impact of IFN on aGvHD is temporally related: IFNcan have immunosuppressive effects if present immediately aftertransplant but inflammatory effects if it is added later (Brok et al.,1998). This inflammatory cascade contributes to a systemicsyndrome of weight loss, diarrhea, skin changes and high mortality.

Key events involved in the development of aGvHD in mice areillustrated in Fig. 1. First, priming of the immune responseestablishes a milieu for enhanced T-cell activation and expansion,and occurs as a direct result of ‘conditioning’. Conditioningsuppresses the recipient’s immune system and is applied to allowengraftment of donor hematopoietic stem cells. It can be achievedwith cytotoxic agents such as irradiation or chemotherapy, or withimmunosuppressive agents such as anti-thymocyte globulin ordrugs that target recipient T cells, such as fludarabine. The mostcommon method of conditioning in mouse models is total bodyirradiation, which ablates the recipient’s bone marrow, allowingdonor stem cell engraftment and preventing rejection of the graftby limiting the proliferation of recipient T cells in response to donorcells. Subsequent activation of donor T cells depends on cognateantigen recognition. T cells are also activated by non-cognatestimulation by cytokines such as IL-1 and TNF that are releasedduring priming by many different cell types (Ferrara et al., 1993).Subsequent expansion of donor T cells and release of cytokinessuch as IFN leads to the activation of additional effector cell types,including neutrophils, monocytes and natural killer (NK) cells,which can result in further donor T-cell proliferation independentof their interactions with cognate antigen (Teshima et al., 2002).In addition, proinflammatory macrophages are activated, which canrelease IL-1, TNF and nitric oxide (Nestel et al., 1992; Cooke etal., 1998).

Box 1. Glossary of immunological termsAntigen cross presentation: a process by which exogenous antigens derivedfrom endogenous cells (such as damaged epithelium in the case of GvHD) aretaken up by APCs and presented, typically in MHC class I molecules, to CD8+ Tcells. In setting of GvHD, donor APCs cross-prime donor T cells by presentingrecipient antigens.Antigen presentation: the process by which either self or non-self antigensare captured, processed and presented on the surface of APCs for cognateantigen recognition by lymphocytes.Antigen-presenting cells (APCs): a heterogenous population of cells(including B cells, monocytes/macrophages and dendritic cells) that presentself and non-self antigens to T cells.Chimerism: the presence of two or more genetically distinct cell populations(from donor and recipient) in the same organism, as occurs following HSCT;can be measured by determining the percentage of donor cells present in theblood or bone marrow through genetic or immunocytometry testing.Cognate antigen recognition: recognition of antigen by T or B cellsexpressing receptors specific for that antigen.Cytokine storm: elevated levels of inflammatory mediators (such as TNF, IL-1and IL-6) that potentiate further stimulation of immune effector cells such as Tcells and macrophages in a positive-feedback loop; can occur in GvHD or inresponse to infections.Effector T cells: mature activated T cells that produce large amounts ofcytokines (in the case of CD4+ T cells) or kill target cells via perforin andgranzymes (in the case of CD8+ T cells).Epitope: a specific region of an antigen that is recognized by antibodies, or byB-cell or T-cell receptors.Epitope spreading: a normal or pathological (in the case of auto-antigens)immune response in which epitopes distinct from the inducing epitopebecome targets of the immune response. This typically occurs in the setting ofchronic inflammatory responses. APCs process and present a variety ofepitopes from the same larger antigenic peptide, resulting in an increasedrepertoire of T-cell responses against multiple epitopes from the originalantigenic peptide.Major histocompatibility complex (MHC): a cluster of highly polymorphicgenes that encode proteins involved in antigen presentation (also known asHLA antigens in humans and H2 in mice); important for enabling the immunesystem to differentiate between self and foreign tissues. MHC class I enablespresentation of mainly endogenous antigens to CD8+ T cells, whereas MHCclass II enables presentation of mainly exogenous antigens to CD4+ T cells.Minor histocompatibility antigens (miHAs): a class of polymorphic antigensthought to arise from polymorphisms in the genes encoding common cellularproteins; presented by MHC class I and are often responsible for organrejection and GvHD in MHC-matched transplants. An example is the H-Yantigen.Regulatory T (TReg) cells: a subset of CD4+ T cells that is defined byexpression of the transcription factor FoxP3 and that suppresses immuneresponses to a wide variety of antigens, including alloantigens and autologousantigens.T helper 1 (Th1) cell: a CD4+ T-cell effector subset characterized by theproduction of the cytokines IL-2, IFN and IL-12. Th1 cells can activatemacrophages and B cells.T helper 2 (Th2) cell: a CD4+ T-cell effector subset characterized by theproduction of the cytokines IL-4, IL-5, IL-6 and IL-10. Th2 cells provide help to Bcells for antibody production.T-cell alloreactivity: the T-cell response induced when a graft is performedwith tissue of the same species but with a different genotype, owing to therecognition of foreign tissue (allograft) by the T cells.Th17 cells: a subset of CD4+ T effector cells that expresses proinflammatorymolecules of the IL-17 family and mediates cytotoxic responses resulting ininflammation and tissue injury.Thymic negative selection (also known as central tolerance): the deletionof self-reactive T cells (i.e. T cells with autoimmune potential) during theirdevelopment in the thymus.

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Page 3: Mouse models of graft-versus-host disease: advances and … › content › dmm › 4 › 3 › 318.full.pdf · PERSPECTIVE 318 dmm.biologists.org Introduction A major complication

dmm.biologists.org320

Mouse models of GvHDPERSPECTIVE

The final step leading to end organ damage in aGvHD is T-cell-mediated tissue toxicity, which involves soluble mediators,including TNF, perforin, granzymes, Fas and Fas ligand (Bakeret al., 1996; Braun et al., 1996; Graubert et al., 1997; Jiang et al.,2001; Maeda et al., 2005). In addition, T helper 17 (Th17) cells area subset of T cells that have recently been shown to contribute tothis final cytotoxic phase and can cause increased inflammationand cytotoxicity when supplemented in the graft in anexperimental setting. A recent report demonstrated that Th17 cellswere sufficient but not necessary to cause GvHD (Iclozan et al.,

2009). Surprisingly, however, some studies have reported that theirabsence from the graft can also worsen GvHD (Yi et al., 2008;Carlson et al., 2009; Kappel et al., 2009). It is hoped that futurestudies will resolve this discrepancy to clarify the role of Th17 cellsin GvHD pathology. A crucial counterpart to Th17 cells areregulatory T (TReg) cells, which have been shown to suppress awide range of immune responses, including alloreactive andautoreactive T-cell responses (Edinger et al., 2003; Sakaguchi etal., 2006).

cGvHD immunopathologyUnlike aGvHD, cGvHD typically presents as an autoimmune-likesyndrome. As well as effects mediated by T cells, cGvHD involvesB-cell stimulation, autoantibody production and systemic fibrosis.Mouse models of cGvHD are associated with prolonged survivalcompared with mouse models of aGvHD. Additional clinicalcharacteristics depend on the mouse model and can includesplenomegaly and lymphadenopathy resulting from B-cellexpansion, glomerulonephritis, biliary cirrhosis and scleroderma.

The immunopathology underlying cGvHD is less wellunderstood than that of aGvHD, in part because of the limitationsof currently available mouse models. Unlike aGvHD, there is nounifying theory for the development of cGvHD, and thepathological mechanisms differ depending on the model system.Mouse models of cGvHD typically involve three main pathologicalmechanisms: autoantibody production, pro-fibrotic pathways anddefective thymic function. Here, we describe the main events thatunderlie the phenotype, although models exploring the three mainmechanisms will be discussed in detail later.

A summary of our current knowledge of the presumed sequenceof events leading to end organ damage in cGvHD is outlined inFig. 2. First, unlike in aGvHD, immune priming in cGvHD modelsis not dependent on tissue damage from conditioning: comparedwith aGvHD models, the initial inflammatory milieu is reducedor absent in cGvHD models, which involve reduced levels ofradiation or no conditioning at all. The outcome of transplantationis mixed T-cell chimerism in most models. Thymic damage canresult in dysfunctional negative selection of autoreactive T cells,and there can be decreased TReg cell number or function. Inaddition, donor and recipient B cells recognize and processextracellular antigens for presentation in the context of MHC. Allof these processes can contribute to the initiation of cGvHD.Second, unlike in aGvHD, T-cell activation is not dependent onrecipient APCs, but is a mix of cognate antigen recognition andcross-priming of donor T cells. CD4+ cells are necessary andsufficient to cause cGvHD in mouse models, whereas cytotoxicCD8+ T cells are not required, in contrast to aGvHD (Rolink andGleichmann, 1983; Rolink et al., 1983; Zhang et al., 2006). RecipientB cells are activated by minor antigens and Th2 cytokines, andmature to present processed antigens in the context of MHC todonor CD4+ T cells, which in turn co-stimulate B cells to produceautoantibodies and additional stimulatory cytokines (Morris et al.,1990a; Shimabukuro-Vornhagen et al., 2009). Third, T and B cellsclonally expand in response to antigenic stimulation and co-stimulation, and form memory and effector cells. In addition,epitope spreading can occur, resulting in oligoclonal T-cellexpansion. Fourth, clonally expanded T and B cells can traffic totarget organ sites and mediate damage through cytokine-mediated

Host cell

Chemotherapy

Damage to host tissues (skin, gut, liver)

Skin, gut, liver, lung

Lymph nodes

Blood and lymph

APC maturationand antigenpresentation

T-cellexpansion

Tissue damage

Trafficking

TNFαIL-1IFNγ

Perforin/granzymeFas/Fas-ligandTNFα

IL-1IFNγ

TNFαIL-1IL-6

MHC I MHC II

RecipientAPC

MHC I MH

RecipientAPC

Endogenousantigen

Exogenousantigen

DonorAPC

Inflammatorycytokines

CD8+

T cellCD4+

T cell

ood and lymp

CD4+ orCD8+ T cell

Tissue-residentDC or

macrophage

Lymph nodes and target organs

Skin, gut, liver, lung

A Immune priming

B T-cell activation and co-stimulation

C Alloreactive T-cell expansion

D Trafficking

E Tissue injury and destruction

Damage

XRT

Fig. 1. Steps to aGvHD in mice. The progression of events occurring in thedevelopment of aGvHD in the mouse is illustrated. The five crucial steps ofimmune priming (A), activation (B), T-cell expansion (C), T-cell trafficking (D)and host tissue injury (E) are outlined. DC, dendritic cell; XRT, radiationconditioning. See main text for full details.

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Page 4: Mouse models of graft-versus-host disease: advances and … › content › dmm › 4 › 3 › 318.full.pdf · PERSPECTIVE 318 dmm.biologists.org Introduction A major complication

Disease Models & Mechanisms 321

Mouse models of GvHD PERSPECTIVE

fibrosis and chronic inflammation. Additionally, bone-marrow-derived T cells inappropriately selected in a dysfunctional thymus,or through dysfunctional peripheral tolerance mechanisms, trafficto target organs, can be activated by self antigens and perpetuatethe Th2 response. Finally, tissue injury results from activated Tand B cells that release inflammatory and fibrosing cytokines, suchas IL-13, IL-10 and IL-4, and activated macrophages producingtransforming growth factor- (TGF) stimulate collagenproduction from fibroblasts, collectively leading to a scleroderma-like syndrome. Autoantibodies against double-stranded DNA,single-stranded DNA and chromatin can be deposited at the siteof target organs, leading to immune-complex glomerulonephritisor causing activation of the platelet derived growth factor receptor,leading to fibrosis (Svegliati et al., 2007).

Mouse models of aGvHDMost mouse models of aGvHD (summarized in Table 1) involvethe transplantation of T-cell-depleted bone marrow supplementedwith varying numbers and phenotypic classes of donor lymphocytes(either splenocytes or lymph node T cells) into lethally irradiatedrecipients. The bone marrow provides donor stem cells that allowhematopoietic reconstitution after transplant; T-cell depletion iscarried out to control the dose and type of immune cells that aredelivered.

The discovery of the MHC and minor histocompatibility antigens(miHAs) has been integral to advancing the field of HSCT.Differences in MHC antigens are largely responsible for immune-mediated rejection of foreign tissues and cells; however, even in anMHC-matched setting, differences in miHAs can cause grafts tobe slowly rejected. Unlike humans, inbred mouse strains are closelyrelated with respect to MHC antigens (note that class I MHC isreferred to as H2 in mice). Although two strains of mice mightshare the same H2 (i.e. they are MHC-I-matched), differences inmiHAs can result in GvHD in the context of experimental HSCT.MHC-mismatched and miHA-mismatched models are discussedbelow. In addition, we describe more recently developed modelsinvolving the xenotransplantation of human cells intoimmunodeficient mice.

The severity of aGvHD depends on several factors. First, the doseand type of T-cell subsets (i.e. CD4+, CD8+ or TReg cells) that aretransferred with donor bone marrow can affect the severity of thedisease (Sprent et al., 1988; Korngold, 1992; Edinger et al., 2003).Second, irradiation dose is proportional to the degree of tissuedamage and the subsequent cytokine storm, and thus is directlyproportional to aGvHD-related mortality in the mouse (Hill et al.,1997; Schwarte and Hoffmann, 2005; Schwarte et al., 2007). Inaddition, the dose of irradiation that is myeloablative varies bymouse strain: inbred strains are generally more susceptible toradiation than their F1 progeny, and different strains showsusceptibility differences (e.g. BALB/c>C3H>C57Bl/6). Third,genetic disparities play a major role in disease. For example, evenmice that are MHC matched can vary by miHAs that influenceaGvHD severity. Fourth, variation in environmental pathogensbetween labs and in mice from different suppliers can affect GvHD(Nestel et al., 1992). Therefore, the same model of aGvHD can varybetween investigators because of differences in radiation dose anddelivery rate, differences in pathogens between animal facilities,and differences in T-cell dose and sources of T cells.

