Kit Regulates HSC Engraftment across the Human-Mouse Species Barrier

12
Cell Stem Cell Resource Kit Regulates HSC Engraftment across the Human-Mouse Species Barrier Kadriye Nehir Cosgun, 1,9 Susann Rahmig, 1,9 Nicole Mende, 1 So ¨ ren Reinke, 1 Ilona Hauber, 2 Carola Scha ¨ fer, 2 Anke Petzold, 3 Henry Weisbach, 1,10 Gordon Heidkamp, 4 Ariawan Purbojo, 5 Robert Cesnjevar, 5 Alexander Platz, 6 Martin Bornha ¨ user, 7 Marc Schmitz, 3 Diana Dudziak, 4 Joachim Hauber, 2 Jo ¨ rg Kirberg, 8 and Claudia Waskow 1, * 1 Regeneration in Hematopoiesis and Animal Models in Hematopoiesis, Institute for Immunology, Faculty of Medicine, TU Dresden, Fetscherstrasse 74, 01307 Dresden, Germany 2 Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Martinistrasse 52, 20251 Hamburg, Germany 3 Institute for Immunology, Faculty of Medicine, TU Dresden, Fetscherstrasse 74, 01307 Dresden, Germany 4 Laboratory of Dendritic Cell Biology, Department of Dermatology, Research Module II, University Hospital of Erlangen, Friedrich-Alexander Universita ¨ t Erlangen-Nu ¨ rnberg, Hartmannstrasse 14, 91052 Erlangen, Germany 5 Department of Paediatric Cardiac Surgery, University Hospital of Erlangen, Friedrich-Alexander Universita ¨ t Erlangen-Nu ¨ rnberg, Loschgestrasse 15, 91054 Erlangen, Germany 6 DKMS Lifeline Cord Blood Bank, Blasewitzer Strasse 43, 01307 Dresden, Germany 7 Department of Hematology/Oncology, University Hospital, TU Dresden, Fetscherstr 74, 01307 Dresden, Germany 8 Paul Ehrlich Institut, Federal Institute for Vaccines and Biomedicines, Paul-Ehrlich-Strasse 51-59, 63225 Langen, Germany 9 Co-first authors 10 Present address: Children’s Hospital, Laboratory for Molecular Biology, Charite Universita ¨ tsmedizin Berlin, Zieglerstrasse 5-9, 10117 Berlin, Germany *Correspondence: [email protected] http://dx.doi.org/10.1016/j.stem.2014.06.001 SUMMARY In-depth analysis of the cellular and molecular mech- anisms regulating human HSC function will require a surrogate host that supports robust maintenance of transplanted human HSCs in vivo, but the currently available options are problematic. Previously we showed that mutations in the Kit receptor enhance engraftment of transplanted HSCs in the mouse. To generate an improved model for human HSC trans- plantation and analysis, we developed immune-defi- cient mouse strains containing Kit mutations. We found that mutation of the Kit receptor enables robust, uniform, sustained, and serially transplant- able engraftment of human HSCs in adult mice without a requirement for irradiation conditioning. Using this model, we also showed that differential KIT expression identifies two functionally distinct subpopulations of human HSCs. Thus, we have found that the capacity of this Kit mutation to open up stem cell niches across species barriers has sig- nificant potential and broad applicability in human HSC research. INTRODUCTION Genetically altered mouse models are useful tools for functional analysis of the biology of human hematopoietic stem and pro- genitor cells (HSPCs) after the transplantation of HSPCs or hu- man hematopoietic stem cells (HSCs), a condition referred to as humanization (Doulatov et al., 2012; Shultz et al., 2012). After humanization these mouse models make it possible to study complex biological processes such as cell fate decisions, line- age commitment, or immune function in an almost physiological setting without a need for invasive procedures in healthy volun- teers or patients. A number of different mouse mutants have been developed for human HSPC functional analysis (Shultz et al., 2012; Willinger et al., 2011). The most commonly used strains are immune-defi- cient mice on BALB/c or NOD genetic backgrounds lacking T, B, and NK cells. The different strains vary in terms of the quality of engraftment of human HSCs (Shultz et al., 2012). In general, NOD-based recipient strains (e.g., NOD Prkdc / Il2rg / , NSG) support higher levels of human HSC engraftment compared to BALB/c-based recipients (e.g., BALB/c Rag1 / Il2rg / , BRg) (Brehm et al., 2010), apparently as a result of poly- morphisms in the signal-regulatory protein alpha (Sirpa) gene, which encodes a receptor that abrogates the phagocytosis of human cells. BRg mice carrying a human Sirpa transgene are able to show significantly enhanced human chimerism relative to wild-type, but successful engraftment still requires irradiation conditioning before HSC transplantation into newborn recipient mice (Strowig et al., 2011). Thus, although the survival of human HSCs was increased in these recipients, the availability of appro- priate niches seemed to remain a limiting factor for successful engraftment. Alternative approaches to supporting long-term human HSC engraftment have included overexpression of membrane-bound human stem cell factor (SCF) (Brehm et al., 2012; Takagi et al., 2012) and knockin of human thrombopoietin (Rongvaux et al., 2011). Increased human chimerism was obtained in both cases, but engraftment still depended on irradiation therapy of the newborn hosts before transplantation (Brehm et al., 2012; Rong- vaux et al., 2011; Takagi et al., 2012). Robust and long-term maintenance of human HSCs after transplantation into adult Cell Stem Cell 15, 227–238, August 7, 2014 ª2014 Elsevier Inc. 227

Transcript of Kit Regulates HSC Engraftment across the Human-Mouse Species Barrier

Page 1: Kit Regulates HSC Engraftment across the Human-Mouse Species Barrier

Cell Stem Cell

Resource

Kit Regulates HSC Engraftmentacross the Human-Mouse Species BarrierKadriye Nehir Cosgun,1,9 Susann Rahmig,1,9 Nicole Mende,1 Soren Reinke,1 Ilona Hauber,2 Carola Schafer,2

Anke Petzold,3 Henry Weisbach,1,10 Gordon Heidkamp,4 Ariawan Purbojo,5 Robert Cesnjevar,5 Alexander Platz,6

Martin Bornhauser,7 Marc Schmitz,3 Diana Dudziak,4 Joachim Hauber,2 Jorg Kirberg,8 and Claudia Waskow1,*1Regeneration in Hematopoiesis and Animal Models in Hematopoiesis, Institute for Immunology, Faculty of Medicine, TU Dresden,Fetscherstrasse 74, 01307 Dresden, Germany2Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Martinistrasse 52, 20251 Hamburg, Germany3Institute for Immunology, Faculty of Medicine, TU Dresden, Fetscherstrasse 74, 01307 Dresden, Germany4Laboratory of Dendritic Cell Biology, Department of Dermatology, Research Module II, University Hospital of Erlangen,Friedrich-Alexander Universitat Erlangen-Nurnberg, Hartmannstrasse 14, 91052 Erlangen, Germany5Department of Paediatric Cardiac Surgery, University Hospital of Erlangen, Friedrich-Alexander Universitat Erlangen-Nurnberg,Loschgestrasse 15, 91054 Erlangen, Germany6DKMS Lifeline Cord Blood Bank, Blasewitzer Strasse 43, 01307 Dresden, Germany7Department of Hematology/Oncology, University Hospital, TU Dresden, Fetscherstr 74, 01307 Dresden, Germany8Paul Ehrlich Institut, Federal Institute for Vaccines and Biomedicines, Paul-Ehrlich-Strasse 51-59, 63225 Langen, Germany9Co-first authors10Present address: Children’sHospital, Laboratory forMolecular Biology, Charite Universitatsmedizin Berlin, Zieglerstrasse 5-9, 10117Berlin,Germany*Correspondence: [email protected]://dx.doi.org/10.1016/j.stem.2014.06.001

SUMMARY

In-depth analysis of the cellular andmolecular mech-anisms regulating human HSC function will require asurrogate host that supports robust maintenanceof transplanted humanHSCs in vivo, but the currentlyavailable options are problematic. Previously weshowed that mutations in the Kit receptor enhanceengraftment of transplanted HSCs in the mouse. Togenerate an improved model for human HSC trans-plantation and analysis, we developed immune-defi-cient mouse strains containing Kit mutations. Wefound that mutation of the Kit receptor enablesrobust, uniform, sustained, and serially transplant-able engraftment of human HSCs in adult micewithout a requirement for irradiation conditioning.Using this model, we also showed that differentialKIT expression identifies two functionally distinctsubpopulations of human HSCs. Thus, we havefound that the capacity of this Kit mutation to openup stem cell niches across species barriers has sig-nificant potential and broad applicability in humanHSC research.