MHC I MHC II

CD8+

T cellCD4+

T cell

DonorAPC

Thymic damage Decreased numberof TReg cells

Antigen

Antibody

B-cell priming

B cell

Antigen-stimulatedclonal T-cell expansion

AutoreactiveT cell

Antigenrecognition

Cytokines(IL-13,

IL-4, IL-10)

Cytokinereceptor

TGFβ

CollagenDirectcytotoxicity

Epitope spreading

AutoantibodiesB-cell clonalexpansion

A Immune priming

B T-cell and B-cell activation and co-stimulation

C T-cell and B-cell expansion and autoantibody production

E Tissue injury, inflammation and fibrosis

ThymusAutoreactive

T cell

TReg cells MHC II

B cell

Th2cytokines

(IL-4, IL-10)

+

RecipientAPC

Recipient anddonor B cells

MHC I MH

B T-cell an

RecipientAPC

Endogenousantigen

Exogenousantigen

CD4+ orCD8+ T cells

Skin, gut, liver, lung, salivary glands, oral mucosa

Blood and lymphd and lymph

D Trafficking

B cell

B cell

Autoantibodies

AutoreactiveT cell

Target organs

AutoactivatedT cell

Macrophage

Host cell

+

+

Autoantibodies

Fibroblasts

PDGFreceptor

Fig. 2. Steps to cGvHD in mice. The progression of events occurring in thedevelopment of cGvHD in the mouse is illustrated. The five steps illustrateimmune priming (A), activation of T and B cells (B), T-cell expansion and B-cellautoantibody production (C), trafficking to sites of tissue damage (D), and endorgan damage through chronic inflammation and fibrosis (E). See main textfor full details.

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Page 5: Mouse models of graft-versus-host disease: advances and … › content › dmm › 4 › 3 › 318.full.pdf · PERSPECTIVE 318 dmm.biologists.org Introduction A major complication

dmm.biologists.org322

Mouse models of GvHDPERSPECTIVE

Ta

ble

1. M

ou

se m

od

els

of

aG

vH

D

Do

no

r st

rain

Re

cip

ien

t st

rain

Co

nd

itio

nin

g

reg

ime

nG

en

eti

cs

Ma

in T

-ce

ll t

yp

e

con

trib

uti

ng

to

ph

en

oty

pe

Ce

ll t

yp

e a

nd

do

seO

utc

om

eR

efe

ren

ce e

xa

mp

le(s

)

MH

C m

ism

atc

he

d

C5

7/B

l6 (

H2

b)

BA

LB/c

(H

2d)

90

0 c

Gy

Mis

ma

tch

ed

fo

r

MH

CI,

MH

CII

an

dm

iHA

s

CD

4+ a

nd

CD

8+

Sp

len

ic T

ce

lls (

0.5

-

21

06)

an

d T

CD

do

no

r B

M c

ells

Sys

tem

ic d

ise

ase

by

10

-

21

da

ys; l

eth

al

va

n L

ee

uw

en

et

al.,

20

02

C3

H/H

eJ

(H2

k )C

57

/Bl6

(H

2b)

90

0 c

Gy

Mis

ma

tch

ed

fo

r

MH

CI,

MH

CII

an

d

miH

As

CD

4+ a

nd

CD

8+

21

07 B

M c

ells

an

d 5

-

25

10

6 s

ple

en

ce

lls

or

0.1

-11

06 ly

mp

hn

od

e c

ells

Sys

tem

ic d

ise

ase

by

10

-

30

da

ys; l

eth

al

Bla

zar

et

al.,

19

91

C5

7/B

l6 (

H2

b)

B6

C3

F1

(H

2k/

b)

60

0-1

05

0 c

Gy

Mis

ma

tch

ed

fo

r

MH

CI,

MH

CII

an

d

miH

As

CD

4+ a

nd

CD

8+

70

10

6 s

ple

en

ce

lls

an

d 1

10

7 T

CD

BM

Sys

tem

ic d

ise

ase

; le

tha

lK

an

am

aru

et

al.,

19

84

;

Fo

wle

r e

t a

l., 1

99

6

C5

7/B

l6 (

H2

b)

B6

D2

F1

(H

2b

/d)

No

XR

T o

r 1

10

0-1

30

0

cGy

Mis

ma

tch

ed

fo

r

MH

C I,

MH

C II

an

d

miH

As

CD

4+ a

nd

CD

8+

Wh

ole

sp

len

ocy

tes:

1-

10

10

7 c

ells

if n

o

XR

T;

2-5

10

6 p

uri

fie

d T

cells

wit

h X

RT

Sys

tem

ic d

ise

ase

; le

tha

l

by

30

da

ys if

XR

T u

sed

;

sym

pto

ms

in 3

0-5

0d

ays

if n

o X

RT

Pic

kel a

nd

Ho

ffm

an

n, 1

97

7;

Via

et

al.,

19

96

a; K

ren

ge

r

et

al.,

20

00

a; H

ilde

bra

nd

te

t a

l., 2

00

4; B

lase

r e

t a

l.,

20

05

; Nic

ule

scu

et

al.,

20

05

C5

7/B

l6 (

H2

b)

B6

AF

1 (

H2

b/a

)N

o X

RT

Mis

ma

tch

ed

fo

rM

HC

I, M

HC

II a

nd

miH

As

CD

4+ a

nd

CD

8+

Wh

ole

lym

ph

oid

ce

lls(2

-61

07)

Sys

tem

ic d

ise

ase

; le

tha

ld

ep

en

din

g o

n T

-ce

ll

do

se

Gh

ayu

r e

t a

l., 1

98

8; N

est

el

et

al.,

19

92

; Ge

nd

elm

an

et

al.,

20

04

C5

7/B

l6 (

H2

b)

B1

0.B

R (

H2

k )7

50

-90

0 c

Gy

+/–

Cyt

oxa

n

12

0 m

g/k

g

Mis

ma

tch

ed

fo

r

MH

C I,

MH

C II

an

dm

iHA

s

CD

4+ a

nd

CD

8+

2.5

10

5 t

o 2

10

7 T

CD

BM

ce

lls a

nd

5-

25

10

6 s

ple

en

ce

lls

or

0.5

-11

06 ly

mp

hn

od

e c

ells

Sys

tem

ic d

ise

ase

; le

tha

l

de

pe

nd

ing

on

T-c

ell

do

se a

nd

so

urc

e;

sym

pto

ms

by

14

-28

da

ys

Va

llera

et

al.,

19

81

; Bla

zar

et

al.,

19

91

; Pa

no

ska

ltsi

s-M

ort

ari

et

al.,

19

98

C5

7/B

l6 (

H2

b)

B6

.C-H

2b

m1 (

bm

1)

(H2

b b

ack

gro

un

d w

ith

mu

tati

on

at

MH

C I)

95

0 c

Gy

or

sub

leth

al

XR

TM

HC

I m

ism

atc

hC

D8

+4

10

6 T

CD

BM

ce

lls

an

d 1

.5–

7.5

10

6

CD

8+ T

ce

lls

Sys

tem

ic d

ise

ase

if C

D8

+

T c

ells

infu

sed

wit

h

de

ath

by

30

-80

da

ys

Ro

link

et

al.,

19

83

; Sp

ren

t

et

al.,

19

86

C5

7/B

l6 (

H2

b)

B6

.C-H

2b

m1

2 (

bm

12

)

(H2

b b

ack

gro

un

d w

ith

mu

tati

on

at

MH

C II

)

95

0 c

Gy

or

sub

leth

al

XR

TM

HC

II m

ism

atc

hC

D4

+4

10

6 T

CD

BM

an

d 1

-

51

06 C

D4

+ T

ce

lls

Sys

tem

ic d

ise

ase

if C

D4

+

T c

ells

infu

sed

wit

hd

ea

th b

y 2

0-4

0 d

ays

Ro

link

et

al.,

19

83

; Sp

ren

t

et

al.,

19

86

miH

A m

ism

atc

he

d

B1

0.B

R (

H2

k )C

BA

or

BA

LB.K

(H

2k )

75

0 c

Gy

miH

A m

ism

atc

he

sC

D8

+8

10

6 T

CD

BM

ce

lls

an

d 1

10

7 w

ho

le

sple

no

cyte

s o

r5

10

6 C

D4

+ o

r C

D8

+

T c

ells

Sys

tem

ic d

ise

ase

;

sym

pto

ms

incl

ud

e t

ail

sca

ling

, ea

r e

rosi

on

s,d

iarr

he

a, h

un

chin

g

Ko

rng

old

an

d S

pre

nt,

19

87

;

Ko

rng

old

, 19

92

; Co

oke

et

al.,

19

96

; Ka

pla

n e

t a

l.,2

00

4; F

an

nin

g e

t a

l., 2

00

9

B1

0.D

2 (

H2

d)

DB

A/2

(H

2d)

80

0 c

Gy

miH

A m

ism

atc

he

sC

D4

+ (

min

or

con

trib

uti

on

by

CD

8+)

41

06 T

CD

BM

ce

lls

an

d 1

10

6 T

ce

lls, o

rp

uri

fie

d C

D4

+ o

r

CD

8+ T

ce

lls

Sys

tem

ic d

ise

ase

; le

tha

l

by

da

y 8

0

Ko

rng

old

an

d S

pre

nt,

19

87

;

Ko

rng

old

, 19

92

Ta

ble

1 c

on

tin

ue

d o

n n

ext

pa

ge

.

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Page 6: Mouse models of graft-versus-host disease: advances and … › content › dmm › 4 › 3 › 318.full.pdf · PERSPECTIVE 318 dmm.biologists.org Introduction A major complication

Disease Models & Mechanisms 323

Mouse models of GvHD PERSPECTIVE

Ta

ble

1.

Co

nti

nu

ed

Do

no

r st

rain

Re

cip

ien

t st

rain

Co

nd

itio

nin

g

reg

ime

nG

en

eti

cs

Ma

in T

-ce

ll t

yp

e

con

trib

uti

ng

to

ph

en

oty

pe

Ce

ll t

yp

e a

nd

do

seO

utc

om

eR

efe

ren

ce e

xa

mp

le(s

)

miH

A m

ism

atc

he

d

B1

0.D

2 (

H2

d)

BA

LB/c

(H

2d)

Su

b-l

eth

al 6

00

-75

0

cGy

or

Fra

ctio

na

ted

10

00

cGy

miH

A m

ism

atc

he

sC

D4

(m

ino

r

con

trib

uti

on

by

CD

8+)

11

07 B

M c

ells

an

d

11

08 w

ho

lesp

len

ocy

tes

Sys

tem

ic d

ise

ase

Ko

rng

old

an

d S

pre

nt,

19

87

;

Eyr

ich

et

al.,

20

05

B1

0 (

H2

b)

BA

LB.b

(H

2b)

77

5 c

Gy

miH

A m

ism

atc

he

sC

D4

+ (

wit

h s

om

e

con

trib

uti

on

fro

mC

D8

+)

81

06 T

CD

BM

ce

lls

an

d 1

07

un

fra

ctio

na

ted

sple

en

ce

lls o

r

1.4

10

6 C

D4

+ o

r6

10

5 C

D8

+ T

ce

lls

Sys

tem

ic d

ise

ase

; no

cuta

ne

ou

sin

vo

lve

me

nt

Ka

pla

n e

t a

l., 2

00

4

C5

7/B

l6 (

H2

b)

BA

LB.b

(H

2b)

85

0–

10

00

cG

ym

iHA

mis

ma

tch

es

CD

4+

51

06 T

CD

BM

ce

lls

an

d 2

10

6 T

ce

llsS

yste

mic

dis

ea

seB

erg

er

et

al.,

19

94

; Zh

an

g

et

al.,

20

05

DB

A/2

(H

2d)

B1

0.D

2 (

H2

d)

82

0 c

Gy

miH

A m

ism

atc

he

sC

D8

+4

10

6 T

CD

BM

ce

lls

an

d 1

10

6 T

ce

lls, o

rp

uri

fie

d C

D4

+ o

r

CD

8+ T

ce

lls

Min

ima

l sys

tem

ic d

ise

ase

wit

h w

ho

le s

ple

en

ce

llsb

ut

incr

ea

sed

wit

h

pu

rifi

ed

CD

8+ T

ce

lls;

up

to

50

% m

ort

alit

y a

t

da

y 2

5

Ko

rng

old

an

d S

pre

nt,

19

87

Xe

no

tra

nsp

lan

tati

on

mo

de

ls

Hu

ma

n P

BM

Cs

NO

D/S

CID

IL2

rγ-n

ull

(NS

G)

No

XR

T o

r

20

0-2

50

cG

y

Mis

tma

tch

ed

fo

r

MH

C I,

MH

C II

an

d

miH

As

CD

4+

11

07 o

r

5-1

01

06

De

ath

fro

m G

vH

D in

30

-

50

da

ys

Ito

et

al.,

20

09

; Kin

g e

t a

l.,

20

09

Hu

ma

n P

BM

Cs

BA

LB/c

A-R

AG

2–

/––

/– IL

2r

35

0 c

Gy

Mis

ma

tch

ed

fo

r

MH

C I,

MH

C II

an

d

miH

As

CD

4+ a

nd

CD

8+

5-3

01

06

Sys

tem

ic d

ise

ase

va

n R

ijn e

t a

l., 2

00

3

Hu

ma

n p

eri

ph

era

l

blo

od

T c

ells

:n

aiv

e o

r

CD

3/C

D2

8 b

ea

de

xpa

nd

ed

NO

D/S

CID

2

m-n

ull

25

0-3

00

cG

yM

ism

atc

he

d f

or

MH

C I,

MH

C II

an

dm

iHA

s

–1

10

7 h

um

an

T c

ells

RO

inje

ctio

nS

yste

mic

dis

ea

seN

erv

i et

al.,

20

07

BM

, bo

ne

ma

rro

w; c

Gy,

ce

nti

gra

y; R

O, r

etr

o-o

rbit

al i

nje

ctio

n; T

CD

, T-c

ell

de

ple

ted

; XR

T, r

ad

iati

on

co

nd

itio

nin

g.