INTRODUCTION

Genetically altered mouse models are useful tools for functionalanalysis of the biology of human hematopoietic stem and pro-genitor cells (HSPCs) after the transplantation of HSPCs or hu-man hematopoietic stem cells (HSCs), a condition referred toas humanization (Doulatov et al., 2012; Shultz et al., 2012). After

humanization these mouse models make it possible to studycomplex biological processes such as cell fate decisions, line-age commitment, or immune function in an almost physiologicalsetting without a need for invasive procedures in healthy volun-teers or patients.A number of different mousemutants have been developed for

human HSPC functional analysis (Shultz et al., 2012; Willingeret al., 2011). The most commonly used strains are immune-defi-cient mice on BALB/c or NOD genetic backgrounds lacking T, B,and NK cells. The different strains vary in terms of the quality ofengraftment of human HSCs (Shultz et al., 2012). In general,NOD-based recipient strains (e.g., NOD Prkdc!/! Il2rg!/!,NSG) support higher levels of human HSC engraftmentcompared to BALB/c-based recipients (e.g., BALB/c Rag1!/!

Il2rg!/!, BRg) (Brehm et al., 2010), apparently as a result of poly-morphisms in the signal-regulatory protein alpha (Sirpa) gene,which encodes a receptor that abrogates the phagocytosis ofhuman cells. BRg mice carrying a human Sirpa transgene areable to show significantly enhanced human chimerism relativeto wild-type, but successful engraftment still requires irradiationconditioning before HSC transplantation into newborn recipientmice (Strowig et al., 2011). Thus, although the survival of humanHSCswas increased in these recipients, the availability of appro-priate niches seemed to remain a limiting factor for successfulengraftment.Alternative approaches to supporting long-term human HSC

engraftment have included overexpression of membrane-boundhuman stem cell factor (SCF) (Brehm et al., 2012; Takagi et al.,2012) and knockin of human thrombopoietin (Rongvaux et al.,2011). Increased human chimerism was obtained in both cases,but engraftment still depended on irradiation therapy of thenewborn hosts before transplantation (Brehm et al., 2012; Rong-vaux et al., 2011; Takagi et al., 2012). Robust and long-termmaintenance of human HSCs after transplantation into adult

Cell Stem Cell 15, 227–238, August 7, 2014 ª2014 Elsevier Inc. 227

Page 2: Kit Regulates HSC Engraftment across the Human-Mouse Species Barrier

nonconditioned recipient mice remains a challenge and hampersthe study of human HSC function under steady-state conditionsusing mouse models.

It is not clear whether modulation of specific signals or regula-tory molecules would allow for more efficient replacement ofmouse HSPCs by their human counterparts in transplantationstudies. Previous chimera analyses in mice have shown thatthe introduction of loss-of-function Kit alleles allows efficientHSC engraftment in nonconditioned allogeneic recipient mice,possibly because functional impairment of the endogenousHSCs means that an irradiation conditioning step is not neces-sary (Waskow et al., 2009). To investigate whether reduced Kitfunction could have a similar effect for human HSC transplanta-tion, we introduced a mutant Kit allele into immune-deficientmouse strains on the BALB/c (BRg KitWv/Wv, BRgWv) or NOD(NSG KitWv/Wv, NSGWv; NSG KitWv/+, NSGWv/+; and NSGKitW41/W41, NSGW41) genetic backgrounds and analyzedwhether the HSC niche becomes permissive for human donorHSCs in these animals. We found that human HSCs engraft effi-ciently in these mice, even in adult nonconditioned recipients,leading to sustained multilineage contribution that was main-tained through serial transplantation. Thus, mice with mutationsin the Kit receptor are effective tools for analysis of human HSPCfunction with significantly broader applicability than previouslyexisting models.

RESULTS

Efficient Engraftment of Human HSPCs into Kit MutantMice without Irradiation PreconditioningWith currently available models, successful xenogeneic humanHSC transplantation into mice requires irradiation conditioningbefore transplantation. Efficient engraftment without condition-ing would be advantageous because irradiation has systemictoxic effects on many cell types and impairs HSC function bydirect (Shao et al., 2014; Shen et al., 2012) and indirect (Carbon-neau et al., 2012) mechanisms that may also impair the functionof donor HSCs (Nilsson et al., 1997). To investigate whether func-tional impairment of the endogenous HSC compartment via mu-tation of Kit could provide an advantage for the engraftment ofhuman HSCs, we bred a loss-of-function Kit allele (KitWv) into aBALB/c Rag2- and Il2rg-deficient background already used forhuman HSC transplantation (BALB/c Rag2! Il2rg! KitWv/Wv

mice, referred to as BRgWv).We then tested the engraftment po-tential of these mice by transplantation of CD34+-enrichedHSPCs from human cord blood into adult BRgWv mice withoutprior irradiation conditioning, and we compared the engraftmentto similar mice lacking the Kit mutation that were transplantedwith and without prior irradiation. Chimerism 15 weeks aftertransplantation was substantially higher in the blood of BRgWvrecipient mice compared to BRg recipients (Figure 1A) andremained higher over a time period of 27 weeks (Figure 1B).To assess whether the introduction of the mutant Kit receptoropens up the stem cell niches for human HSCs irrespectiveof the genetic background of the recipient, we compared thecontribution of human leukocytes to the blood of adult C57BL/6 Rag2!/! Il2rg!/! KitWv/Wv (similar to the protocol in Waskowet al., 2009) or BRgWv (Figure 1C) recipient mice. Human leuko-cytes were readily detected in BRgWv, but not C57BL/6Rag2!/!

Il2rg!/! KitWv/Wv, mice (Figure 1C), suggesting that differences inthe Sirpa allele (Strowig et al., 2011; Takenaka et al., 2007) orother genetic modifier loci present in the BALB/c mouse strainare advantageous for human HSPC engraftment in this context.The frequencies of human leukocytes in the bone marrow of

BRgWv mice were uniformly very high and remained high overtime (Figure 1D). Notably, the engraftment rate at late time pointsafter transplantation was significantly increased compared toirradiated NSG recipient mice (one of the best existing mousemodels; Shultz et al., 2012). Consistently, human leukocytenumbers were higher in the bone marrow of BRgWv recipients(Figure 1E), and engraftment was detected after the transplanta-tion of as few as 1.2 3 104 human donor HSPCs (Figure 1F).Thus, our data suggest that introduction of a mutant Kit allele

confers increased receptiveness for human cell engraftment inthe blood and bone marrow of recipient mice, and that thisreceptiveness is improved to a sufficient extent to mean that effi-cient engraftment can be achieved in adult recipient micewithout irradiation conditioning before transplantation.

SustainedMultilineageEngraftment byHumanHSPCs inBRgWv Recipient MiceTo assess which human cell types were generated in the bonemarrow of recipient mice after transplantation, we analyzed therecipient mice for the presence of human lymphoid, myeloid,and erythroid lineage cells 11–37 weeks after human HSPCtransplantation into adult mice (Figure 2). Only B cells were de-tected in the bonemarrow of nonconditioned BRgmice, and irra-diation led to the additional appearance of monocytes andgranulocytes (Figure 2A). However, we were able to detect thesecell types at high frequencies after transplantation into nonirradi-ated BRgWv recipient mice (Figure 2B). Detailed analyses ofmyeloid cell types by flow cytometry (Figure 2C) combinedwith morphological analyses confirmed the cellular identities(Figure 2D) and revealed elevated repopulation of myeloid celltypes in BRgWv recipients compared to irradiated NSG andBRg mice (Figure 2E). The composition of the myeloid cell typesof BRgWv and irradiated NSG recipient mice was comparable(Figure 2F). These data suggest that nonconditioned BRgWvmice support robust human myelopoiesis.Human HSPCs transplanted into irradiated NSG mice can

give rise to erythrocytes, but this population has not yet beendescribed in strains with a BALB/c genetic background. In ourexperiments, however, we were able to detect human erythroidlineage cells in the bone marrow of 20 out of 29 BRgWv mice,while only 8 out of 17 irradiated NSG recipients showed engraft-ment with human erythroid cells (Figure 2G). Overall, therefore,BRgWv recipient mice show long-term repopulation by myeloidand erythroid cell types after human HSPC transplantation athigher frequency than has been obtained thus far with existingmodels.

De Novo Generation of Human Lymphocytes inTransplanted BRgWv MiceTo test for continuous hematopoiesis from transplanted HSCs,we analyzed B and T cell development in the bone marrow andthymus, respectively, in the recipient mice. The developmentalstages of B cell development are defined by the status of D-Jor V-DJ recombination of the immunoglobulin heavy chain

Cell Stem Cell

Human HSC Engraftment in Adult Kit Mutant Mice

228 Cell Stem Cell 15, 227–238, August 7, 2014 ª2014 Elsevier Inc.

Page 3: Kit Regulates HSC Engraftment across the Human-Mouse Species Barrier

genes, which in turn is associated with defined expression ofCD10, CD19, CD34, IgM, and IgD on the cell surface (Ghiaet al., 1996) (Figure 3A). In the bone marrow of reconstitutedBRgWv mice, we found that the CD19+CD10+CD34!IgM! preB cell compartment was overrepresented at the expense ofmature B cells (Figure 3B), suggesting that there is a defectin B cell maturation that may be a result of species-specificincompatibilities.We also found that the mature T cell pool in the bone marrow

was small, but thymocyte numbers were greater in BRgWvmice relative to control recipient mice, suggesting ongoingT cell development (Figure 1E). In fact, the sizable fraction ofCD4+CD8+ (double positive, DP) human thymocytes that weobserved in BRgWv mice (Figures 3C and 3D) is similar to thesituation seen in transplanted irradiated NSGmice and in thymusbiopsies from human infants. We also detected DP thymocytesafter transplanting cell-sorter purified human lineage!CD38!

CD34+ cells into nonconditioned BRgWv and irradiated NSGrecipient mice (Figure 3D, open symbols), suggesting that these

Figure 1. Elevated Human White Blood CellChimerism in the Bone Marrow of BRgWvMice following Human HSPC Transplan-tation(A) Dot plots show murine (mCD45) and human

(hCD45) leukocytes in nonconditioned BRgWv

and control BRg recipient mice that had received

33 105 human CD34-enriched cord-blood HSPCs

15 weeks earlier. Donor cell preparation and

number is the same for Figures 1B–1E.

(B) Frequencies of human leukocytes in the blood

of humanized BRgWv (n = 23), irradiated BRg

(n = 9), and nonconditioned BRg (n = 7) mice over

28 weeks of time.

(C) Frequencies of human leukocytes in the blood

of BALB/c RgWv (n = 5) or C57BL/6 RgWv (n = 4)

recipient mice treated as in (A).

(D) Frequencies of human leukocytes in the bone

marrow of BRgWv and control recipient mice

analyzed 17–24 weeks (left) or 25–37 weeks (right)

after transplantation of human HSPCs.

(E) Human leukocyte numbers in two femurs (left),

the spleen (middle), or in the thymus (right) of hu-

manized mice 17–37 weeks after transplantation.

Open symbols represent values detected in recip-

ient mice that were transplanted with sorter-puri-

fied human lineage!CD38!CD34+ cells.

(F) Frequencies of human leukocytes in the bone

marrow of BRgWv mice that had received indi-

cated numbers of human CD34-enriched HSPCs

19–24 weeks before.

cells could be generated de novo from en-grafted human HSCs. Clear evidence forde novo T cell generation in BRgWv micecame from the analysis of T cell receptorexcision circles (TRECs) in the thymocytesof the recipient mice (Figure 3E). Thymo-cytes from repopulated thymi, but notfrom control thymi, could proliferate inresponse to polyclonal T cell stimulation(Figure 3F).

To confirm that the continuous human hematopoiesis thatwe observed was based on HSC activity and not on the differ-entiation of cotransplanted progenitor cells, we transplantedsorter-purified lineage!CD38!CD34+ cord blood HSPCs anddetermined their multilineage differentiation potential. We founddonor-derived myeloid and erythroid cells in the bone marrow(Figures 3G–3I) and short-lived T cell precursors in the thymusas described above (Figure 3D, open symbols), suggestingthat at the time point of analysis de novo hematopoiesis wasoccurring. Overall, therefore, we conclude that continuous hu-man lymphopoiesis is supported in BRgWv recipient mice.

Enhanced Self-Renewal of Human HSCs in Mice Is aFunction of Mutant KitWe next looked at whether the mouse bone marrow microenvi-ronment in Kit mutant recipients is permissive for the self-renewal of human HSPCs by examining the localization andphenotype of the transplanted cells (Figure 4A). We found thathuman HSPCs engrafted in the bone marrow, but not in the

Cell Stem Cell

Human HSC Engraftment in Adult Kit Mutant Mice

Cell Stem Cell 15, 227–238, August 7, 2014 ª2014 Elsevier Inc. 229

Page 4: Kit Regulates HSC Engraftment across the Human-Mouse Species Barrier

Figure 2. Multilineage Repopulation of Humanized Mice(A) Human cell types that engrafted in the bone marrow of indicated mouse strains after the transplantation of 3 3 105 human CD34-enriched HSPCs 29 weeks

earlier. Human CD3+ T, CD19+ B, SSChi granulocyte, and CD14+ monocyte cell populations were superimposed on SSC/FSC dot-plots. Donor cell preparation

and number is the same throughout Figure 2.

(B) Relative composition of human-graft-derived leukocytes in the bone marrow of recipient mice of the indicated genotypes that received human HSPC 11–

37 weeks earlier. For comparison the relative composition of BALB/c murine bonemarrow is shown. Human cells were identified as CD3+ T, CD19+ B, and CD33+

myeloid cells, and immature cells lacking the expression of CD3, CD19, and CD33. Mouse cell types were identified as CD45R+ B cells, CD3+ T cells, CD11b+

myeloid cells, and lineage! (lineage = CD3 CD11b CD19) immature cells. Number of mice analyzed per group is indicated above each column.

(C) Identification of indicated cell types in the bone marrow of humanized BRgWv mice 27 weeks after humanization (top) and control human bone marrow

(bottom). Arrow indicates population that is shown in the next dot plot. pDC, plasmacytoid dendritic cells; cDC, conventional DC; Baso, basophil granulocyte; Eo,

eosinophilic granulocyte; MC, mast cell; PMN, polymorph nucleated neutrophil; NP, neutrophil precursors.

(D) Pictures show May-Grunwald-Giemsa stained cells of the indicated lineages (purified as described in A and C) purified from human bone marrow and from

humanized BRgWv mice that were transplanted with human HSPCs 14 weeks earlier. Size bar applies to all photographs.

(E) Plot shows frequencies of human myeloid cells (CD33+ cells) in the bone marrow of indicated humanized mice that were transplanted with human HSPCs

20–37 weeks before.

(F) Composition of human myeloid cells in irradiated NSG and BRgWv recipient mice.

(G) Frequencies of human erythroid cells (hCD45!mCD45!Ter119!hCD235+) in humanized mice 14–37 weeks after transplantation.

Cell Stem Cell

Human HSC Engraftment in Adult Kit Mutant Mice

230 Cell Stem Cell 15, 227–238, August 7, 2014 ª2014 Elsevier Inc.

Page 5: Kit Regulates HSC Engraftment across the Human-Mouse Species Barrier

Figure 3. Lymphocyte Differentiation in Humanized BRgWv Mice(A) B cell development in human bone marrow (top) or in bone marrow and spleen of BRgWv mice (middle and bottom, respectively) that had received 3 3 105

human CD34-enriched HSPCs 28 weeks before. Dot plots depict the expression of CD19 and CD10 on human leukocytes (hCD45+ SSClo, left), the expression of

IgM and CD34 on CD19 CD10 double positive cells (middle), and the expression of IgM and IgD on CD19 single positive cells (right). Donor cell preparation and

number is the same in Figures 3B and 3F.

(B) Frequencies of pro-B cells (BI, CD10+CD19+CD34+IgM!), pre-B cells (BII, CD10+CD19+CD34!IgM!), immature B cells (BIII, CD10+CD19+CD34!IgM+), and

mature B cells (BIV, CD10!CD19+CD34!IgM+) of total B cells (CD19+) in the femurs of BRgWv or irradiated BRg mice transplanted with human HSPCs

21–32 weeks earlier.

(C) Dot plots show the expression of CD4 and CD8 on hCD45+ CD3+ thymocytes of a humanized BRgWv mouse 23 weeks after transplantation and human

thymus control (donor age: 5 months). Data shown are representative for five independent experiments.

(D) Plot shows the frequency of CD4+CD8+ double positive thymocytes in humanized mice of the indicated genotype that had received 33 105 CD34-enriched

(closed symbols) or 3 3 104 sorted (lineage!CD38!CD34+, open symbols) human HSPCs 16–31 weeks before.

(E) Relative frequency of T cell receptor excision circles (TRECs) in thymocytes of humanized BRgWv mice that were transplanted as described in (D). TREC

frequencies were determined in purified T cells (CD3+) from human cord blood (CB) and blood (PB) samples from adult donors (age: 44–58 years) for control.

(F) Plot shows 3H-thymidine incorporation into thymocytes after in vitro trigger with PHA. Thymocytes from transplanted BRgWv (Tx) mice, but not from non-

transplanted control BRg (w/o) mice, expanded after polyclonal trigger.

(G) Dot plot shows repopulated bone marrow 17 weeks after transplantation of 3 3 104 sorter-purified lineage!CD38!CD34+ human HSPCs (purity: 99.8% ±

0.23%, three independent experiments; human CD45+ cells: 59.4% ± 28%, n = 6 mice.)

(H)Graph shows composition of human bonemarrow graft 16, 17, and 30weeks after transplantation as described in (G). Data shown are as described in Figure 2B.

(I) Sorter-purified donor cells give rise to erythroid cells.