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Page 7: Mouse models of graft-versus-host disease: advances and … › content › dmm › 4 › 3 › 318.full.pdf · PERSPECTIVE 318 dmm.biologists.org Introduction A major complication

dmm.biologists.org324

Mouse models of GvHDPERSPECTIVE

MHC-mismatched modelsThe most commonly studied MHC-mismatched model of aGvHDis C57/Bl6 (H2b) r BALB/c (H2d) (transplantation of cellularisolates from C57/Bl6 (H2b) donors into BALB/c (H2d) recipients).MHC-mismatched models of aGvHD also contain a number ofmiHA mismatches, although the impact of miHA mismatches onthe pathophysiology of the clinical phenotype observed is limited.All models incorporate myeloablative radiation conditioning eitheras a single dose or, when radiation doses of more than 1100 cGyare delivered, as a fractionated dose separated by 3-8 hours, whichdecreases gut toxicity. Most MHC-mismatched models aredependent on both CD8+ and CD4+ T cells. To dissect themechanisms by which these T-cell subsets induce aGvHD,transgenic mice that have a mutant MHC I (which is involved inCD8+ T-cell activation) [B6.C-H2bm1 (bm1)] or MHC II (which isinvolved in CD4+ T-cell activation) [B6.C-H2bm12 (bm12)] havebeen used. The H2bm1 and H2bm12 transgenic mouse models havebeen important in dissecting the interaction of T cells withrecipient and donor APCs (Sprent et al., 1990). CD8+ T cells induceaGvHD by engagement of the TCR and release of perforin,granzymes and Fas ligand (Via et al., 1996b; Graubert et al., 1997;Maeda et al., 2005). Only recipient APCs can stimulate CD8+ Tcells in CD8+ T-cell-dependent models (Shlomchik et al., 1999).Conversely, either recipient or donor APCs can stimulate CD4+

T-cell responses (Anderson et al., 2005). TNF mediates thecytotoxic effects of alloreactive CD4+ T cells. The target of thesecytotoxic T-cell responses is recipient epithelium, although theepithelium is not required to present antigen to T cells (Teshimaet al., 2002).

Another MHC-mismatched model of aGvHD is the ‘parent-to-F1’ model. Parent-to-F1 transplants have been studied using eitherlethal irradiation regimens or no irradiation conditioning. The lattermodel allows the study of aGvHD without the effects of radiation,thus mimicking the situation in humans that undergo non-myeloablative or ‘reduced intensity conditioning’ regimens. Thecells transplanted into these models are either whole splenocytes(in the non-irradiated models) or purified T cells with T-cell-depleted bone marrow (in the irradiated models). The developmentof aGvHD in these models depends on differences in class I andclass II MHC antigens, and involves the effects of CD4+ and CD8+

T cells (Hakim et al., 1991). The characteristics of aGvHD differin the irradiated and non-irradiated parent-to-F1 model: whereasthe irradiated model exhibits weight loss and skin problems, thenon-irradiated model has relatively limited skin pathology orweight loss, and aGvHD mainly manifests as immune deficiencyand mixed chimerism, with lower mortality. Importantly, not allparental donor strains cause aGvHD. Some can cause cGvHD (e.g.DBA/2 r B6D2F1), possibly because of the natural bias of somestrains to mount either a Th1- or Th2-cell response (De Wit et al.,1993; Nikolic et al., 2000; Tawara et al., 2008).

miHA-mismatched modelsmiHA-mismatched models of aGvHD represent human HSCTmore closely than MHC-mismatched models, because MHC-mismatched transplants in humans are not commonly performed.Similar to the human setting, miHA-mismatched models exhibitless morbidity than MHC-mismatched models, but still result inlethal aGvHD. An example of a miHA-mismatched transplant

model is B10.D2 (H2d) r BALB/c or DBA/2 (H2d). The aGvHDthat occurs in this model is primarily dependent on CD4+ T cellsand less on CD8+ T cells (Korngold and Sprent, 1987). However,transplants between congenic strains illustrate that differences inMHC (which means that alloantigens are presented differently invivo) influence the type and specificity of the T-cell responseunderlying the GvHD phenotype (Kaplan et al., 2004). For example,in other miHA-mismatched transplant models, such as C3H.SWr Bl6 (H2b) and DBA/2 r B10.D2 (H2d), GvHD depends primarilyon CD8+ T cells.

Xenogeneic transplant modelsThese models have been developed to generate a system wherebyhuman T-cell-mediated aGvHD can be studied and manipulatedin vivo. However, transplanting across species (xenotransplantation)requires significant immunosuppression to prevent graft rejection,because the immune response to xenografts is typically much morerobust than to allografts. Initial attempts to xenotransplant humancells into mice were conducted using non-obese diabetic severecombined immunodeficiency (NOD/SCID) mice and resulted in a1-20% rate of engraftment (Mosier et al., 1988; Hoffmann-Fezer etal., 1993; Christianson et al., 1997). Low engraftment was due tothe fact that NOD/SCID mice lack functional T and B cells butretain NK cell function and can therefore respond to foreignantigens. Subsequently, better engraftment of xenotransplantedcells was obtained using RAG2-deficient and IL-2-receptor--deficient mice (BALB/cA-RAG2–/– IL2r–/–), which lack functionalT, B and NK cells (van Rijn et al., 2003). Our group has used theNOD/SCID 2-microglobulin-null mouse (Christianson et al.,1997; Nervi et al., 2007) to image the trafficking of xenotransplantedhuman T cells in vivo. Using this system, we demonstrated thatretro-orbital injection of human T cells into sublethally irradiatedNOD/SCID 2m-null recipients resulted in consistent engraftmentand expansion of human T cells, the trafficking of T cells to targetorgans including the lung, skin, liver and kidney, and clinicalsymptoms consistent with severe aGvHD. T cells could be detected1 hour after retro-orbital injection and trafficked to secondarylymphoid organs, resulting in lethal GvHD; by contrast, intravenousinjection of T cells caused them to traffic first to the lungs, andwas associated with the development of less severe aGvHD andless morbidity and mortality (Nervi et al., 2007).

The most permissive mouse model for human stem-cell and T-cell engraftment was derived by back-crossing mice carrying ahomozygous deletion of the common -chain of the IL-2 receptoronto a NOD/SCID background [referred to as NOD/SCID IL2r-null (NSG) mice]. NSG mice lack T, B and NK cells, and also havereduced function of macrophages and dendritic cells.Transplantation of human peripheral blood mononuclear cells(PBMCs) causes an aGvHD-like syndrome that results in death by20-50 days, and is manifested by overt human T-cell infiltration ofmouse skin, liver, intestine, lungs and kidneys (Ito et al., 2009).Xenogeneic transplants of human PBMCs into immunodeficientmice require human APCs to process mouse antigens and presentthem in the presence of class II MHC. Because T-cell recognitionof MHC molecules is restricted by species, human TCRs do notrecognize mouse MHC, making this a primarily CD4+ T-cell-dependent model, at least in the case of NOD/SCID (Lucas et al.,1990).

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Page 8: Mouse models of graft-versus-host disease: advances and … › content › dmm › 4 › 3 › 318.full.pdf · PERSPECTIVE 318 dmm.biologists.org Introduction A major complication

Disease Models & Mechanisms 325

Mouse models of GvHD PERSPECTIVE

Antigen-specific transgenic TCR modelsA major limitation of all MHC- and miHA-mismatched transplantmodels of aGvHD is that it is difficult to determine the specificalloantigen that the responding T cells recognize. In addition, thecytokine storm induced by conditioning can lead to antigen-independent bystander activation of T cells. One solution to thisproblem has been to use mouse strains that have T cells expressingTCRs of defined specificity (TCR-transgenic mice); these strainswere generated initially to study thymic selection, but have morerecently been useful for understanding T-cell alloresponses inGvHD. The specificity of the T cells in a TCR-transgenic mouseis generally restricted to a single peptide epitope that is eitherconstitutively expressed by the transplanted allogeneic APCs oris administered as an exogenous antigen to stimulate a T-cellresponse. Examples of TCR-transgenic mice include H-Y, TEa,2C, TS1, D011.10 and OT-II (Kisielow et al., 1988; Sha et al., 1988;Murphy et al., 1990; Grubin et al., 1997; Barnden et al., 1998;Wilkinson et al., 2009). These mice have been used to investigateseveral aspects of GvHD, including the role of antigen affinity(Yu et al., 2006), antigen-induced T-cell activation andproliferation, TReg cell function (Albert et al., 2005; Damdinsurenet al., 2010) and, more recently, cross presentation of antigens(Wang et al., 2009).

Mouse models of cGvHDAlthough the clinical phenotype and kinetics of cGvHD are distinctfrom aGvHD, with fibrosis and chronic inflammationpredominating in a manner resembling autoimmune disease, mostof the same organs are affected. There are five factors that cancontribute to the pathology of cGvHD in humans and mousemodels after HSCT: cytokine-mediated fibrosis, defective negativeselection of T cells, deficiency of TReg cells, genetic polymorphismsand dysregulated B-cell responses. However, no mouse modeldeveloped thus far recapitulates all of these characteristics or exactlymimics what is seen in human cGvHD.

Current mouse models of cGvHD (summarized in Table 2) canbe broadly divided into sclerodermatous (pro-fibrotic) models,autoantibody-mediated (lupus-like) models and a more recentlyreported model in which thymic function is defective (Sakoda etal., 2007; Chu and Gress, 2008). The distinctions in the phenotypesof models of cGvHD and aGvHD result from differences in thefactors underlying the pathogenesis of each model.

Sclerodermatous modelsSclerodermatous (pro-fibrotic) cGvHD models are characterizedby fibrotic changes in the dermis, which can involve the lung, liverand salivary glands (Jaffee and Claman, 1983; Claman et al., 1985;Howell et al., 1989; McCormick et al., 1999; Levy et al., 2000).Transplantation results in full donor chimerism, and the resultingphenotype resembles scleroderma in humans, which occurs in upto 15% of patients with cGvHD (Skert et al., 2006). However, fibrosisin the mouse models begins within 30 days of transplantation,which is atypical of human cGvHD, which occurs months to yearsafter transplantation.

These models are MHC matched but differ in numerous miHAs.The pathology is dependent on CD4+ T cells that release Th2cytokines, which can stimulate other cells to release fibrosingcytokines (such as IL-13 and TGF) (for a review, see Wynn, 2004),

resulting in the sclerodermatous changes (Hamilton, 1987;Korngold and Sprent, 1987). In addition, other cellular mediatorshave also been implicated: macrophages are responsible for TGFproduction, whereas other cell types such as eosinophils and mastcells have been implicated in the pathogenesis of fibrosis in targetorgans. Mediators secreted by eosinophils and mast cells can causefibroblast proliferation and collagen production (Levi-Schaffer andWeg, 1997). Two examples of sclerodermatous models are B10.D2r BALB/c and LP/J r Bl6, both of which require total bodyirradiation conditioning (Jaffee and Claman, 1983; DeClerck et al.,1986). The dose of radiation can also affect the severity of disease(Jaffee and Claman, 1983).

The sclerodermatous cGvHD models provide importantinsights into the pathogenesis of fibrosis that occurs in cGvHD;however, there are a number of limitations of these models. First,the fibrosis in human cGvHD can be systemic or pleiotropic,unlike mouse models, which mainly involve the skin, suggestingother mechanisms might be at play in the development of systemicsclerosis. Second, the time course of development of skin changesis less rapid in humans than in mouse models. Finally, these mousemodels fail to reproduce the full clinical spectrum seen inhumans, which can involve target organs including the intestine,lungs, liver, salivary glands, eyes, oral mucosa, muscles, tendonsand nerves.

Autoantibody-mediated modelsAutoantibody-mediated (lupus-like) cGvHD models, which exhibita pathology similar to that of lupus, are manifested by nephritis,biliary cirrhosis, salivary gland fibrosis, lymphadenopathy,splenomegaly, autoantibody production and, to a lesser extent, skinpathology. These models are MHC mismatched and classicallyinvolve parent-to-F1 transplants that result in mixed chimerism.The most commonly studied autoantibody model is the DBA/2 rB6D2F1 model. This model results in expansion of recipient B cells,leading to lymphadenopathy, splenomegally and autoantibodyproduction. Both MHC I and II are mismatched in these models,but the pathology is dependent on donor CD4+ T cells (Rolink andGleichmann, 1983; Rolink et al., 1983; Hakim et al., 1991) and isassociated with the production of Th2 cytokines. Manipulating thecytokine balance by skewing the CD4+ T-cell response to a Th1phenotype, by exposure to IL-12, causes a shift to a more aGvHD-like phenotype (Via et al., 1994). Importantly, the choice of parentaldonor determines the phenotypic outcome. For example, if C57/Bl6is the T-cell donor for a B6D2F1 recipient, an aGvHD phenotyperesults. This might be due to naturally high proportions of cytotoxicCD8+ T cells in the C57/Bl6 mouse strain (Via et al., 1987).

Unlike models of aGvHD, which depend primarily on donor Tcells, pathology in autoantibody models of cGvHD depends onrecipient B cells (Morris et al., 1990a). Specifically, the recipient Bcells interact with donor T cells (Morris et al., 1990b) via cell-surfaceMHC and are also stimulated by cytokines, including IL-4 and IL-10 (De Wit et al., 1993; Durie et al., 1994; Via et al., 1996a; Kim etal., 2008). Recipient B cells act as potent APCs that stimulate donorT cells, perpetuating the cycle that leads to autoantibodyproduction.

Mouse autoantibody cGvHD models are similar to cGvHD thatoccurs in humans in that they also involve the production ofautoantibodies, although there are a number of distinctions. First,

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Page 9: Mouse models of graft-versus-host disease: advances and … › content › dmm › 4 › 3 › 318.full.pdf · PERSPECTIVE 318 dmm.biologists.org Introduction A major complication

dmm.biologists.org326

Mouse models of GvHDPERSPECTIVE

Ta

ble

2.