Cell Stem Cell

Human HSC Engraftment in Adult Kit Mutant Mice

Cell Stem Cell 15, 227–238, August 7, 2014 ª2014 Elsevier Inc. 231

Page 6: Kit Regulates HSC Engraftment across the Human-Mouse Species Barrier

spleen, of BRgWv mice. Donor cell engraftment in the bonemarrow was more efficient in BRgWv mice than in irradiatedBRg or NSG recipients (Figure 4B). Recent analyses haveindicated that within the CD38!CD34+ compartment, humanHSCs can be more precisely defined as CD90+CD45RA!

cells (Majeti et al., 2007) (Figure 4C). The frequency oflineage!CD38!CD34+CD90+CD45RA! HSCs was significantlyhigher in the bone marrow of BRgWv recipients compared toirradiated NSG recipients (Figure 4D). In fact, frequencies of hu-man HSCs in reconstituted BRgWv mice were equivalent to thatin human bonemarrow, suggesting that the HSCniche in BRgWvmice is suitable for the maintenance of human HSCs (Figure 4D).Increased maintenance of human HSCs was also observed aftertransplantation into newborn BRgWv mice, suggesting that theimproved accessibility of niche space for human HSCs brought

Figure 4. Maintenance of Human HSCs inMice(A) Dot plots show human HSPCs (CD38!CD34+)

within immature cells (lineage!, cells lacking CD2

CD3 CD10 CD11b CD14 CD15 CD16 CD19

GlyA expression) of human origin (hCD45+) in the

bone marrow of nonconditioned BRgWv or non-

conditioned or irradiated BRg or NSG recipient

mice, and in a human bone marrow control sample.

Recipient mice were analyzed 19 weeks after

transplantation of 3 3 105 human CD34-enriched

HSPCs. Donor cell preparation and number is the

same in Figures 4B–4E.

(B) Plot shows numbers of human HSPCs (hCD45+

lineage!CD34+CD38!) in two femurs of humanized

indicated mouse strains transplanted 11–37 weeks

before.

(C) Human HSPCs were further analyzed for the

expression of CD90 and CD45RA in indicated hu-

manized recipient mice and human bone marrow

control.

(D) Plot shows frequencies of human HSCs

(% lineage!CD38!CD34+CD90+CD45RA!) in indi-

cated mouse strains and human control samples.

(E) Plot shows numbers of human HSPCs in the

bone marrow 27–32 weeks after humanization of

newborn mice with 3 3 105 CD34-enriched cord

blood cells.

(F) Dot plots show engrafted human cell types in the

bonemarrow of secondary recipientmice 13weeks

after transplantation of 107 total bone marrow cells

from primary recipient mice. Cells were identified

as shown in Figure 2A.

(G) Frequencies of human CD45+ cells in the bone

marrow of secondary recipient mice 11–20 weeks

after secondary transplantation of 1 to 23 107 total

bone marrow cells from primary recipient mice. The

number of positively repopulated mice out of all

secondary recipients is indicated.

about by the Kit mutation is independentof the age of the recipient mice (Figure 4E).

To determine whether these phenotypi-cally defined human HSCs were func-tional, we conducted serial transplantationof total bone marrow cells from primaryrecipient mice into secondary recipients.

Secondary transplantation into BRgWv mice was successfuland led to the presence of detectable human B cells, NK cells,monocytes, and granulocytes. By contrast, we found mainly Bcells in the bone marrow of irradiated BRg secondary recipients(Figure 4F). The number of successfully repopulated secondaryBRgWv recipient mice (18 out of 28 recipients) was greaterthan the success rate with irradiated BRgmice (5 out of 13 recip-ients) receiving a graft from primary recipient mice of the samegenotype (Figure 4G). Moreover, the frequency of human leuko-cytes in the bone marrow was increased in secondary BRgWvrecipients compared to secondary irradiated BRg recipients,confirming the improved maintenance of functional multipotenthuman HSCs in primary BRgWv recipients.Taken together our data show efficient and effective mainte-

nance of phenotypic and functional human HSCs in the bone

Cell Stem Cell

Human HSC Engraftment in Adult Kit Mutant Mice

232 Cell Stem Cell 15, 227–238, August 7, 2014 ª2014 Elsevier Inc.

Page 7: Kit Regulates HSC Engraftment across the Human-Mouse Species Barrier

marrow of immune-deficient recipient mice with Kit receptor mu-tations, and that the level of functional engraftment that can beachieved in these mice is significantly improved relative to exist-ing host mouse strains.

Mechanism of Human Stem Cell MaintenanceTo assess whether the engrafted human immature hematopoiet-ic cells replace precursor cells of murine origin, we determinedthe number ofmouse progenitors in the bonemarrow of recipientmice (Figure 5A). We found that the murine progenitors weresignificantly reduced in numbers in the bone marrow of trans-planted BRgWv recipients, but not in irradiated NSG or BRgrecipients, suggesting that the clone size of human HSPCsincreased in humanized BRgWv mice because human donorcells are able to compete effectively with the endogenousmousecells. Thus, human cells with a fully functional KIT receptor mayhave a proliferative advantage over mouse HSPCs expressing amutant Kit allele.The increased engraftment rates in the BRgWv mice were in-

dependent of any alteration of homing activities (Figure 5B) orof changes in the availability of the ligand for the Kit receptor,SCF, (Figures 5C and 5D), which can modulate the engraftmentof human donor cells in mouse models (Brehm et al., 2012;Takagi et al., 2012). The protein level of SCF was comparablebetween Kit mutant and wild-type recipient mice (Figure 5D).Also, the relative abundance of SCF transcripts encodingthe membrane-bound or soluble form of SCF in marrow mesen-chymal stromal cells, the nonhematopoietic niche cell fraction

Figure 5. Kit Receptor Regulates Competi-tion between Human and Mouse HSCs andHPCs(A) Plot shows immature mouse bone marrow

cell numbers (CD3!CD11b!CD19!CD45R!Gr-

1!NK1.1!Ter119!) in nontransplanted (w/o Tx) or

indicated humanized mouse strains (Tx) that had

received 3 3 105 human CD34-enriched HSPCs

12–37 weeks earlier.

(B) Homing of human CD34-enriched HSPC cells

into the bone marrow of indicated recipient mice

within 18 hr after transplantation.

(C) Relative mRNA expression levels of the trans-

membrane (tm) or soluble (s) forms of SCF in niche

cells (CD45!Ter119!CD31!Pdgfra+CD51+) from

nontransplanted BRgWv and BRg mice. Each dot

represents the mean of triplicate analysis from one

mouse.

(D) Concentration of stem cell factor (SCF) in blood

and bone marrow of nontransplanted mice of the

indicated genotypes determined by ELISA. Each

dot represents the mean of duplicate analysis from

one mouse. As a positive control, increased SCF

protein levels in complete Kit null mice are shown

(KitW/W hEPO-tg; Waskow et al., 2004).

that expresses SCF, was similar in micecarrying wild-type or mutant Kit alleles(Figure 5C). These findings suggest thatthe increased engraftment of humanHSCs is based on impaired murineHSC function that results in increased

accessible niche space for donor humanHSCs, and that it allowshuman HSCs to out-compete endogenous HSCs for nichespace.

Expression of Mutant Kit Increases the Receptivenessof NSG Mice for Human HSC EngraftmentTo investigate the capacity of BRgWv mice for human HSCengraftment, we looked at transplantation of very high donorHSC numbers and found elevated engraftment compared to irra-diated NSGmice (Figure 1D), suggesting that BRgWv mice havemore available niche space for human HSCs than NSG mice do.Consistently, we find increased endogenous HSC numbers innontransplanted, Kit wild-type BRg mice compared to NSGmice (Figure S1A available online), suggesting that immune-defi-cient mice on the BALB/c genetic background have more HSCniches. To compare the engraftment of human donor cells undernonsaturating conditions, we looked at transplantation of limitednumbers of donor HSPCs. In this experiment we saw increasedengraftment of human leukocytes in irradiated NSG micecompared to nontreated BRgWv recipients (Figure 6A), prompt-ing us to test whether mutant Kit also confers an advantage forthe engraftment of human HSCs in NOD-based mouse strains.We generated three more mouse strains: NSG KitWv/+ (termed

NSGWv/+), NSG KitWv/Wv (termed NSGWv), and NSG KitW41/W41

(termed NSGW41), and we also transplanted them with humanHSPCs. A large fraction of the NSGWv mice became moribundand were not available for analysis (Figure 6B). We continued touse the NSGWv/+ and NSGW41 mouse strains, which survive

Cell Stem Cell

Human HSC Engraftment in Adult Kit Mutant Mice

Cell Stem Cell 15, 227–238, August 7, 2014 ª2014 Elsevier Inc. 233

Page 8: Kit Regulates HSC Engraftment across the Human-Mouse Species Barrier

humanization for long time periods. NSGW41 and NSGWv/+ re-cipients showed very uniform and robust engraftment with hu-man donor cells, and blood chimerism reflected the engraftmentfrequency in the bone marrow (Figure 6C). In NSGW41 mice, wesaw increased generation of myeloid cells (Figure 6D) and animproved balance between T and B lymphocytes (Figure 6E)compared to standard irradiated NSG recipients. However, asin the BRgWv mice, cells of the B lymphoid lineage were alsoblocked in their terminal differentiation (data not shown). Trans-plantation of titrated numbers of donor cells into irradiated NSGandnonconditionedNSGW41mice revealed an increased recep-tivenessofNSGW41micecompared to irradiatedNSGrecipients(Figure 6F), similar to the situation that we saw when comparingBRgWv to irradiated BRg mice. Consistently, the SCID repopu-lating cell (SRC) activity in CD34-enriched HSPCs was foundto be increased in NSGW41 recipient mice (irradiated NSG: 1 in4,732, NSGW41: 1 in 514; p = 0.001), suggesting an enhancedsensitivity for the engraftment of human HSCs. Thus, it seemsthat Kit mutation also enables efficient human HSC engraftment

Figure 6. Mutant Kit Confers IncreasedReceptiveness on the NOD Genetic Back-ground(A) Plot shows human leukocyte frequencies in the

bone marrow of BRgWv and irradiated NSG

recipient mice 30 weeks after the transplantation

of indicated titrated numbers of sorted human

lineage!CD38!CD34+ cord blood HSPCs.