Mo

use

mo

de

ls o

f cG

VH

D

Do

no

r

stra

inR

eci

pie

nt

stra

in

Co

nd

itio

nin

g

reg

ime

nG

en

eti

cs

Ma

in T

-ce

ll t

yp

e

con

trib

uti

ng

to

ph

en

oty

pe

Ce

lls

an

d d

ose

Ou

tco

me

Re

fere

nce

ex

am

ple

(s)

Scl

ero

de

rma

(p

ro-fi

bro

tic

mo

de

ls)

B1

0.D

2

(H2

d)

BA

LB/c

(H

2d)

70

0-9

00

cG

ym

iHA

mis

ma

tch

ed

CD

4+

TC

D B

M c

ells

an

d 1

10

7 w

ho

le

sple

no

cyte

s o

r p

uri

fie

d T

ce

lls

Scl

ero

de

rma

; pro

gre

ssiv

e

fib

rosi

s o

f o

rga

ns

Ko

rng

old

an

d S

pre

nt,

19

78

; Ja

ffe

e

an

d C

lam

an

, 19

83

; Cla

ma

n e

t a

l.,1

98

5; H

am

ilto

n, 1

98

7; K

orn

go

ld

an

d S

pre

nt,

19

87

; McC

orm

ick

et

al.,

19

99

; Le

vy

et

al.,

20

00

; Ka

pla

n

et

al.,

20

04

; Zh

ou

et

al.,

20

07

LP/J

(H

2b)

C5

7/B

l6 (

H2

b)

90

0-1

10

0 c

Gy

Mis

ma

tch

ed

fo

r

miH

As

an

d M

HC

ND

20

-50

10

6 s

ple

en

ce

lls a

nd

51

07

BM

Scl

ero

de

rma

occ

urr

ing

by

da

y 2

8

Ha

milt

on

an

d P

ark

ma

n, 1

98

3;

De

Cle

rck

et

al.,

19

86

B1

0.D

2

(H2

d)

BA

LB/c

Ra

g2

–/–

(H

2d

d)

No

ne

H2

ma

tch

ed

bu

t

miH

Am

ism

atc

he

d

ND

Wh

ole

sp

lee

n c

ells

(2-5

10

7)

Scl

ero

de

rma

; pro

gre

ssiv

e

fib

rosi

s o

f o

rga

ns;

au

toa

nti

bo

die

s

Ru

zek

et

al.,

20

04

Au

toa

nti

bo

dy

-me

dia

ted

(lu

pu

s-li

ke

) m

od

els

DB

A/2

(H

2d)

B6

D2

F1

(H

2b

/d)

No co

nd

itio

nin

g

Mis

ma

tch

ed

fo

r

MH

C I,

MH

C II

an

d m

iHA

s

CD

4+

1-1

01

07

Lup

us

Sjo

rgre

n’s

(S

ialo

ad

en

itis

);

au

toa

nti

bo

die

s

Via

et

al.,

19

87

; Fu

jiwa

ra e

t a

l., 1

99

1;

De

Wit

et

al.,

19

93

; Via

et

al.,

19

96

a; S

layb

ack

et

al.,

20

00

; Kim

et

al.,

20

06

; Kim

et

al.,

20

08

BA

LB/c

(H

2d)

(BA

LB/c

xA)F

1 (

H2

d/a

)N

o con

dit

ion

ing

Mis

ma

tch

ed

fo

rM

HC

I, M

HC

II

an

d m

iHA

s

CD

4+

8-1

21

07 s

ple

en

ce

llsP

oly

art

hri

tis,

scl

ero

de

rma

,g

lom

eru

lon

ep

hri

tis,

Sjo

rgre

n’s

, scl

oro

sin

gch

ola

ng

itis

Pa

ls e

t a

l., 1

98

5; V

ida

l et

al.,

19

96

C5

7/B

l6 (

H2

b)

(B6

xBA

LB/c

)F1

(H2

b/d

)N

o con

dit

ion

ing

Mis

ma

tch

ed

fo

rM

HC

I, M

HC

II

an

d m

iHA

s

CD

4+

61

07 w

ho

le s

ple

en

ce

llsA

cute

pro

gre

ssin

g t

och

ron

ic G

vH

DT

sch

ett

er

et

al.,

20

00

DB

A/2

(H

2d)

BA

LB/c

(H

2d)

65

0 c

Gy

miH

A m

ism

atc

he

s–

51

07 w

ho

le s

ple

en

ce

llsA

uto

an

tib

od

ies;

ski

nsc

lero

de

rma

Zh

an

g e

t a

l., 2

00

6

C5

7/B

l6 (

H2

b)

(C5

7/B

l6

B6

.C-

H2

bm

1)F

1 (

H2

b

ba

ckg

rou

nd

wit

h

mu

tati

on

at

MH

CI)

No co

nd

itio

nin

g

MH

C I

mis

ma

tch

ed

CD

8+

51

07 s

ple

en

ce

lls o

r 1

08 s

ple

en

(2/3

) a

nd

lym

ph

no

de

(1

/3)

cells

;

2 d

ose

s o

f ce

lls o

n d

ay

0 a

nd

7

Glo

me

rulo

ne

ph

riti

s; n

on

-

leth

al i

n u

nir

rad

iate

dm

ice

; le

tha

l in

irra

dia

ted

mic

e a

t 3

0-6

0 d

ays

Ro

link

et

al.,

19

83

; Ito

et

al.,

19

92

C5

7/B

l6 (

H2

b)

(C5

7/B

l6

B6

.C-

H2

bm

12)F

1

(H2

b b

ack

gro

un

dw

ith

mu

tati

on

at

MH

C II

)

No

ne

or

90

0

cGy

MH

C II

mis

ma

tch

ed

CD

4+

No

co

nd

itio

nin

g:

4-5

10

6 s

ple

en

CD

4+ T

ce

lls, 2

-

31

06 B

M c

ells

;

2 d

ose

s o

f ce

lls o

n d

ay

0 a

nd

7

Co

nd

itio

ne

d:

4-5

10

6 C

D4

+ T

ce

lls a

nd

1-2

10

6

TC

D B

M c

ells

Sys

tem

ic d

ise

ase

an

d d

ea

th

by

20

-40

da

ys

Ro

link

et

al.,

19

83

; Ito

et

al.,

19

92

;

Bro

wn

et

al.,

20

02

Th

ym

ic d

ysf

un

ctio

n m

od

el

Bl6

-H2

-Ab

l–/–

(H2

b)

C3

H/H

eN

(H

2k )

13

00

cG

y sp

lit

in t

wo

fra

ctio

ns

Mis

ma

tch

ed

fo

r

MH

C I,

MH

C II

an

d m

iHA

s

BM

-de

riv

ed

CD

4+

an

d C

D8

+

51

06 T

CD

BM

ce

llsE

pid

erm

al a

tro

ph

y, f

at

loss

,

folli

cula

r d

rop

ou

t a

nd

de

rma

l th

icke

nin

g; b

iled

uct

loss

an

d p

eri

po

rta

l

fib

rosi

s in

liv

er;

sa

liva

ryg

lan

d fi

bro

sis

an

d

atr

op

hy;

cyt

op

en

ias

Sa

kod

a e

t a

l., 2

00

7

Ab

bre

via

tio

ns

as

for

Ta

ble

1, p

lus:

B6

, C5

7/B

l6; B

6D

2F

1, (

C5

7/B

l6 x

DB

A/2

)F1

; ND

, no

t d

ete

rmin

ed

.

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Page 10: Mouse models of graft-versus-host disease: advances and … › content › dmm › 4 › 3 › 318.full.pdf · PERSPECTIVE 318 dmm.biologists.org Introduction A major complication

Disease Models & Mechanisms 327

Mouse models of GvHD PERSPECTIVE

nephritis in cGvHD in humans is rare. Second, the repertoire ofautoantibodies associated with cGvHD is more diverse in humansthan in mice. Finally, lymphadenopathy and splenomegaly do notoccur in the human setting.

A cGvHD model involving both autoantibodies andsclerodermaTo more accurately model cGvHD that occurs in humans, attemptshave been made to combine the features of autoantibody-mediatedand scleroderma models in a single mouse model. For example,transfer of splenocytes from DBA/2 (H2d) mice into sublethallyirradiated BALB/c (H2d) recipients results in a lupus-like andsclerodermatous phenotype (Zhang et al., 2006). These micedevelop proteinuria with basement membrane immune complexdeposits, and skin fibrosis with collagen I deposition; the severityof these disorders depends on the number of donor cellstransplanted. In addition, these mice have increased levels ofautoantibodies specific for double-stranded DNA. In this case,donor CD4+ T cells are required for the pathology, whereas donorCD8+ T cells do not contribute. Unlike pathology in the parent-to-F1 autoantibody-mediated model, which involves recipient B cells,pathology in this model involves donor B cells. Finally, the transferof TReg cells rescues the cGvHD phenotype, indicating that TRegcells are functional in treating cGvHD, as has been demonstratedin aGvHD models. In summary, this model indicates theimportance of donor CD4+ T cells as well as B cells (from bothdonor and recipient) in the development of cGvHD. However, itdoes not support a role for the thymus in the pathophysiology ofcGvHD, in contrast to the model discussed below.

Defective thymic function modelGiven the similarity between the presentation of cGvHD and someautoimmune diseases, as well as the fact that thymectomy of 3-day-old mice results in symptoms similar to some aspects of cGvHD(Nishizuka and Sakakura, 1969; Bonomo et al., 1995), it has longbeen hypothesized that defects in thymic negative selection mighthave a role in the pathogenesis of cGvHD. The thymus is a crucialtarget of aGvHD (Fukushi et al., 1990; Krenger et al., 2000b; Hauri-Hohl et al., 2007), and it is known that aGvHD results in destructionof the normal thymic architecture (Ghayur et al., 1988), especiallyHassall’s corpuscles, which have been shown to be crucial for thedevelopment of TReg cells (Watanabe et al., 2005). The presentationof tissue-restricted antigens by thymic APCs and epithelial cellscould result in their direct attack by cytotoxic donor T cells.Cytotoxic attack of these cell types, which are crucial for negativeselection, could result in impaired central tolerance and subsequentautoreactivity (Krenger and Hollander, 2008; Mathis and Benoist,2009).

A potential role for thymic damage in GvHD was initiallyproposed by a group that demonstrated dysfunctional negativeselection of autoreactive T-cell clones in mice with aGvHD (vanden Brink et al., 2000). More recently, a transgenic mouse transplantmodel was developed: T-cell-depleted bone marrow from MHC-II-deficient mice on a C57/Bl6 background [H2-Ab1–/– (H2b)] weretransferred to lethally irradiated C3H/HeN (H2k) recipients.Recipient mice developed a phenotype similar to clinical cGvHD,with skin fibrosis characterized by epidermal atrophy, fat loss,follicular drop out and dermal thickening; bile duct loss and

periportal fibrosis in the liver; salivary gland fibrosis and atrophy;and cytopenias (Sakoda et al., 2007). This phenotype developedwithin 6 weeks of donor cell transfer, and fewer than 20% of animalslived for more than 100 days. Importantly, thymectomized recipientmice abrogated the phenotype, highlighting the role of the thymusin the pathology seen in this model. Experiments involving chimericmice showed that donor APCs were essential for the developmentof the cGvHD phenotype in this model: donor APCs that traffickedto the thymus after transplantation were principally responsiblefor negative selection. In a series of adoptive transfer studies, it wasshown that CD4+ T cells isolated from C3H/HeN recipients ofMHC-II-deficient bone marrow that develop cGvHD causedaGvHD when transferred to the C57/Bl6 donor strain, but causedcGvHD when transferred to established chimeric mice created froma C57/Bl6 r C3H/HeN transfer. This implies that donor APCs areessential for the development of the cGvHD phenotype not onlythrough their role in negative selection but through peripheralantigen presentation and T-cell stimulation.

Although the mouse model described above (H2-Ab1–/– r

C3H/HeN) is the first that reproduces multiple clinical features ofhuman cGvHD, it might not be clinically relevant. The modelcannot rule out the possibility that additional insult to the thymuscaused by GvHD might be the cause of dysfunctional negativeselection. It is also difficult to determine the exact kinetics ofcGvHD in this model, and whether there is a critical period ofthymic damage that is necessary to cause the resulting phenotype.Nonetheless, this model suggests that a crucial component ofcGvHD is dysfunctional thymic selection, which may or may notbe caused by donor T-cell-induced damage of the recipient thymusas a consequence of aGvHD.

Linking acute and chronic GvHD in a mouse modelThe high incidence of cGvHD that occurs following aGvHD inhumans suggests that there is a direct causal link between thesetwo disease states. If the mechanisms underlying this link could beidentified, targeted interventions could be used to prevent thedevelopment of autoimmune-like responses that characterizecGvHD from the alloresponses that underlie aGvHD.

Zhang et al. attempted to link acute and chronic GvHD in amouse model (Zhang et al., 2007). Donor bone-marrow-derivedCD4+ T cells that arose from hematopoietic precursors followingtransplantation and were selected (educated) in the recipientthymus during the development of aGvHD could mediate a cGvHDphenotype when transferred to secondary allogeneic recipients,resulting in cGvHD affecting the skin, liver and gut. Whentransferred to syngeneic secondary recipients, the same cellsresulted in a lethal aGvHD phenotype, suggesting that appropriatepositive selection had taken place in the recipient thymus but thatthymic negative selection was absent or dysfunctional. Thisillustrates a possible link between the cytotoxic CD8+ T-cellresponses of aGvHD and the CD4+ T-cell-mediated responsesdemonstrated in these models of cGvHD. The exact mechanism isunknown, but these data suggest that the thymus-educated donorCD4+ T cells that should be self-tolerant are autoreactive, suggestingthat the link between aGvHD and cGvHD is related to the targetingto the thymus by alloreactive T cells (both CD4+ and CD8+) andsubsequent elimination or alteration of the function of thymiccellular elements that are essential for appropriate negative

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Page 11: Mouse models of graft-versus-host disease: advances and … › content › dmm › 4 › 3 › 318.full.pdf · PERSPECTIVE 318 dmm.biologists.org Introduction A major complication

dmm.biologists.org328

Mouse models of GvHDPERSPECTIVE

selection. The effects of these autoreactive T cells can only bedemonstrated by adoptive transfer experiments because of thehighly lethal nature of aGvHD models.

Another attempt to investigate the link between aGvHD andcGvHD was performed using an F1 donor and F1 recipient [(B10� B10.D2)F1 (H2b/d) r (BALB.b � BALB/c)F1 (H2b/d)] transplant:this resulted in both systemic symptoms of aGvHD and skinmanifestations of cGvHD (Kaplan et al., 2004). In this model, MHCwas matched between donor and recipient, but miHAs weremismatched. This same group investigated transplantation betweenmultiple strain combinations matched at MHC loci, anddemonstrated that certain MHC haplotypes were associated withthe development of GvHD involving specific target organs, whichthey suspected was due to differences in MHC affinity for antigensfound in certain target tissues. For example, immunodominantantigens in the skin might be bound with high affinity in only certainMHC strains. This illustrates the important role of MHC inselecting immunodominant antigens, and how this might influencecGvHD.