(B) Survival plot for humanized nonconditioned

BRgWv, NSGWv/+, NSGWv, and NSGW41

recipient mice after the transplantation of 3 3 105

(BRgWv) or 3 to 5 3 104 (NSGWv, NSGWv/+,

NSGW41) CD34-enriched human HSPCs.

(C) Frequencies of human leukocytes in the blood

as a function of the frequency of human leukocytes

in the bonemarrow for the indicatedmouse strains

15–40 weeks after transplantation of 3 3 105

(closed circles) or 3 to 53 104 (open circles) CD34-

enriched human HSPCs.

(D) Frequency of human myeloid cells within the

bone marrow of humanized mice 15–40 weeks

after the transplantation of 3 to 5 3 104 CD34-

enriched cord blood cells.

(E) Plot shows relative composition of human-

graft-derived leukocytes in the bone marrow of

recipient mice 15–40 weeks after transplantation

of 3 to 5 3 104 CD34-enriched human HSPCs.

(F) Kinetic of the repopulation in the blood of

NSGW41 and irradiated NSG recipient mice after

the transplantation of 103 (left) or 104 (right) human

CD34-enriched HSPCs. Data from two indepen-

dent experiments are pooled (103 donor cells were

transplanted in six irradiated NSG and eight

NSGW41 mice; 104 donor cells were transplanted

in five irradiated NSG and seven NSGW41 mice).

See also Figure S1.

in the immune-deficient NOD geneticbackground without a requirement forirradiation preconditioning.To test whether the increased levels of

engraftment seen in the Kit mutant mousestrains makes them suitable for studies

using the human immunodeficiency virus type 1 (HIV-1), werepopulated newborn irradiated NSG mice and adult noncondi-tioned NSGWv/+ and NSGW41 mice using StemReginin1-expanded human HSCs, and we infected them 20 weeks laterwith HIV. The viral load was determined 4, 7, and 9 weeks post-infection and proved to be comparable between all three human-izedmouse strains (Figure S1B). This analysis therefore suggeststhat the NSGWv/+ and NSGW41 mouse strains would be highlysuitable for studies using HIV.

Adoptive Transfer of Prospectively Separated HumanStem Cell Populations Reveals Two FunctionalSubpopulations in the Human HSC PoolWe then looked at whether NSGWv/+ recipientmice can be usedto assay human HSC function. Subtle differences in the densityof Kit receptor expression on the surface of murine HSCs areassociated with strikingly different functions (Grinenko et al.,2014; Shin et al., 2014). More specifically, HSCs expressing in-termediate levels of Kit (Kitint) show more potent engraftment

Cell Stem Cell

Human HSC Engraftment in Adult Kit Mutant Mice

234 Cell Stem Cell 15, 227–238, August 7, 2014 ª2014 Elsevier Inc.

Page 9: Kit Regulates HSC Engraftment across the Human-Mouse Species Barrier

potential, and the Kitint and Kithi (high levels of Kit) populationsrepresent consecutive developmental stages. Thus, the transi-tion from Kitint to Kithi HSCs indicates the initiation of differentia-tion (Grinenko et al., 2014). To assesswhether the level of KIT cellsurface receptor expression also allows the prospective separa-tion of two discrete HSCpopulations in humans, we transplantedsorted cord blood HSCs expressing high or intermediate levelsof KIT into nonconditioned NSGWv/+ mice (Figure 7A). HumanHSCs expressing high densities of KIT showed higher repopula-tion activity for human leukocytes in the blood of the recipientmice over time compared to KITint human-cord-blood-derivedHSCs (Figure 7B). This higher contribution in the blood wasbased on more efficient engraftment of human HSCs in thebonemarrow, where the progeny of KIThi humanHSCswere pre-sent at a higher frequency compared to KITint-derived humanHSCs (Figure 7C). Interestingly, this situation is reversed relativeto that seen in mouse where Kitint HSCs are the best at repopu-lation, suggesting that the role of Kit-mediated signals differs be-tween the two species generally or that it shifts during ontogenyin the same organism. We conclude that KITint and KIThi cells aretwo functionally distinct human HSC subpopulations that can bedistinguished based on their repopulation capacity in this mousemodel.

DISCUSSION

We show here that combining a loss-of-function allele of the Kitreceptor with profound immune deficiency allows the efficientand stable engraftment of human HSCs into adult recipient

mice without a need for prior irradiation. Our data suggest thatthe Kit mutation functionally impairs endogenous HSCs in away that permits efficient engraftment of human HSCs, andthey show that this approach is effective in immune-deficientstrains with BALB/c or NOD genetic backgrounds.Existing mousemodels used for xenogeneic HSC transplanta-

tion require irradiation conditioning before transplantation todeplete the endogenous HSCs and provide niche space for thedonor HSCs (Shultz et al., 2012). Engraftment without condition-ing would be advantageous because irradiation has systemictoxic effects on many proliferating cell types (Shao et al., 2014;Shen et al., 2012; Carbonneau et al., 2012), and the inducedinflammation may have a major impact on the level and qualityof engraftment and on immune-mediated complications (e.g.,graft versus host disease). Our approach was designed to facil-itate human HSC engraftment in mice by increasing the HSCniche space through impairment of the endogenous mouseHSCs by introducing a loss-of-function Kit allele. Using thisapproach, we found that human HSPCs successfully engraftedinto three strains of immune-compromised mice, BRgWv,NSGWv/+, and NSGW41, and that successful reconstitution ofsecondary recipient mice provided evidence for functional main-tenance of human HSCs. Furthermore, NSGWv/+ and NSGW41mice are very suitable mouse models for the study of HIV andpossibly other human pathogens. Taken together, we concludethat functional impairment of endogenous host murine HSCs us-ing Kit mutation conveys a competitive advantage for incominghuman stem and progenitor cells and allows the engraftmentof human HSCs into adult mice without irradiation conditioning.Engraftment of human HSCs into mice of a young age is

generally considered advantageous (Brehm et al., 2010; Shultzet al., 2007), and newborn BRg or NSG are successfully usedfor the generation of humanized mice harboring all componentsof the innate or adaptive immune system (Ishikawa et al., 2005;Ito et al., 2002; Rongvaux et al., 2014; Traggiai et al., 2004). How-ever, our experiments reveal no advantage for human HSCengraftment in newborn versus adult BRgWv recipient mice.In principle, it seemed to us that mutation of the Kit receptor

could provide an advantage for the engraftment of humanHSCs in mice through two distinct mechanisms: a direct impair-ment of endogenous HSCs that allows their replacement bydonor cells and/or increased expression of SCF caused byimpaired ligand-receptor interactions in cells expressing Kit.However, in BRgWv mice we found that SCF protein levels andtranscripts in bone marrow niche cells are unaltered, suggestingthat the increased HSC engraftment in these mice is indepen-dent of SCF expression/availability. In line with this hypothesis,prior work has shown that increased levels of human SCFcan support increased differentiation, but not engraftment ofhuman HSCs (Brehm et al., 2012). We therefore propose thatthe increased engraftment that we observe is based largelyon impaired murine HSC function, resulting in accessible nichespace for human HSCs. Thus, our experiments suggest thatthe Kit receptor is a regulator of HSC competitiveness acrossspecies.Recently a number of mouse strains have successfully been

generated that show improved engraftment of human myeloidcell types by harboring overexpression or knockin approachesfor human growth factors (Miller et al., 2013; Rongvaux et al.,

Figure 7. Two Functional Subpopulations of Human HSCs(A) Prospective separation of Kithi and Kitint human HSCs from cord blood (left

and middle) and postsort analysis of both populations before transplantation

(right).

(B) Repopulation of mouse blood by human leukocytes (human CD45+) over

the time course of 30 weeks after transplantation of sorter-purified

lineage!CD38!CD34+CD90+CD45RA! Kithi or Kitint human HSCs in NSGWv/+

recipient mice.

(C) Frequency of human HSPCs (CD38!CD34+) in the bone marrow (hCD45+)

of NSGWv/+ mice that had received purified Kithi or Kitint HSCs 30 weeks

before. Data from two independent experiments were pooled.