A final example of the link between alloreactive T-cell responsesof aGvHD and the autoimmune responses of cGvHD involved theuse of immunodeficient mice (Tivol et al., 2005; Chen et al., 2007).In this case, aGvHD was initiated through a C57/Bl6 r BALB/clethally irradiated MHC-mismatched transplant. After 20 days,recipient animals were sacrificed and their splenic T cells wereisolated. Transfer of these isolated T cells into unconditionedimmunodeficient (Rag–/–) C57/Bl6 recipients (i.e. the samebackground as the original donor) resulted in inflammatoryinfiltration of the colon and liver (Chen et al., 2007). Thisautoimmune phenotype was dependent on CD4+ T cells (with aTh1 and Th17 cytokine profile) and could be rescued by theinfusion of immunosuppressive donor TReg cells. This supports thedata of Zhang et al. described above that a CD4+ T cell – derivedeither centrally (through the thymus) or peripherally (from theinitial donor T-cell inoculum) – in the context of aGvHD canstimulate the autoimmune responses observed in cGvHD. In fact,recent evidence suggests that mature donor-derived CD4+ T cellshave the ability to cause both alloreactive and autoreactiveresponses (Zhao et al., 2010). In a series of adoptive transferexperiments using the DBA/2 r BALB/c cGvHD model, Zhao etal. characterized the CD4+ T cells down to single cells, and showedthat a specific TCR predominated in mice with cGvHD. These Tcells recognized antigen in the presence of either allogeneic orautologous dendritic cells.

Considerations when selecting a mouse model of GvHDWith the large number of GvHD models now available, it can bedifficult to determine which model is appropriate for the researchquestion at hand. We offer in this section a few suggestions.

(1) First, decide whether you are interested in studying acute orchronic GvHD. It is of course useful to first establish from publishedliterature which models have been successfully validated for testinga given intervention.

(2) Next, determine what mechanism or T-cell population youwant to be dominant in your model for testing a specific therapeuticintervention. For example, a drug that targets CD8+ T cells wouldnot be effective in a model in which CD4+ T cells mediatepathology. Conversely, if limiting the degree of cytokine storm

produced from conditioning is important, then miHA-mismatchedor TCR-transgenic models would be useful, because conditioningcan be reduced or eliminated.

(3) Next, consider measurement outcomes such as: severity, coatcolor and the ability to track specific cell populations. If easy clinicalscoring of GvHD severity and survival are the main outcomesmeasured, then a model that has consistent rapid kinetics and 100%penetrance is desirable. In this case, the C57/Bl6 r BALB/ctransplant model could be a good choice. In general, the less thedegree of MHC mismatch, the less severe the model. Ifbioluminescence imaging (BLI) will be used (see Box 2), considerthe coat color of the mouse that will be imaged, because imagingis more difficult with mice that have dark coats (which requireshaving prior to BLI). If the tracking of donor cells in the recipientis necessary, it is possible to use flow cytometry to track MHCdisparity, to track fluorescent donor cells using BLI, or to usecongenic mice that have allelic differences at the CD45 locus (e.g.CD45.1 or CD45.2) to track cellular subsets from donor versusrecipient when they have a common MHC.

(4) It is also necessary to consider more complex issues such asthe production of transgenic strains. Sophisticated modeling canallow for elegant mechanistic studies, but if you are going to use atransgenic strain as a donor or recipient, extensive backcrossing toa single background is essential to eliminate significant geneticdisparity that could affect results.

(5) Finally, it is crucial to personally validate all new models inyour lab. Even a basic model (such as Bl6 r BALB/c) and certainlyany novel strain combinations require careful validation toinvestigate the influence of various factors – including pre-conditioning radiation dose, mouse strain and cleanliness ofcolonies (which varies between labs) – on, for example, diseaseincidence, severity and pathologic evidence of GvHD.

When working with any mouse model of GvHD, there are anumber of caveats to be aware of when interpreting and translatingexperimental data, which are outlined in Box 3.

Measuring the GVL effect in GvHD modelsThe graft-versus-leukemia (GVL) effect, also referred to as graft-versus-tumor effect, describes the alloimmune responses of donorimmune cells to residual leukemia or tumor, which are indicationsfor which HSCT is frequently performed as a treatment. Althougha complete review of models of GVL is beyond the scope of thisarticle [the reader is instead referred to reviews on the mechanisms

Box 2. Imaging pathological events during GvHDBioluminescent imaging (BLI) has become an important tool for trackingtransplanted cells in vivo. This technique has been reviewed recently (Negrinand Contag, 2006). Briefly, this technique uses donor mice that are transgenicfor a luciferase-GFP (green fluorescent protein) fusion protein, which is underthe control of the constitutively active chicken -actin promoter andcytomegalovirus enhancer [L2G85 mice (FVB background, H2q)]. The activity ofthe luciferase enzyme results in light emission that can be detected in vivo inrecipient mice using a specialized device. This technique is sensitive forvisualizing 100-1000 cells per region. It has been used to study the in vivotrafficking and proliferation of T cells (Beilhack et al., 2005), TReg cells (Edingeret al., 2003) and other T-cell subsets, including Th17 cells (Iclozan et al., 2009).It can also be used to measure the GVL effect by tracking the expansion orelimination of luciferase-expressing tumor cells over time after HSCT(Nishimura et al., 2008).

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Page 12: Mouse models of graft-versus-host disease: advances and … › content › dmm › 4 › 3 › 318.full.pdf · PERSPECTIVE 318 dmm.biologists.org Introduction A major complication

Disease Models & Mechanisms 329

Mouse models of GvHD PERSPECTIVE

of GVL (Bleakley and Riddell, 2004; Kolb et al., 2004) as well a recenthistorical perspective on GVL (Kolb, 2008)], it is worth mentioningthat separating GvHD from the GVL effect is a long-standingdilemma in transplantation, and is still an area of activeinvestigation. In fact, separating the immunopathological aspectsof GvHD from the curative effect of GVL is perhaps the mostclinically meaningful endpoint of HSCT. However, this is acomplicated issue because T cells mediate both GvHD and GVL.

Research in this area has focused on identifying the mechanismsthat might differentiate cytotoxic T-cell responses in GvHD fromcytotoxic T-cell responses in GVL, as well as identifying accessorycells that could block GvHD while allowing or augmenting GVL.The first evidence for GVL came from mouse transplant models(Barnes and Loutit, 1957; Weiss et al., 1983). Initial studies in the1950s demonstrated that a mouse leukemia could not be eliminatedby irradiation or by irradiation and syngeneic bone marrow, butirradiation plus allogeneic bone marrow prolonged survival of miceand eliminated leukemia from detection. However, transplantedmice developed a wasting syndrome that later became known asGvHD. Later efforts in the early 1980s were better able to definethe immunological basis for this GVL effect, and subsequent workdefined the importance of T cells and miHAs, as well as of innateimmune cells, such as NK cells, on the GVL effect. The modelsdiscussed above can also be used to test GVL effects of allogeneiccellular therapies, as well as to test treatments that are designed toprevent GvHD while preserving the GVL effect.

It could be argued that any novel therapy designed in thesemodels to target GvHD should be assessed for its effect on GVLbecause of the clinical implications for translation to humans. Totest the GVL effect, a labeled congenic tumor cell line (i.e.genetically compatible with the recipient, such as the A20 cell linefor a BALB/c background) or primary tumor cells [such as mouseacute promyelocytic leukemia cells from PML/RAR transgenicmice (Bl6/129 background) (Westervelt et al., 2003)] can betransduced to stably express a fluorescent marker or luciferase forbioluminescent studies. After a lethal injection of tumor cells atthe time of allogeneic transplantation, mice that develop aGvHDgenerally eliminate the genetically compatible tumor, owing to thealloreactive response mounted by donor T cells. The ultimate goalof interventions has been to prevent GvHD while preserving GVL.There have been a number of recent articles demonstrating this(Liang et al., 2008; Nishimura et al., 2008; Zheng et al., 2009). Theavailability of BLI (Box 2) has improved the capacity to detectresidual leukemia after transplantation and is used as a measureof response to treatments, together with survival and GvHDscoring, in these models.

ConclusionMouse models of GvHD are crucial for advancing ourunderstanding of the disease in its acute and chronic forms. As ourunderstanding grows, it is hoped that targeted therapies will bedeveloped to treat GvHD and preserve GVL. Recent advances intreatment that have evolved owing to studies of mouse models ofaGvHD include targeting mediators of the cytokine storm, such asTNF and IL-1, with antagonists (Uberti et al., 2005; Alousi et al.,2009; Antin et al., 2002), as well as reducing the intensity ofconditioning regimens to avoid excess inflammation. Other novelinterventions include the proteasome inhibitor bortezomib (Sun

Box 3. Caveats and considerations when interpretingfindings from mouse models of GvHD and translatingthem to the clinicDifferences in conditioning regimensPre-transplant conditioning in humans mainly consists of chemotherapyregimens, given with or without radiation, which is delivered at a rate (typicallyfractionated) that differs from that used in mouse models of GvHD (usuallysingle dose). Conditioning results in tissue damage and a proinflammatoryreaction that might differ between mice and humans and therefore influencethe GvHD phenotype. In addition, conditioning regimens and the timing oftransplantation can impact experimental outcomes (Gendelman et al., 2004;Mabed et al., 2005; Schwarte and Hoffmann, 2005; Mapara et al., 2006). Thiscan be directly related to the residual host-versus-graft (HVG) effect, which candecrease GvHD severity if the host immune system is not adequatelysuppressed. The HVG reaction is a rare complication after transplant in humansbut increases as conditioning regimen intensity is decreased.Genetic and immunological disparities between donor and recipientIn humans, HLA typing occurs via high-resolution DNA typing, and mosttransplants are matched at the allele level with respect to MHC loci, althoughthey usually differ at the level of miHAs that lie outside of the MHC locus. Bycontrast, many mouse models of GvHD involve mismatches in both MHCantigens and miHAs. There are also key species differences between themouse and human immune system that can make the direct translation ofresults difficult. [These have been reviewed elsewhere (Mestas and Hughes,2004).] Some of these differences include: involution of the thymus in humansbut not mice; expression of MHC II by human but not mouse T cells; anddifferences in the proportions of circulating lymphocytes and their effectormechanisms in the two species. Furthermore, strain-specific disparities in theproportion of lymphocyte subsets (such as CD4+, CD8+ and TReg cells)influence the phenotype between different mouse models of GvHD. Finally,recent reports suggest that MHC-matched mice can carry geneticpolymorphisms outside of the MHC locus that can affect the clinicalphenotype of GvHD, which might explain observed differences in results indifferent labs, even when the same strain combinations are used (Cao et al.,2003; Fanning et al., 2009).Source of donor immune cellsMaterial for HSCT in humans is now most commonly derived from mobilizedstem cell products and contains donor immune cells from the circulation. Thenumber, origin and type of circulating immune cells can differ depending onthe mobilization regimen used. By contrast, immune cells delivered togetherwith transplants in mouse models are usually isolated from spleen or lymphnodes. It is important to consider that immune-cell populations from differentsources might have different trafficking capacities and composition (i.e.different proportions of T-cell subsets, dendritic cells and early myeloidprogenitors), and therefore have a different influence on the GvHD phenotype.Differences in normal floraEnteric pathogens can play a role in the kinetics and severity of GvHD, whichtargets the gut in many cases. Microorganisms express molecules that canactivate the immune system and therefore can augment the severity ofdisease (Cooke et al., 1998). In line with this, changes in the environment ofthe mice, which in turn affect the composition of gut flora, have been shownto affect disease severity (Bennett et al., 1998; Gerbitz et al., 2004; Gorski et al.,2007). Given that the gut flora of a human is markedly different from that of amouse kept in a pathogen-free facility, this should be taken into account whentranslating experimental data.AgeHSCT is commonly carried out in adult humans, whereas mice used to studyGvHD are typically 8- to 14-weeks old (and have a 2-year lifespan). Given thatage can influence the efficiency of immune reconstitution after transplant, aswell as susceptibility to GvHD (Ordemann et al., 2002), this factor must betaken into account when translating experimental results.[Adapted, with permission, from Socié and Blazar (Socié and Blazar, 2009).]

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Page 13: Mouse models of graft-versus-host disease: advances and … › content › dmm › 4 › 3 › 318.full.pdf · PERSPECTIVE 318 dmm.biologists.org Introduction A major complication

dmm.biologists.org330

Mouse models of GvHDPERSPECTIVE

et al., 2004; Sun et al., 2005), and the DNA methyltransferaseinhibitors decitabine and azacitidine (Choi et al., 2010). The exactmechanism by which these agents work is not known, but theymight involve suppression of alloreactive T-cell responses and/oran increase in TReg cell number or function followingtransplantation. In addition, cellular therapies, such as depleting Tcells from the graft, or infusing TReg cells, have been developedbased on studies performed in the mouse. Finally, cGvHD modelshave been instrumental in developing imatinib – a tyrosine kinaseinhibitor that targets PDGFR, which has been implicated in thepathogenesis of fibrosis (McCormick et al., 1999; Daniels et al., 2004;Abdollahi et al., 2005) – and rituximab, an antibody that targetspotential autoreactive B cells (Cutler et al., 2006). Areas that warrantfurther study include the influence of reduced intensityconditioning regimens (Sadeghi et al., 2008), in vivo imaging, andthe genomics and epigenomics of GvHD.

As discussed, advances in in vivo imaging techniques to monitortrafficking of immune cells have recently been developed and willbe crucial for dissecting the temporal and spatial relationships ofdonor and recipient T cells, B cells and APCs in these models(Panoskaltsis-Mortari et al., 2004). Furthermore, recent advancesin proteomics, gene profiling and whole genome sequencing willhelp to elucidate the genetic basis of GvHD, provide ways to bettermatch donor and recipient to prevent GvHD, and improve ourability to predict the severity and kinetics of the disease (Baron etal., 2007; Hori et al., 2008; Paczesny et al., 2009).

Despite the advances made through preclinical modeling ofGvHD in mice, many questions remain. Mouse models willcontinue to allow for the dissection of mechanistic pathways ofGvHD through the use of informative genetic knock-out, knock-in and knock-down experiments that are only possible in the mouse.Fundamental research using mouse models of GvHD shouldenhance our understanding of GvHD in humans and result inimproved and novel therapeutic approaches to minimize GvHDwhile optimizing the GVL effect, as well as immune reconstitution,following HSCT.COMPETING INTERESTSThe authors have no relevant financial or competing interests to disclose.

REFERENCESAbdollahi, A., Li, M., Ping, G., Plathow, C., Domhan, S., Kiessling, F., Lee, L. B.,

McMahon, G., Grone, H. J., Lipson, K. E. et al. (2005). Inhibition of platelet-derivedgrowth factor signaling attenuates pulmonary fibrosis. J. Exp. Med. 201, 925-935.

Albert, M. H., Liu, Y., Anasetti, C. and Yu, X. Z. (2005). Antigen-dependentsuppression of alloresponses by Foxp3-induced regulatory T cells in transplantation.Eur. J. Immunol. 35, 2598-2607.