Cell Stem Cell

Human HSC Engraftment in Adult Kit Mutant Mice

Cell Stem Cell 15, 227–238, August 7, 2014 ª2014 Elsevier Inc. 235

Page 10: Kit Regulates HSC Engraftment across the Human-Mouse Species Barrier

2014; Shultz et al., 2012; Willinger et al., 2011). However, theincreased myeloid differentiation achieved by these approachesmay well be achieved at the expense of HSC maintenance,suggesting that human cytokine levels must be finely tuned toprovide appropriate differentiation signals and allow stem cellmaintenance at the same time (Nicolini et al., 2004). Direct com-parison to Kit wild-type mice revealed significantly increasedgeneration of myeloid cells in Kit mutant NSGW41 or BRgWv re-cipients, suggesting that improved engraftment of human HSCsin the murine stem cell niche also leads to an improved engraft-ment of myeloid cell types. Also, mouse progenitor cells in thebone marrow were largely replaced by human hematopoieticprogenitors, pointing at a proliferative advantage for humancells expressing a fully functional Kit receptor over mouse cellsexpressing mutant Kit. We conclude that increased myeloidcell contribution can also be achieved through improved HSCengraftment and a competitive proliferation advantage of humanmyeloid progenitor cells.

We repopulated nonirradiated NSGWv/+ mice with humanHSCs that were prospectively separated into cells expressinghigh and intermediate densities of the KIT receptor, and throughthis analysis we found that the human HSC pool can be subdi-vided into two cell populations that functionally differ in theirengraftment capacity. Human HSCs expressing high densitiesof KIT (KIThi) containmore potent repopulation activity comparedto HSCs expressing intermediate levels (KITint). In contrast, inmice Kitint HSCs exhibit a very efficient engraftment activity,and the reduced signaling activity by the Kit receptor seemscrucial for the maintenance of ‘‘stemness’’ in situ (Grinenkoet al., 2014). It will be interesting to determine in the futurewhether this difference is due to distinct species-specific re-quirements for Kit signaling or due to some switch altering therelevance of the level of Kit signaling during ontogeny (Kawa-shima et al., 1996).

When our data are taken together, our overall conclusionis that impairment of endogenous host HSCs by Kit receptor mu-tation leads to a competitive advantage for incoming humanstem and progenitor cells and allows efficient engraftment ofhuman HSCs into adult mice without irradiation conditioning.In this study we have generated three mouse strains, BRgWv,NSGWv/+, and NSGW41 mice, that take advantage of thisapproach and facilitate analysis of human HSC function. Basedon these results, we would expect thesemice to also be superiorto irradiated mice for the transplantation of leukemic cells, whichhave proved difficult to engraft efficiently in xenotransplantationmodels. We are planning to explore this line of investigation infuture experiments. Finally, our data also suggest that Kit mightbe an attractive target for approaches aimed at facilitating HSCengraftment without prior conditioning in human therapeuticsettings.

EXPERIMENTAL PROCEDURES

MiceNOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ mice (NSG) were obtained from Jackson

Laboratory. C57BL/6Rag2!/! Il2rg!/!KitWv/Wv (Figure 1C)mice were obtained

by intercrossing Rag2!/! Il2rg!/! KitWv/+ mice (Waskow et al., 2009), and

KitWv/Wv offspring were identified by their white coat color. BRgWv mice were

generated by backcrossing the KitWv allele (C57BL/6-Kit < Wv > ) to N10–

N20 onto the H-2d (BALB/c haplotype) Rag2tm1Alt Il2rgtm1Brn strain (Kirberg

et al., 1997), which is permissive for humanization (Scheeren et al., 2008).

The latter strain is N4 with respect to BALB/c and had been maintained

by inbreeding before the introduction of theKitWv allele.KitWv andKitW41 alleles

werebackcrossed toNSGmice for 18and10generations, respectively. Result-

ing BALB/c Rag2!/! Il2rg!/! KitWv/+ or NSG KitWv/+ mice were intercrossed to

obtain the experimental homozygous BRgWv or NSGWvmice. Also NSGWv/+

mice were used for transplantations. To genotype for Kit alleles (Kit<+>,

Kit<Wv>), PCR was performed on tail-snip DNA (forward: 50-AAAGAGAG

GCCCTAATGTCG; reverse: 50-CTCGAGACTACCTCCCACC) and the prod-

ucts were sequenced using the forward primer (Kit<Wv> harbors a C to T con-

version at position 2007; Nocka et al., 1990) or by digestion of a PCR product

with NsiI (forward: 50- CCACGCTTTGTTTTGCTAAAATGCATCAC; reverse:

50- AGGCACTGCCTAAATCATACTTTGATAACC; PCR mix volume: 11 ml). A

band of 182 bp is indicative for the wild-type allele and bands of 118 bp and

64 bp are indicative for the Kit<Wv> allele. KitW41 allele typing was by PCR

(forward: 50-AAGGAAGGTTAGAACCCCTGG; reverse: 50-AGCTCCCAGAGGA

AAATCCC). The products were sequenced using the forward primer. The

Kit<W41> allele contains a point mutation at position 2519 (G / A) (Nocka

et al., 1990). All mice were bred and maintained under specific pathogen-free

conditions at the animal facility of theDresdenUniversity of Technology. Animal

experiments were approved by the relevant authorities, the Landesdirektion

Dresden.

Human Donor CellsHuman umbilical cord blood samples were provided by the DKMS Cord Blood

Bank Dresden and by the Department of Hematology/Oncology, University

Hospital Dresden and were used in accordance with the guidelines approved

by the Ethics Committee of theDresdenUniversity of Technology. Two to six in-

dividual cord blood samples were pooled before Ficoll-Hypaque density centri-

fugation andmagnetic enrichment for CD34+ cells according tomanufacturers’

instructions (Miltenyi Biotech). Purity of CD34+ cells was on average 93%± 4%

(n = 23). In indicated experiments sorted human HSCs were transplanted. The

sort was lineage! (lineage = CD2 CD3 CD10 CD11b CD14 CD15 CD16 CD19

CD56 CD235) CD38! CD34+ and the purity was 99.8% ± 0.2% (n = 3) by

FACS Aria III (BD). Human thymi were obtained from living children undergoing

heart surgery and were used in accordance with the guidelines approved

by the Ethics Committee of the University Hospital of Erlangen. Single-cell

suspensions were obtained by tissue disruption and Collagenase D digestion.

Thymocytes were further purified by gradient centrifugation on Bicoll.

Flow CytometryBonemarrow, spleen, and thymus cell suspensionswere prepared and stained

as described (Arndt et al., 2013; von Bonin et al., 2013). Leukocyte counts

of cells of mouse (mCD45+) or human (hCD45+) origin were determined

by analyzing viable cells (DAPI!, 4,6-diamidino-2-phenylindole, Molecular

Probes) usingMACSQuant Analyzer (Miltenyi). Human cellswere stained using

antibodies specific for (clone names given in parentheses) the following: CD10

(HI10A), CD11c (3.9), CD16 (3G8), CD45 (HI30), CD117 (A3C6E2), CD203

(NP4D6), CD235 (HIR2), IgM (MHM-88), and NKp46 (9E2) (all BioLegend);

CD2 (RPA-2.10), CD3 (UCHT1), CD4 (RPA-T4), CD8 (RPA-T8), CD10

(eBioSN5c), CD11b (CBRM1/5), CD14 (M5E2), CD15 (HI98), CD16 (CB16),

CD19 (HIB19), CD33 (WM-53), CD38 (HIT2), CD45RA (HI100), CD56

(MEM188), CD123 (6H6), CD125 (A14), and CD235a (HIR2) (all eBioscience);

and CD34 (581/CD34), CD38 (HIT2), IgD (IA6-2), CD90 (5E10), CD125 (A14),

IgD (IA6-2), TCRab (T10B9.1A-31), and TCRgd (11F2) (all BD Biosciences).

Reagents specific for mouse antigens were as follows: CD3 (2C11), CD11b

(M170), CD19 (eBio1D3), CD31 (390), CD45 (30-F11), CD45R (RA3-6B2),

Gr-1 (RB6-8C5), NK1.1 (PK136), CD51 (RMV-7), CD140a (APA5), CD144

(eBioBV13), Gr-1 (RB6-8C5), and Ter119 (Ter-119) (all eBioscience). Samples

were acquired or sorted using an LSRII or Aria III cytometer (BD Biosciences),

respectively, and were analyzed using FlowJo software (Tree Star).