Alousi, A. M., Weisdorf, D. J., Logan, B. R., Bolanos-Meade, J., Carter, S., Difronzo,N., Pasquini, M., Goldstein, S. C., Ho, V. T., Hayes-Lattin, B. et al. (2009).Etanercept, mycophenolate, denileukin, or pentostatin plus corticosteroids for acutegraft-versus-host disease: a randomized phase 2 trial from the Blood and MarrowTransplant Clinical Trials Network. Blood 114, 511-517.

Anderson, B. E., McNiff, J. M., Jain, D., Blazar, B. R., Shlomchik, W. D. andShlomchik, M. J. (2005). Distinct roles for donor- and host-derived antigen-presenting cells and costimulatory molecules in murine chronic graft-versus-hostdisease: requirements depend on target organ. Blood 105, 2227-2234.

Antin, J. H., Weisdorf, D., Neuberg, D., Nicklow, R., Clouthier, S., Lee, S. J., Alyea, E.,McGarigle, C., Blazar, B. R., Sonis, S. et al. (2002). Interleukin-1 blockade does notprevent acute graft-versus-host disease: results of a randomized, double-blind,placebo-controlled trial of interleukin-1 receptor antagonist in allogeneic bonemarrow transplantation. Blood 100, 3479-3482.

Baker, M. B., Altman, N. H., Podack, E. R. and Levy, R. B. (1996). The role of cell-mediated cytotoxicity in acute GVHD after MHC-matched allogeneic bone marrowtransplantation in mice. J. Exp. Med. 183, 2645-2656.

Barnden, M. J., Allison, J., Heath, W. R. and Carbone, F. R. (1998). Defective TCRexpression in transgenic mice constructed using cDNA-based alpha- and beta-chaingenes under the control of heterologous regulatory elements. Immunol. Cell Biol. 76,34-40.

Barnes, D. W. and Loutit, J. F. (1957). Treatment of murine leukaemia with x-rays andhomologous bone marrow. II. Br. J. Haematol. 3, 241-252.

Baron, C., Somogyi, R., Greller, L. D., Rineau, V., Wilkinson, P., Cho, C. R., Cameron,M. J., Kelvin, D. J., Chagnon, P., Roy, D. C. et al. (2007). Prediction of graft-versus-host disease in humans by donor gene-expression profiling. PLoS Med. 4, e23.

Beilhack, A., Schulz, S., Baker, J., Beilhack, G. F., Wieland, C. B., Herman, E. I.,Baker, E. M., Cao, Y. A., Contag, C. H. and Negrin, R. S. (2005). In vivo analyses ofearly events in acute graft-versus-host disease reveal sequential infiltration of T-cellsubsets. Blood 106, 1113-1122.

Bennett, M., Taylor, P. A., Austin, M., Baker, M. B., Schook, L. B., Rutherford, M.,Kumar, V., Podack, E. R., Mohler, K. M., Levy, R. B. et al. (1998). Cytokine andcytotoxic pathways of NK cell rejection of class I-deficient bone marrow grafts:influence of mouse colony environment. Int. Immunol. 10, 785-790.

Berger, M., Wettstein, P. J. and Korngold, R. (1994). T cell subsets involved in lethalgraft-versus-host disease directed to immunodominant minor histocompatibilityantigens. Transplantation 57, 1095-1102.

Blaser, B. W., Roychowdhury, S., Kim, D. J., Schwind, N. R., Bhatt, D., Yuan, W.,Kusewitt, D. F., Ferketich, A. K., Caligiuri, M. A. and Guimond, M. (2005). Donor-derived IL-15 is critical for acute allogeneic graft-versus-host disease. Blood 105, 894-901.

Blazar, B. R., Carroll, S. F. and Vallera, D. A. (1991). Prevention of murine graft-versus-host disease and bone marrow alloengraftment across the major histocompatibilitybarrier after donor graft preincubation with anti-LFA1 immunotoxin. Blood 78, 3093-3102.

Bleakley, M. and Riddell, S. R. (2004). Molecules and mechanisms of the graft-versus-leukaemia effect. Nat. Rev. Cancer 4, 371-380.

Bonomo, A., Kehn, P. J., Payer, E., Rizzo, L., Cheever, A. W. and Shevach, E. M.(1995). Pathogenesis of post-thymectomy autoimmunity. Role of syngeneic MLR-reactive T cells. J. Immunol. 154, 6602-6611.

Braun, M. Y., Lowin, B., French, L., Acha-Orbea, H. and Tschopp, J. (1996). CytotoxicT cells deficient in both functional fas ligand and perforin show residual cytolyticactivity yet lose their capacity to induce lethal acute graft-versus-host disease. J. Exp.

Med. 183, 657-661.Brok, H. P., Vossen, J. M. and Heidt, P. J. (1998). IFN-gamma-mediated prevention of

graft-versus-host disease: pharmacodynamic studies and influence on proliferativecapacity of chimeric spleen cells. Bone Marrow Transplant. 22, 1005-1010.

Brown, G. R., Lee, E. and Thiele, D. L. (2002). TNF-TNFR2 interactions are critical forthe development of intestinal graft-versus-host disease in MHC class II-disparate(C57BL/6J->C57BL/6J x bm12)F1 mice. J. Immunol. 168, 3065-3071.

Cao, T. M., Lo, B., Ranheim, E. A., Grumet, F. C. and Shizuru, J. A. (2003). Variablehematopoietic graft rejection and graft-versus-host disease in MHC-matched strainsof mice. Proc. Natl. Acad. Sci. USA 100, 11571-11576.

Carlson, M. J., West, M. L., Coghill, J. M., Panoskaltsis-Mortari, A., Blazar, B. R. andSerody, J. S. (2009). In vitro-differentiated TH17 cells mediate lethal acute graft-versus-host disease with severe cutaneous and pulmonary pathologicmanifestations. Blood 113, 1365-1374.

Chen, X., Vodanovic-Jankovic, S., Johnson, B., Keller, M., Komorowski, R. andDrobyski, W. R. (2007). Absence of regulatory T-cell control of TH1 and TH17 cells isresponsible for the autoimmune-mediated pathology in chronic graft-versus-hostdisease. Blood 110, 3804-3813.

Choi, J., Ritchey, J., Prior, J. L., Holt, M., Shannon, W. D., Deych, E., Piwnica-Worms,D. R. and DiPersio, J. F. (2010). In vivo administration of hypomethylating agentsmitigate graft-versus-host disease without sacrificing graft-versus-leukemia. Blood

116, 129-139.Christianson, S. W., Greiner, D. L., Hesselton, R. A., Leif, J. H., Wagar, E. J.,

Schweitzer, I. B., Rajan, T. V., Gott, B., Roopenian, D. C. and Shultz, L. D. (1997).Enhanced human CD4+ T cell engraftment in beta2-microglobulin-deficient NOD-scid mice. J. Immunol. 158, 3578-3586.

Chu, Y. W. and Gress, R. E. (2008). Murine models of chronic graft-versus-host disease:insights and unresolved issues. Biol. Blood Marrow Transplant. 14, 365-378.

Claman, H. N., Jaffee, B. D., Huff, J. C. and Clark, R. A. (1985). Chronic graft-versus-host disease as a model for scleroderma. II. Mast cell depletion with deposition ofimmunoglobulins in the skin and fibrosis. Cell. Immunol. 94, 73-84.

Cooke, K. R., Kobzik, L., Martin, T. R., Brewer, J., Delmonte, J., Jr, Crawford, J. M.and Ferrara, J. L. (1996). An experimental model of idiopathic pneumoniasyndrome after bone marrow transplantation: I. The roles of minor H antigens andendotoxin. Blood 88, 3230-3239.

Cooke, K. R., Hill, G. R., Crawford, J. M., Bungard, D., Brinson, Y. S., Delmonte, J., Jrand Ferrara, J. L. (1998). Tumor necrosis factor- alpha production to

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Page 14: Mouse models of graft-versus-host disease: advances and … › content › dmm › 4 › 3 › 318.full.pdf · PERSPECTIVE 318 dmm.biologists.org Introduction A major complication

Disease Models & Mechanisms 331

Mouse models of GvHD PERSPECTIVE

lipopolysaccharide stimulation by donor cells predicts the severity of experimentalacute graft-versus-host disease. J. Clin. Invest. 102, 1882-1891.

Cutler, C., Miklos, D., Kim, H. T., Treister, N., Woo, S. B., Bienfang, D., Klickstein, L.B., Levin, J., Miller, K., Reynolds, C. et al. (2006). Rituximab for steroid-refractorychronic graft-versus-host disease. Blood 108, 756-762.

Damdinsuren, B., Zhang, Y., Khalil, A., Wood, W. H., 3rd, Becker, K. G., Shlomchik,M. J. and Sen, R. (2010). Single round of antigen receptor signaling programs naiveB cells to receive T cell help. Immunity 32, 355-366.

Daniels, C. E., Wilkes, M. C., Edens, M., Kottom, T. J., Murphy, S. J., Limper, A. H.and Leof, E. B. (2004). Imatinib mesylate inhibits the profibrogenic activity of TGF-beta and prevents bleomycin-mediated lung fibrosis. J. Clin. Invest. 114, 1308-1316.

De Wit, D., Van Mechelen, M., Zanin, C., Doutrelepont, J. M., Velu, T., Gerard, C.,Abramowicz, D., Scheerlinck, J. P., De Baetselier, P., Urbain, J. et al. (1993).Preferential activation of Th2 cells in chronic graft-versus-host reaction. J. Immunol.

150, 361-366.DeClerck, Y., Draper, V. and Parkman, R. (1986). Clonal analysis of murine graft-vs-

host disease. II. Leukokines that stimulate fibroblast proliferation and collagensynthesis in graft-vs. host disease. J. Immunol. 136, 3549-3552.

Durie, F. H., Aruffo, A., Ledbetter, J., Crassi, K. M., Green, W. R., Fast, L. D. andNoelle, R. J. (1994). Antibody to the ligand of CD40, gp39, blocks the occurrence ofthe acute and chronic forms of graft-vs-host disease. J. Clin. Invest. 94, 1333-1338.

Edinger, M., Hoffmann, P., Ermann, J., Drago, K., Fathman, C. G., Strober, S. andNegrin, R. S. (2003). CD4+CD25+ regulatory T cells preserve graft-versus-tumoractivity while inhibiting graft-versus-host disease after bone marrow transplantation.Nat. Med. 9, 1144-1150.

Eyrich, M., Burger, G., Marquardt, K., Budach, W., Schilbach, K., Niethammer, D.and Schlegel, P. G. (2005). Sequential expression of adhesion and costimulatorymolecules in graft-versus-host disease target organs after murine bone marrowtransplantation across minor histocompatibility antigen barriers. Biol. Blood Marrow

Transplant. 11, 371-382.Fanning, S. L., Appel, M. Y., Berger, S. A., Korngold, R. and Friedman, T. M. (2009).

The immunological impact of genetic drift in the B10.BR congenic inbred mousestrain. J. Immunol. 183, 4261-4272.

Ferrara, J. L., Abhyankar, S. and Gilliland, D. G. (1993). Cytokine storm of graft-versus-host disease: a critical effector role for interleukin-1. Transplant. Proc. 25,1216-1217.

Ferrara, J. L., Levine, J. E., Reddy, P. and Holler, E. (2009). Graft-versus-host disease.Lancet 373, 1550-1561.

Fowler, D. H., Breglio, J., Nagel, G., Eckhaus, M. A. and Gress, R. E. (1996).Allospecific CD8+ Tc1 and Tc2 populations in graft-versus-leukemia effect and graft-versus-host disease. J. Immunol. 157, 4811-4821.

Fujiwara, K., Sakaguchi, N. and Watanabe, T. (1991). Sialoadenitis in experimentalgraft-versus-host disease. An animal model of Sjogren’s syndrome. Lab. Invest. 65,710-718.

Fukushi, N., Arase, H., Wang, B., Ogasawara, K., Gotohda, T., Good, R. A. and Onoe,K. (1990). Thymus: a direct target tissue in graft-versus-host reaction after allogeneicbone marrow transplantation that results in abrogation of induction of self-tolerance. Proc. Natl. Acad. Sci. USA 87, 6301-6305.

Gendelman, M., Hecht, T., Logan, B., Vodanovic-Jankovic, S., Komorowski, R. andDrobyski, W. R. (2004). Host conditioning is a primary determinant in modulatingthe effect of IL-7 on murine graft-versus-host disease. J. Immunol. 172, 3328-3336.

Gerbitz, A., Schultz, M., Wilke, A., Linde, H. J., Scholmerich, J., Andreesen, R. andHoller, E. (2004). Probiotic effects on experimental graft-versus-host disease: letthem eat yogurt. Blood 103, 4365-4367.

Ghayur, T., Seemayer, T. A., Xenocostas, A. and Lapp, W. S. (1988). Completesequential regeneration of graft-vs.-host-induced severely dysplastic thymuses.Implications for the pathogenesis of chronic graft-vs.-host disease. Am. J. Pathol. 133,39-46.

Gorski, J., Chen, X., Gendelman, M., Yassai, M., Krueger, A., Tivol, E., Logan, B.,Komorowski, R., Vodanovic-Jankovic, S. and Drobyski, W. R. (2007). Homeostaticexpansion and repertoire regeneration of donor T cells during graft versus hostdisease is constrained by the host environment. Blood 109, 5502-5510.

Graubert, T. A., DiPersio, J. F., Russell, J. H. and Ley, T. J. (1997). Perforin/granzyme-dependent and independent mechanisms are both important for the developmentof graft-versus-host disease after murine bone marrow transplantation. J. Clin. Invest.

100, 904-911.Grubin, C. E., Kovats, S., deRoos, P. and Rudensky, A. Y. (1997). Deficient positive

selection of CD4 T cells in mice displaying altered repertoires of MHC class II-boundself-peptides. Immunity 7, 197-208.

Hakim, F. T., Sharrow, S. O., Payne, S. and Shearer, G. M. (1991). Repopulation ofhost lymphohematopoietic systems by donor cells during graft-versus-host reactionin unirradiated adult F1 mice injected with parental lymphocytes. J. Immunol. 146,2108-2115.

Hamilton, B. L. (1987). L3T4-positive T cells participate in the induction of graft-vs-host disease in response to minor histocompatibility antigens. J. Immunol. 139,2511-2515.