TransplantationsAdult recipient mice (4–12 weeks of age) were transplanted with (BRg 400cGy,

NSG 200cGy; X-Ray source, MaxiShot, Yxlon) or without irradiation condition-

ing (BRgWv, BRg, NSGWv, NSGWv/+, NSGW41) prior to intravenous injection

of 103 to 3 3 105 CD34+-enriched cord blood cells (HSPCs) or 3 3 104 sorted

lineage!CD38!CD34+ cells in 150 ml PBS/5% FCS. Alternative donor cell

Cell Stem Cell

Human HSC Engraftment in Adult Kit Mutant Mice

236 Cell Stem Cell 15, 227–238, August 7, 2014 ª2014 Elsevier Inc.

Page 11: Kit Regulates HSC Engraftment across the Human-Mouse Species Barrier

numbers are indicated in the figures. SRC values were calculated using ELDA

software as previously described (Grinenko et al., 2014) scoring nonre-

sponders as mice that showed <2% hCD45+ cells in the bone marrow 24–

26 weeks after transplantation of 5 3 104, 104, and 103 CD34-enriched cord

blood HSPCs into NSGW41 or irradiated NSG mice. Pairwise differences in

active cell frequencies between groups were calculated as described previ-

ously (Hu and Smyth, 2009). Newborn recipientmice (1–3 days old) were trans-

planted with (BRg 300cGy, NSG 75cGy) or without irradiation conditioning

(BRgWv or BRg) prior to intrahepatical (i.h.) transfer of 33 105 CD34+-enriched

cord blood cells in 30 ml volume. For secondary transplantations 1 to 2 3 107

nonseparated bone marrow cells were injected per recipient. To assess the

function of prospectively separated human HSC populations, 1,000 (experi-

ment 1) or 6,000 (experiment 2) sorted lineage!CD38!CD34+CD90+CD45RA!

Kithi or Kitint human HSCs were transplanted into 3- to 5-week-old non-

conditioned NSGWv/+ recipient mice. After transplantation mice were given

neomycin-containing drinking water for 3 weeks. For the homing assay, 4 or

7 3 105 human HSPCs were transplanted into adult recipient mice (n =

8–12) with or without previous irradiation (850cGy) and human CD34+ cell

numbers in two femurs were determined 18 hr later. The number of human

HSPCs detected in the bone marrow was normalized to the number of trans-

planted cells.

Polyclonal Stimulation of ThymocytesThymocytes of humanized BRgWv and control BRgWv or BRg mice were

cultured in 200 ml RPMI with or without Phytohemagglutinin (PHA, 1 mg/ml,

Sigma Aldrich) for 72 hr, and subsequently 1 mCi 3H-thymidine (Hartmann An-

alytic) was added for 18 hr. The incorporation of 3H-thymidine was quantified

using a beta counter (Perkin Elmer). Data are presented as fold changes in the

numbers of PHA treated-cells relative to untreated controls.

Morphological AnalysisCells were sorted and cytospun (500 3 g, 5 min) onto glass slides and were

stainedwithMay-Gruenwald-Giemsaasdescribedbefore (Waskowetal., 2008).

SCF ELISALevels of mouse SCF in serum and bone marrow were determined using the

Quantikine mouse SCF ELISA Kit (R&D) according to the manufacturer’s in-

structions. We prepared bone marrow samples by crushing two femurs in

500 ml PBS and centrifuging to remove cells and bone splinters. Blood samples

were allowed to clot for 2 hr at room temperature before centrifugation. All

samples were stored at !20"C and assayed within 1 month of collection.

Molecular AnalysisTREC analysis was performed as follows: genomic DNAwas isolated using the

RNA-Bee (Ams Biotechnology #CS-104B) following the manufacturer’s in-

structions. Relative TREC levels were determined using primers and condi-

tions as described (Hazenberg et al., 2000). PCRs were performed in triplicate

on a Stratagene Mx3500P qPCR Cycler (Agilent). TREC frequencies were

calculated using the DDCt method. Average frequency of TRECs in T cells

from adult donors (40–70 years) was set to 1 to calculate the relative frequency

of TRECs in cord blood CD3+ T cells and in thymocytes from BRgWv recipient

mice. For SCF RT-PCR, bone marrow cells were digested with 0.25%

collagenase type I and 1 mg/ml DNase I at 37"C. Nonhematopoietic cells

(CD45!Ter119!) were enriched (Miltenyi), and for RNA isolation (RNeasy

MicroKit; QIAGEN) 3,000 CD45!Ter119!CD31!PDGFRa+CD51+ MSCs were

sorted (purity: 98.6% ± 1.1%, n = 11). cDNA was synthesized using the

SuperScript First-Strand Synthesis System for RT-PCR (Invitrogen). qPCR re-

actions were run in triplicate for each sample in 20 ml of reaction mix including

2 ml of cDNA, SYBR green (Fermentas), and 0.1 mM of the following primers.

Soluble (s) SCF: forward, 50-CTCTCTTCAACATTAGGTCCCGAGAAAGA

TTCCA; reverse, 50-CTTCCAGTATAAGGCTCCAAAAGCAAAGCCA; trans-

membrane (tm) SCF: forward, 50-CTCTTCAACATTAGGTCCCGAGAAAGG

GAAAG; reverse, 50-CTTCCAGTATAAGGCTCCAAAAGCAAAGCCA; GAPDH

forward, 50-AAGGGGCGGAGATGATGAC; reverse, 50-GGTGCTGAGTATGTC

GTGGAG. Annealing temperatures of 60"C were used. Incorporation of SYBR

green was analyzed using the Mx3005P QPCR system (Agilent) and theMxPro

QPCR Software (MxPro, version 4.10). SCF transcript level was normalized on

GAPDH expression.

Statistical AnalysisA two-tailed Student’s t test was used for all statistical analysis. *p = 0.05–0.01,

**p = 0.01–0.001, and ***p < 0.001. Mean ± standard deviation is shown

throughout figures.

SUPPLEMENTAL INFORMATION

Supplemental Information for this article includes one figure and Supplemental

Experimental Procedures and can be found with this article online at http://dx.

doi.org/10.1016/j.stem.2014.06.001.

AUTHOR CONTRIBUTIONS

K.N.C. and S. Rahmig planned, conducted, and interpreted experiments using

BRgWv, NSGWv, and NSGW41 mice. N.M. and A. Petzold performed SCF

qPCR. I.H., C.S., and J.H. planned, conducted, and interpreted experiments

using HIV. H.W., M.S., and S. Reinke provided crucial help for the analysis.

J.K. provided crucial reagents. G.H., A. Purbojo, R.C., and D.D. provided hu-

man thymus samples, and A.P. and M.B. provided human cord blood and

bone marrow samples. C.W. planned and interpreted experiments and wrote

the paper.

ACKNOWLEDGMENTS

We thank Melanie Portz, Sabrina Piontek, and Sindy Bohme for expert help

with mouse maintenance and genotyping; Tatyana Grinenko for discussion;

and Ulrike Lohr for critical reading of the manuscript. We thank the entire lab-

oratory staff from the DKMS Lifeline Cord Blood Bank for providing cord blood

samples, and we thank Frank Miedema and Sigrid Otto for providing the

cloned Sj fragment for TREC analysis. C.W. is supported by the Center for

Regenerative Therapies Dresden, by the German Research Foundation

(DFG) WA2837, SFB655-B9, SFB127-A3, and FOR2033-A03, and by funding

from the European Union’s Seventh Programme for research, technological

development, and demonstration under grant agreement No. 261387 (CELL-

PID). D.D. is supported by BayGene and the DFG (SFB643-A7).

Received: June 21, 2011

Revised: March 4, 2014

Accepted: June 2, 2014

Published: July 10, 2014

REFERENCES

Arndt, K., Grinenko, T., Mende, N., Reichert, D., Portz, M., Ripich, T.,

Carmeliet, P., Corbeil, D., and Waskow, C. (2013). CD133 is a modifier of he-

matopoietic progenitor frequencies but is dispensable for the maintenance of

mouse hematopoietic stem cells. Proc. Natl. Acad. Sci. USA 110, 5582–5587.

Brehm, M.A., Cuthbert, A., Yang, C., Miller, D.M., DiIorio, P., Laning, J.,

Burzenski, L., Gott, B., Foreman, O., Kavirayani, A., et al. (2010). Parameters

for establishing humanized mouse models to study human immunity: analysis

of human hematopoietic stem cell engraftment in three immunodeficient

strains of mice bearing the IL2rgamma(null) mutation. Clin. Immunol. 135,

84–98.

Brehm, M.A., Racki, W.J., Leif, J., Burzenski, L., Hosur, V., Wetmore, A., Gott,

B., Herlihy, M., Ignotz, R., Dunn, R., et al. (2012). Engraftment of human HSCs

in nonirradiated newborn NOD-scid IL2rg null mice is enhanced by transgenic

expression of membrane-bound human SCF. Blood 119, 2778–2788.

Carbonneau, C.L., Despars, G., Rojas-Sutterlin, S., Fortin, A., Le, O., Hoang,

T., and Beausejour, C.M. (2012). Ionizing radiation-induced expression of

INK4a/ARF in murine bone marrow-derived stromal cell populations interferes

with bone marrow homeostasis. Blood 119, 717–726.

Doulatov, S., Notta, F., Laurenti, E., and Dick, J.E. (2012). Hematopoiesis: a

human perspective. Cell Stem Cell 10, 120–136.

Ghia, P., ten Boekel, E., Sanz, E., de la Hera, A., Rolink, A., and Melchers, F.

(1996). Ordering of human bone marrow B lymphocyte precursors by single-

cell polymerase chain reaction analyses of the rearrangement status of the

immunoglobulin H and L chain gene loci. J. Exp. Med. 184, 2217–2229.

Cell Stem Cell

Human HSC Engraftment in Adult Kit Mutant Mice

Cell Stem Cell 15, 227–238, August 7, 2014 ª2014 Elsevier Inc. 237

Page 12: Kit Regulates HSC Engraftment across the Human-Mouse Species Barrier

Grinenko, T., Arndt, K., Portz, M., Mende, N., Gunther, M., Cosgun, K.N.,

Alexopoulou, D., Lakshmanaperumal, N., Henry, I., Dahl, A., and Waskow,

C. (2014). Clonal expansion capacity defines two consecutive developmental

stages of long-term hematopoietic stem cells. J. Exp. Med. 211, 209–215.

Hazenberg, M.D., Otto, S.A., Cohen Stuart, J.W., Verschuren, M.C., Borleffs,

J.C., Boucher, C.A., Coutinho, R.A., Lange, J.M., Rinke de Wit, T.F.,

Tsegaye, A., et al. (2000). Increased cell division but not thymic dysfunction

rapidly affects the T-cell receptor excision circle content of the naive T cell

population in HIV-1 infection. Nat. Med. 6, 1036–1042.

Hu, Y., and Smyth, G.K. (2009). ELDA: extreme limiting dilution analysis for

comparing depleted and enriched populations in stem cell and other assays.

J. Immunol. Methods 347, 70–78.

Ishikawa, F., Yasukawa, M., Lyons, B., Yoshida, S., Miyamoto, T., Yoshimoto,

G., Watanabe, T., Akashi, K., Shultz, L.D., and Harada, M. (2005).

Development of functional human blood and immune systems in NOD/SCID/

IL2 receptor gamma chain(null) mice. Blood 106, 1565–1573.

Ito, M., Hiramatsu, H., Kobayashi, K., Suzue, K., Kawahata, M., Hioki, K.,

Ueyama, Y., Koyanagi, Y., Sugamura, K., Tsuji, K., et al. (2002). NOD/SCID/

gamma(c)(null) mouse: an excellent recipient mouse model for engraftment

of human cells. Blood 100, 3175–3182.

Kawashima, I., Zanjani, E.D., Almaida-Porada, G., Flake, A.W., Zeng, H., and

Ogawa, M. (1996). CD34+ human marrow cells that express low levels of

Kit protein are enriched for long-term marrow-engrafting cells. Blood 87,

4136–4142.

Kirberg, J., Berns, A., and von Boehmer, H. (1997). Peripheral T cell survival

requires continual ligation of the T cell receptor to major histocompatibility

complex-encoded molecules. J. Exp. Med. 186, 1269–1275.

Majeti, R., Park, C.Y., andWeissman, I.L. (2007). Identification of a hierarchy of

multipotent hematopoietic progenitors in human cord blood. Cell Stem Cell 1,

635–645.

Miller, P.H., Cheung, A.M., Beer, P.A., Knapp, D.J., Dhillon, K., Rabu, G.,

Rostamirad, S., Humphries, R.K., and Eaves, C.J. (2013). Enhanced normal

short-term human myelopoiesis in mice engineered to express human-spe-

cific myeloid growth factors. Blood 121, e1–e4.

Nicolini, F.E., Cashman, J.D., Hogge, D.E., Humphries, R.K., and Eaves, C.J.

(2004). NOD/SCID mice engineered to express human IL-3, GM-CSF and

Steel factor constitutively mobilize engrafted human progenitors and compro-

mise human stem cell regeneration. Leukemia 18, 341–347.

Nilsson, S.K., Dooner, M.S., Tiarks, C.Y., Weier, H.U., and Quesenberry, P.J.

(1997). Potential and distribution of transplanted hematopoietic stem cells in

a nonablated mouse model. Blood 89, 4013–4020.

Nocka, K., Tan, J.C., Chiu, E., Chu, T.Y., Ray, P., Traktman, P., and Besmer, P.

(1990). Molecular bases of dominant negative and loss of function mutations

at the murine c-kit/white spotting locus: W37, Wv, W41 and W. EMBO J. 9,

1805–1813.

Rongvaux, A., Willinger, T., Takizawa, H., Rathinam, C., Auerbach, W.,

Murphy, A.J., Valenzuela, D.M., Yancopoulos, G.D., Eynon, E.E., Stevens,

S., et al. (2011). Human thrombopoietin knockin mice efficiently support

human hematopoiesis in vivo. Proc. Natl. Acad. Sci. USA 108, 2378–2383.

Rongvaux, A., Willinger, T., Martinek, J., Strowig, T., Gearty, S.V., Teichmann,

L.L., Saito, Y., Marches, F., Halene, S., Palucka, A.K., et al. (2014).

Development and function of human innate immune cells in a humanized

mouse model. Nat. Biotechnol. 32, 364–372.

Scheeren, F.A., Nagasawa,M.,Weijer, K., Cupedo, T., Kirberg, J., Legrand, N.,

and Spits, H. (2008). T cell-independent development and induction of somatic

hypermutation in human IgM+ IgD+ CD27+ B cells. J. Exp. Med. 205, 2033–

2042.

Shao, L., Feng, W., Li, H., Gardner, D., Luo, Y., Wang, Y., Liu, L., Meng, A.,

Sharpless, N.E., and Zhou, D. (2014). Total body irradiation causes long-

term mouse BM injury via induction of HSC premature senescence in an

Ink4a- and Arf-independent manner. Blood 123, 3105–3115.

Shen, H., Yu, H., Liang, P.H., Cheng, H., XuFeng, R., Yuan, Y., Zhang, P.,

Smith, C.A., and Cheng, T. (2012). An acute negative bystander effect of g-irra-

diated recipients on transplanted hematopoietic stem cells. Blood 119, 3629–

3637.

Shin, J.Y., Hu, W., Naramura, M., and Park, C.Y. (2014). High c-Kit expression

identifies hematopoietic stem cells with impaired self-renewal and megakar-

yocytic bias. J. Exp. Med. 211, 217–231.

Shultz, L.D., Ishikawa, F., and Greiner, D.L. (2007). Humanized mice in trans-

lational biomedical research. Nat. Rev. Immunol. 7, 118–130.

Shultz, L.D., Brehm, M.A., Garcia-Martinez, J.V., and Greiner, D.L. (2012).

Humanized mice for immune system investigation: progress, promise and

challenges. Nat. Rev. Immunol. 12, 786–798.

Strowig, T., Rongvaux, A., Rathinam, C., Takizawa, H., Borsotti, C., Philbrick,

W., Eynon, E.E., Manz, M.G., and Flavell, R.A. (2011). Transgenic expression

of human signal regulatory protein alpha in Rag2-/-gamma(c)-/-mice improves

engraftment of human hematopoietic cells in humanized mice. Proc. Natl.

Acad. Sci. USA 108, 13218–13223.

Takagi, S., Saito, Y., Hijikata, A., Tanaka, S., Watanabe, T., Hasegawa, T.,

Mochizuki, S., Kunisawa, J., Kiyono, H., Koseki, H., et al. (2012). Membrane-

bound human SCF/KL promotes in vivo human hematopoietic engraftment

and myeloid differentiation. Blood 119, 2768–2777.

Takenaka, K., Prasolava, T.K., Wang, J.C., Mortin-Toth, S.M., Khalouei, S.,

Gan, O.I., Dick, J.E., and Danska, J.S. (2007). Polymorphism in Sirpa

modulates engraftment of human hematopoietic stem cells. Nat. Immunol.

8, 1313–1323.

Traggiai, E., Chicha, L., Mazzucchelli, L., Bronz, L., Piffaretti, J.C.,

Lanzavecchia, A., and Manz, M.G. (2004). Development of a human adaptive

immune system in cord blood cell-transplanted mice. Science 304, 104–107.

von Bonin, M., Wermke, M., Cosgun, K.N., Thiede, C., Bornhauser, M.,

Wagemaker, G., and Waskow, C. (2013). In vivo expansion of co-transplanted

T cells impacts on tumor re-initiating activity of human acute myeloid leukemia

in NSG mice. PLoS ONE 8, e60680.

Waskow, C., Terszowski, G., Costa, C., Gassmann, M., and Rodewald, H.R.

(2004). Rescue of lethal c-KitW/W mice by erythropoietin. Blood 104, 1688–

1695.

Waskow, C., Liu, K., Darrasse-Jeze, G., Guermonprez, P., Ginhoux, F., Merad,

M., Shengelia, T., Yao, K., and Nussenzweig, M. (2008). The receptor tyrosine

kinase Flt3 is required for dendritic cell development in peripheral lymphoid

tissues. Nat. Immunol. 9, 676–683.

Waskow, C., Madan, V., Bartels, S., Costa, C., Blasig, R., and Rodewald, H.R.

(2009). Hematopoietic stem cell transplantation without irradiation. Nat.

Methods 6, 267–269.

Willinger, T., Rongvaux, A., Strowig, T., Manz, M.G., and Flavell, R.A. (2011).

Improving human hemato-lymphoid-system mice by cytokine knock-in gene

replacement. Trends Immunol. 32, 321–327.

Cell Stem Cell

Human HSC Engraftment in Adult Kit Mutant Mice

238 Cell Stem Cell 15, 227–238, August 7, 2014 ª2014 Elsevier Inc.