Hamilton, B. L. and Parkman, R. (1983). Acute and chronic graft-versus-host diseaseinduced by minor histocompatibility antigens in mice. Transplantation 36, 150-155.

Hauri-Hohl, M. M., Keller, M. P., Gill, J., Hafen, K., Pachlatko, E., Boulay, T., Peter, A.,Hollander, G. A. and Krenger, W. (2007). Donor T-cell alloreactivity against hostthymic epithelium limits T-cell development after bone marrow transplantation.Blood 109, 4080-4088.

Hildebrandt, G. C., Olkiewicz, K. M., Corrion, L. A., Chang, Y., Clouthier, S. G., Liu,C. and Cooke, K. R. (2004). Donor-derived TNF-alpha regulates pulmonarychemokine expression and the development of idiopathic pneumonia syndromeafter allogeneic bone marrow transplantation. Blood 104, 586-593.

Hill, G. R., Crawford, J. M., Cooke, K. R., Brinson, Y. S., Pan, L. and Ferrara, J. L.(1997). Total body irradiation and acute graft-versus-host disease: the role ofgastrointestinal damage and inflammatory cytokines. Blood 90, 3204-3213.

Hoffmann-Fezer, G., Gall, C., Zengerle, U., Kranz, B. and Thierfelder, S. (1993).Immunohistology and immunocytology of human T-cell chimerism and graft-versus-host disease in SCID mice. Blood 81, 3440-3448.

Hori, T., Naishiro, Y., Sohma, H., Suzuki, N., Hatakeyama, N., Yamamoto, M.,Sonoda, T., Mizue, Y., Imai, K., Tsutsumi, H. et al. (2008). CCL8 is a potentialmolecular candidate for the diagnosis of graft-versus-host disease. Blood 111, 4403-4412.

Howell, C. D., Yoder, T., Claman, H. N. and Vierling, J. M. (1989). Hepatic homing ofmononuclear inflammatory cells isolated during murine chronic graft-vs-hostdisease. J. Immunol. 143, 476-483.

Iclozan, C., Yu, Y., Liu, C., Liang, Y., Yi, T., Anasetti, C. and Yu, X. Z. (2009). T helper17cells are sufficient but not necessary to induce acute graft-versus-host disease. Biol.Blood Marrow Transplant. 16, 170-178.

Ito, R., Katano, I., Kawai, K., Hirata, H., Ogura, T., Kamisako, T., Eto, T. and Ito, M.(2009). Highly sensitive model for xenogenic GVHD using severe immunodeficientNOG mice. Transplantation 87, 1654-1658.

Ito, S., Ueno, M., Nishi, S., Arakawa, M., Ikarashi, Y., Saitoh, T. and Fujiwara, M.(1992). Histological characteristics of lupus nephritis in F1 mice with chronic graft-versus-host reaction across MHC class II difference. Autoimmunity 12, 79-87.

Jaffee, B. D. and Claman, H. N. (1983). Chronic graft-versus-host disease (GVHD) as amodel for scleroderma. I. Description of model systems. Cell. Immunol. 77, 1-12.

Jiang, Z., Podack, E. and Levy, R. B. (2001). Major histocompatibility complex-mismatched allogeneic bone marrow transplantation using perforin and/or Fasligand double-defective CD4(+) donor T cells: involvement of cytotoxic function bydonor lymphocytes prior to graft-versus-host disease pathogenesis. Blood 98, 390-397.

Kanamaru, A., Okamoto, T., Matsuda, K., Hara, H. and Nagai, K. (1984). Elevation oferythroid colony-stimulating activity in the serum of mice with graft-versus-hostdisease. Exp. Hematol. 12, 763-767.

Kaplan, D. H., Anderson, B. E., McNiff, J. M., Jain, D., Shlomchik, M. J. andShlomchik, W. D. (2004). Target antigens determine graft-versus-host diseasephenotype. J. Immunol. 173, 5467-5475.

Kappel, L. W., Goldberg, G. L., King, C. G., Suh, D. Y., Smith, O. M., Ligh, C., Holland,A. M., Grubin, J., Mark, N. M., Liu, C. et al. (2009). IL-17 contributes to CD4-mediated graft-versus-host disease. Blood 113, 945-952.

Kim, J., Kim, H. J., Choi, W. S., Nam, S. H., Cho, H. R. and Kwon, B. (2006).Maintenance of CD8+ T-cell anergy by CD4+CD25+ regulatory T cells in chronicgraft-versus-host disease. Exp. Mol. Med. 38, 494-501.

Kim, J., Park, K., Kim, H. J., Kim, H. A., Jung, D., Choi, H. J., Choi, S. Y., Seo, K. W.,Cho, H. R. and Kwon, B. (2008). Breaking of CD8+ T cell tolerance through in vivoligation of CD40 results in inhibition of chronic graft-versus-host disease andcomplete donor cell engraftment. J. Immunol. 181, 7380-7389.

Kim, Y. M., Sachs, T., Asavaroengchai, W., Bronson, R. and Sykes, M. (2003). Graft-versus-host disease can be separated from graft-versus-lymphoma effects by controlof lymphocyte trafficking with FTY720. J. Clin. Invest. 111, 659-669.

King, M. A., Covassin, L., Brehm, M. A., Racki, W., Pearson, T., Leif, J., Laning, J.,Fodor, W., Foreman, O., Burzenski, L. et al. (2009). Human peripheral bloodleucocyte non-obese diabetic-severe combined immunodeficiency interleukin-2receptor gamma chain gene mouse model of xenogeneic graft-versus-host-likedisease and the role of host major histocompatibility complex. Clin. Exp. Immunol.157, 104-118.

Kisielow, P., Bluthmann, H., Staerz, U. D., Steinmetz, M. and von Boehmer, H.(1988). Tolerance in T-cell-receptor transgenic mice involves deletion of nonmatureCD4+8+ thymocytes. Nature 333, 742-746.

Kolb, H. J. (2008). Graft-versus-leukemia effects of transplantation and donorlymphocytes. Blood 112, 4371-4383.

Kolb, H. J., Schmid, C., Barrett, A. J. and Schendel, D. J. (2004). Graft-versus-leukemiareactions in allogeneic chimeras. Blood 103, 767-776.

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Page 15: Mouse models of graft-versus-host disease: advances and … › content › dmm › 4 › 3 › 318.full.pdf · PERSPECTIVE 318 dmm.biologists.org Introduction A major complication

dmm.biologists.org332

Mouse models of GvHDPERSPECTIVE

Korngold, R. (1992). Lethal graft-versus-host disease in mice directed to multipleminor histocompatibility antigens: features of CD8+ and CD4+ T cell responses. BoneMarrow Transplant. 9, 355-364.

Korngold, R. and Sprent, J. (1978). Lethal graft-versus-host disease after bone marrowtransplantation across minor histocompatibility barriers in mice. Prevention byremoving mature T cells from marrow. J. Exp. Med. 148, 1687-1698.

Korngold, R. and Sprent, J. (1987). Variable capacity of L3T4+ T cells to cause lethalgraft-versus-host disease across minor histocompatibility barriers in mice. J. Exp. Med.165, 1552-1564.

Krenger, W. and Hollander, G. A. (2008). The immunopathology of thymic GVHD.Semin. Immunopathol. 30, 439-456.

Krenger, W., Rossi, S. and Hollander, G. A. (2000a). Apoptosis of thymocytes duringacute graft-versus-host disease is independent of glucocorticoids. Transplantation69, 2190-2193.

Krenger, W., Rossi, S., Piali, L. and Hollander, G. A. (2000b). Thymic atrophy inmurine acute graft-versus-host disease is effected by impaired cell cycle progressionof host pro-T and pre-T cells. Blood 96, 347-354.

Lee, S. J. (2010). Have we made progress in the management of chronic graft-vs-hostdisease? Best Pract. Res Clin. Haematol. 23, 529-535.

Levi-Schaffer, F. and Weg, V. B. (1997). Mast cells, eosinophils and fibrosis. Clin. Exp.Allergy 27 Suppl. 1, 64-70.

Levy, S., Nagler, A., Okon, S. and Marmary, Y. (2000). Parotid salivary glanddysfunction in chronic graft-versus-host disease (cGVHD): a longitudinal study in amouse model. Bone Marrow Transplant. 25, 1073-1078.

Liang, Y., Liu, C., Djeu, J. Y., Zhong, B., Peters, T., Scharffetter-Kochanek, K.,Anasetti, C. and Yu, X. Z. (2008). Beta2 integrins separate graft-versus-host diseaseand graft-versus-leukemia effects. Blood 111, 954-962.

Lu, Y. and Waller, E. K. (2009). Dichotomous role of interferon-gamma in allogeneicbone marrow transplant. Biol. Blood Marrow Transplant. 15, 1347-1353.

Lucas, P. J., Shearer, G. M., Neudorf, S. and Gress, R. E. (1990). The humanantimurine xenogeneic cytotoxic response. I. Dependence on responder antigen-presenting cells. J. Immunol. 144, 4548-4554.

Mabed, M., Maroof, S., Zalta, K. and El-Awadee, M. (2005). Delayed or delayedsequential bone marrow transplantation: relevance for acute graft-versus-hostdisease prevention after major H2 incompatible transplantation. Bone MarrowTransplant. 35, 803-806.

Maeda, Y., Levy, R. B., Reddy, P., Liu, C., Clouthier, S. G., Teshima, T. and Ferrara, J.L. (2005). Both perforin and Fas ligand are required for the regulation of alloreactiveCD8+ T cells during acute graft-versus-host disease. Blood 105, 2023-2027.

Mapara, M. Y., Leng, C., Kim, Y. M., Bronson, R., Lokshin, A., Luster, A. and Sykes,M. (2006). Expression of chemokines in GVHD target organs is influenced byconditioning and genetic factors and amplified by GVHR. Biol. Blood MarrowTransplant. 12, 623-634.

Mathis, D. and Benoist, C. (2009). Aire. Annu. Rev. Immunol. 27, 287-312.McCormick, L. L., Zhang, Y., Tootell, E. and Gilliam, A. C. (1999). Anti-TGF-beta

treatment prevents skin and lung fibrosis in murine sclerodermatous graft-versus-host disease: a model for human scleroderma. J. Immunol. 163, 5693-5699.

Mestas, J. and Hughes, C. C. (2004). Of mice and not men: differences betweenmouse and human immunology. J. Immunol. 172, 2731-2738.

Morris, S. C., Cheek, R. L., Cohen, P. L. and Eisenberg, R. A. (1990a). Allotype-specificimmunoregulation of autoantibody production by host B cells in chronic graft-versus host disease. J. Immunol. 144, 916-922.

Morris, S. C., Cheek, R. L., Cohen, P. L. and Eisenberg, R. A. (1990b). Autoantibodiesin chronic graft versus host result from cognate T-B interactions. J. Exp. Med. 171,503-517.

Mosier, D. E., Gulizia, R. J., Baird, S. M. and Wilson, D. B. (1988). Transfer of afunctional human immune system to mice with severe combinedimmunodeficiency. Nature 335, 256-259.

Murphy, K. M., Heimberger, A. B. and Loh, D. Y. (1990). Induction by antigen ofintrathymic apoptosis of CD4+CD8+TCRlo thymocytes in vivo. Science 250, 1720-1723.

Negrin, R. S. and Contag, C. H. (2006). In vivo imaging using bioluminescence: a toolfor probing graft-versus-host disease. Nat. Rev. Immunol. 6, 484-490.

Nervi, B., Rettig, M. P., Ritchey, J. K., Wang, H. L., Bauer, G., Walker, J., Bonyhadi, M.L., Berenson, R. J., Prior, J. L., Piwnica-Worms, D. et al. (2007). Factors affectinghuman T cell engraftment, trafficking, and associated xenogeneic graft-vs-hostdisease in NOD/SCID beta2mnull mice. Exp. Hematol. 35, 1823-1838.

Nestel, F. P., Price, K. S., Seemayer, T. A. and Lapp, W. S. (1992). Macrophage primingand lipopolysaccharide-triggered release of tumor necrosis factor alpha during graft-versus-host disease. J. Exp. Med. 175, 405-413.

Niculescu, F., Niculescu, T., Nguyen, P., Puliaev, R., Papadimitriou, J. C., Gaspari, A.,Rus, H. and Via, C. S. (2005). Both apoptosis and complement membrane attackcomplex deposition are major features of murine acute graft-vs.-host disease. Exp.Mol. Pathol. 79, 136-145.

Nikolic, B., Lee, S., Bronson, R. T., Grusby, M. J. and Sykes, M. (2000). Th1 and Th2mediate acute graft-versus-host disease, each with distinct end-organ targets. J. Clin.

Invest. 105, 1289-1298.Nishimura, R., Baker, J., Beilhack, A., Zeiser, R., Olson, J. A., Sega, E. I., Karimi, M.

and Negrin, R. S. (2008). In vivo trafficking and survival of cytokine-induced killercells resulting in minimal GVHD with retention of antitumor activity. Blood 112,2563-2574.

Nishizuka, Y. and Sakakura, T. (1969). Thymus and reproduction: sex-linkeddysgenesia of the gonad after neonatal thymectomy in mice. Science 166, 753-755.

Ordemann, R., Hutchinson, R., Friedman, J., Burakoff, S. J., Reddy, P., Duffner, U.,Braun, T. M., Liu, C., Teshima, T. and Ferrara, J. L. (2002). Enhanced allostimulatoryactivity of host antigen-presenting cells in old mice intensifies acute graft-versus-host disease. J. Clin. Invest. 109, 1249-1256.

Paczesny, S., Krijanovski, O. I., Braun, T. M., Choi, S. W., Clouthier, S. G., Kuick, R.,Misek, D. E., Cooke, K. R., Kitko, C. L., Weyand, A. et al. (2009). A biomarker panelfor acute graft-versus-host disease. Blood 113, 273-278.

Pals, S. T., Radaszkiewicz, T., Roozendaal, L. and Gleichmann, E. (1985). Chronicprogressive polyarthritis and other symptoms of collagen vascular disease inducedby graft-vs-host reaction. J. Immunol. 134, 1475-1482.

Panoskaltsis-Mortari, A., Lacey, D. L., Vallera, D. A. and Blazar, B. R. (1998).Keratinocyte growth factor administered before conditioning ameliorates graft-versus-host disease after allogeneic bone marrow transplantation in mice. Blood 92,3960-3967.

Panoskaltsis-Mortari, A., Price, A., Hermanson, J. R., Taras, E., Lees, C., Serody, J. S.and Blazar, B. R. (2004). In vivo imaging of graft-versus-host-disease in mice. Blood

103, 3590-3598.Pickel, K. and Hoffmann, M. K. (1977). Suppressor T cells arising in mice undergoing a

graft-vs-host response. J. Immunol. 118, 653-656.Ringden, O., Karlsson, H., Olsson, R., Omazic, B. and Uhlin, M. (2009). The

allogeneic graft-versus-cancer effect. Br. J. Haematol. 147, 614-633.Rolink, A. G. and Gleichmann, E. (1983). Allosuppressor- and allohelper-T cells in

acute and chronic graft-vs.-host (GVH) disease. III. Different Lyt subsets of donor Tcells induce different pathological syndromes. J. Exp. Med. 158, 546-558.

Rolink, A. G., Pals, S. T. and Gleichmann, E. (1983). Allosuppressor and allohelper Tcells in acute and chronic graft-vs.-host disease. II. F1 recipients carrying mutationsat H-2K and/or I-A. J. Exp. Med. 157, 755-771.

Ruzek, M. C., Jha, S., Ledbetter, S., Richards, S. M. and Garman, R. D. (2004). Amodified model of graft-versus-host-induced systemic sclerosis (scleroderma)exhibits all major aspects of the human disease. Arthritis Rheum. 50, 1319-1331.

Sadeghi, B., Aghdami, N., Hassan, Z., Forouzanfar, M., Rozell, B., Abedi-Valugerdi,M. and Hassan, M. (2008). GVHD after chemotherapy conditioning in allogeneictransplanted mice. Bone Marrow Transplant. 42, 807-818.

Sakaguchi, S., Ono, M., Setoguchi, R., Yagi, H., Hori, S., Fehervari, Z., Shimizu, J.,Takahashi, T. and Nomura, T. (2006). Foxp3+ CD25+ CD4+ natural regulatory T cellsin dominant self-tolerance and autoimmune disease. Immunol. Rev. 212, 8-27.

Sakoda, Y., Hashimoto, D., Asakura, S., Takeuchi, K., Harada, M., Tanimoto, M. andTeshima, T. (2007). Donor-derived thymic-dependent T cells cause chronic graft-versus-host disease. Blood 109, 1756-1764.

Schwarte, S. and Hoffmann, M. W. (2005). Influence of radiation protocols on graft-vs-host disease incidence after bone-marrow transplantation in experimentalmodels. Methods Mol. Med. 109, 445-458.

Schwarte, S., Bremer, M., Fruehauf, J., Sorge, Y., Skubich, S. and Hoffmann, M. W.(2007). Radiation protocols determine acute graft-versus-host disease incidence afterallogeneic bone marrow transplantation in murine models. Int. J. Radiat. Biol. 83,625-636.

Sha, W. C., Nelson, C. A., Newberry, R. D., Kranz, D. M., Russell, J. H. and Loh, D. Y.(1988). Positive and negative selection of an antigen receptor on T cells in transgenicmice. Nature 336, 73-76.

Shimabukuro-Vornhagen, A., Hallek, M. J., Storb, R. F. and von Bergwelt-Baildon,M. S. (2009). The role of B Cells in the pathogenesis of graft-versus-host disease.Blood 114, 4919-4927.

Shlomchik, W. D., Couzens, M. S., Tang, C. B., McNiff, J., Robert, M. E., Liu, J.,Shlomchik, M. J. and Emerson, S. G. (1999). Prevention of graft versus host diseaseby inactivation of host antigen-presenting cells. Science 285, 412-415.

Skert, C., Patriarca, F., Sperotto, A., Cerno, M., Fili, C., Zaja, F., Stocchi, R., Geromin,A., Damiani, D. and Fanin, R. (2006). Sclerodermatous chronic graft-versus-hostdisease after allogeneic hematopoietic stem cell transplantation: incidence,predictors and outcome. Haematologica 91, 258-261.

Slayback, D. L., Dobkins, J. A., Harper, J. M. and Allen, R. D. (2000). Genetic factorsinfluencing the development of chronic graft-versus-host disease in a murine model.Bone Marrow Transplant. 26, 931-938.

Socié, G. and Blazar, B. R. (2009). Acute graft-versus-host disease; from the bench tothe bedside. Blood 114, 4327-4336.

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Page 16: Mouse models of graft-versus-host disease: advances and … › content › dmm › 4 › 3 › 318.full.pdf · PERSPECTIVE 318 dmm.biologists.org Introduction A major complication

Disease Models & Mechanisms 333

Mouse models of GvHD PERSPECTIVE

Sprent, J., Schaefer, M., Lo, D. and Korngold, R. (1986). Properties of purified T cellsubsets. II. In vivo responses to class I vs. class II H-2 differences. J. Exp. Med. 163,998-1011.

Sprent, J., Schaefer, M., Gao, E. K. and Korngold, R. (1988). Role of T cell subsets inlethal graft-versus-host disease (GVHD) directed to class I versus class II H-2differences. I. L3T4+ cells can either augment or retard GVHD elicited by Lyt-2+ cellsin class I different hosts. J. Exp. Med. 167, 556-569.

Sprent, J., Schaefer, M. and Korngold, R. (1990). Role of T cell subsets in lethal graft-versus-host disease (GVHD) directed to class I versus class II H-2 differences. II.Protective effects of L3T4+ cells in anti-class II GVHD. J. Immunol. 144, 2946-2954.

Sun, K., Welniak, L. A., Panoskaltsis-Mortari, A., O’Shaughnessy, M. J., Liu, H.,Barao, I., Riordan, W., Sitcheran, R., Wysocki, C., Serody, J. S. et al. (2004).Inhibition of acute graft-versus-host disease with retention of graft-versus-tumoreffects by the proteasome inhibitor bortezomib. Proc. Natl. Acad. Sci. USA 101, 8120-8125.

Sun, K., Wilkins, D. E., Anver, M. R., Sayers, T. J., Panoskaltsis-Mortari, A., Blazar, B.R., Welniak, L. A. and Murphy, W. J. (2005). Differential effects of proteasomeinhibition by bortezomib on murine acute graft-versus-host disease (GVHD): delayedadministration of bortezomib results in increased GVHD-dependent gastrointestinaltoxicity. Blood 106, 3293-3299.

Svegliati, S., Olivieri, A., Campelli, N., Luchetti, M., Poloni, A., Trappolini, S.,Moroncini, G., Bacigalupo, A., Leoni, P., Avvedimento, E. V. et al. (2007).Stimulatory autoantibodies to PDGF receptor in patients with extensive chronicgraft-versus-host disease. Blood 110, 237-241.

Tawara, I., Maeda, Y., Sun, Y., Lowler, K. P., Liu, C., Toubai, T., McKenzie, A. N. andReddy, P. (2008). Combined Th2 cytokine deficiency in donor T cells aggravatesexperimental acute graft-vs-host disease. Exp. Hematol. 36, 988-996.

Teshima, T., Ordemann, R., Reddy, P., Gagin, S., Liu, C., Cooke, K. R. and Ferrara, J.L. (2002). Acute graft-versus-host disease does not require alloantigen expression onhost epithelium. Nat. Med. 8, 575-581.

Tivol, E., Komorowski, R. and Drobyski, W. R. (2005). Emergent autoimmunity ingraft-versus-host disease. Blood 105, 4885-4891.

Tschetter, J. R., Mozes, E. and Shearer, G. M. (2000). Progression from acute tochronic disease in a murine parent-into-F1 model of graft-versus-host disease. J.Immunol. 165, 5987-5994.

Uberti, J. P., Ayash, L., Ratanatharathorn, V., Silver, S., Reynolds, C., Becker, M.,Reddy, P., Cooke, K. R., Yanik, G., Whitfield, J. et al. (2005). Pilot trial on the use ofetanercept and methylprednisolone as primary treatment for acute graft-versus-hostdisease. Biol. Blood Marrow Transplant. 11, 680-687.

Vallera, D. A., Soderling, C. C., Carlson, G. J. and Kersey, J. H. (1981). Bone marrowtransplantation across major histocompatibility barriers in mice. Effect of eliminationof T cells from donor grafts by treatment with monoclonal Thy-1.2 plus complementor antibody alone. Transplantation 31, 218-222.

van den Brink, M. R., Moore, E., Ferrara, J. L. and Burakoff, S. J. (2000). Graft-versus-host-disease-associated thymic damage results in the appearance of T cell cloneswith anti-host reactivity. Transplantation 69, 446-449.

van Leeuwen, L., Guiffre, A., Atkinson, K., Rainer, S. P. and Sewell, W. A. (2002). Atwo-phase pathogenesis of graft-versus-host disease in mice. Bone MarrowTransplant. 29, 151-158.

van Rijn, R. S., Simonetti, E. R., Hagenbeek, A., Hogenes, M. C., de Weger, R. A.,Canninga-van Dijk, M. R., Weijer, K., Spits, H., Storm, G., van Bloois, L. et al.(2003). A new xenograft model for graft-versus-host disease by intravenous transferof human peripheral blood mononuclear cells in RAG2-/- gammac-/- double-mutantmice. Blood 102, 2522-2531.

Via, C. S., Sharrow, S. O. and Shearer, G. M. (1987). Role of cytotoxic T lymphocytes inthe prevention of lupus-like disease occurring in a murine model of graft-vs-hostdisease. J. Immunol. 139, 1840-1849.

Via, C. S., Rus, V., Gately, M. K. and Finkelman, F. D. (1994). IL-12 stimulates thedevelopment of acute graft-versus-host disease in mice that normally would

develop chronic, autoimmune graft-versus-host disease. J. Immunol. 153, 4040-4047.

Via, C. S., Rus, V., Nguyen, P., Linsley, P. and Gause, W. C. (1996a). Differential effectof CTLA4Ig on murine graft-versus-host disease (GVHD) development: CTLA4Igprevents both acute and chronic GVHD development but reverses only chronicGVHD. J. Immunol. 157, 4258-4267.

Via, C. S., Nguyen, P., Shustov, A., Drappa, J. and Elkon, K. B. (1996b). A major rolefor the Fas pathway in acute graft-versus-host disease. J. Immunol. 157, 5387-5393.

Vidal, S., Labrador, M., Rodriguez-Sanchez, J. L. and Gelpi, C. (1996). The role ofBALB/c donor CD8+ lymphocytes in graft-versus-host disease in (BALB/c x A/J)F1(CAF1) mice. J. Immunol. 156, 997-1005.

Wang, X., Roopenian, D., Martone, C., Li, N., Li, H. and Shlomchik, W. D. (2009).Mechanisms of cross-presentation in graft-vs-host disease. Blood 114, (ASH AnnualMeeting Abstracts), Abstract 687.

Watanabe, N., Wang, Y. H., Lee, H. K., Ito, T., Cao, W. and Liu, Y. J. (2005). Hassall’scorpuscles instruct dendritic cells to induce CD4+CD25+ regulatory T cells in humanthymus. Nature 436, 1181-1185.

Weiss, L., Morecki, S., Vitetta, E. S. and Slavin, S. (1983). Suppression and eliminationof BCL1 leukemia by allogeneic bone marrow transplantation. J. Immunol. 130, 2452-2455.

Westervelt, P., Lane, A. A., Pollock, J. L., Oldfather, K., Holt, M. S., Zimonjic, D. B.,Popescu, N. C., DiPersio, J. F. and Ley, T. J. (2003). High-penetrance mouse modelof acute promyelocytic leukemia with very low levels of PML-RARalpha expression.Blood 102, 1857-1865.

Wilkinson, K. N., Anderson, B., McNiff, J., Demetris, A. J., Shlomchik, W. D. andShlomchik, M. (2009). A new TCR transgenic model of GVHD reveals that,independent of repertoire, effector memory T cells are severely limited, and centralmemory T cells somewhat limited, in their ability to cause GVHD. Blood 114 (ASHAnnual Meeting Abstracts), Abstract 233.

Wynn, T. A. (2004). Fibrotic disease and the T(H)1/T(H)2 paradigm. Nat. Rev. Immunol.4, 583-594.

Wysocki, C. A., Panoskaltsis-Mortari, A., Blazar, B. R. and Serody, J. S. (2005).Leukocyte migration and graft-versus-host disease. Blood 105, 4191-4199.

Yi, T., Zhao, D., Lin, C. L., Zhang, C., Chen, Y., Todorov, I., LeBon, T., Kandeel, F.,Forman, S. and Zeng, D. (2008). Absence of donor Th17 leads to augmented Th1differentiation and exacerbated acute graft-versus-host disease. Blood 112, 2101-2110.

Yu, X. Z., Albert, M. H. and Anasetti, C. (2006). Alloantigen affinity and CD4 helpdetermine severity of graft-versus-host disease mediated by CD8 donor T cells. J.Immunol. 176, 3383-3390.

Zhang, C., Todorov, I., Zhang, Z., Liu, Y., Kandeel, F., Forman, S., Strober, S. andZeng, D. (2006). Donor CD4+ T and B cells in transplants induce chronic graft-versus-host disease with autoimmune manifestations. Blood 107, 2993-3001.

Zhang, Y., Joe, G., Hexner, E., Zhu, J. and Emerson, S. G. (2005). Alloreactive memoryT cells are responsible for the persistence of graft-versus-host disease. J. Immunol.174, 3051-3058.

Zhang, Y., Hexner, E., Frank, D. and Emerson, S. G. (2007). CD4+ T cells generated denovo from donor hemopoietic stem cells mediate the evolution from acute tochronic graft-versus-host disease. J. Immunol. 179, 3305-3314.

Zhao, D., Young, J. S., Chen, Y. H., Shen, E., Yi, T., Todorov, I., Chu, P. G., Forman, S.J. and Zeng, D. (2010). Alloimmune response results in expansion of autoreactivedonor CD4+ T cells in transplants that can mediate chronic graft-versus-host disease.J. Immunol. 186, 856-868.

Zheng, H., Matte-Martone, C., Jain, D., McNiff, J. and Shlomchik, W. D. (2009).Central memory CD8+ T cells induce graft-versus-host disease and mediate graft-versus-leukemia. J. Immunol. 182, 5938-5948.

Zhou, L., Askew, D., Wu, C. and Gilliam, A. C. (2007). Cutaneous gene expression byDNA microarray in murine sclerodermatous graft-versus-host disease, a model forhuman scleroderma. J. Invest. Dermatol. 127, 281-292.

Dise

ase

Mod

els &

Mec

hani

sms

D

MM